Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2016 Apr 11;161(7):1751–1760. doi: 10.1007/s00705-016-2855-2

Glycosylation of dengue virus glycoproteins and their interactions with carbohydrate receptors: possible targets for antiviral therapy

Fakhriedzwan Idris 1,, Siti Hanna Muharram 1, Suwarni Diah 1
PMCID: PMC7087181  PMID: 27068162

Abstract

Dengue virus, an RNA virus belonging to the genus Flavivirus, affects 50 million individuals annually, and approximately 500,000–1,000,000 of these infections lead to dengue hemorrhagic fever or dengue shock syndrome. With no licensed vaccine or specific antiviral treatments available to prevent dengue infection, dengue is considered a major public health problem in subtropical and tropical regions. The virus, like other enveloped viruses, uses the host’s cellular enzymes to synthesize its structural (C, E, and prM/M) and nonstructural proteins (NS1–5) and, subsequently, to glycosylate these proteins to produce complete and functional glycoproteins. The structural glycoproteins, specifically the E protein, are known to interact with the host’s carbohydrate receptors through the viral proteins’ N-glycosylation sites and thus mediate the viral invasion of cells. This review focuses on the involvement of dengue glycoproteins in the course of infection and the virus’ exploitation of the host’s glycans, especially the interactions between host receptors and carbohydrate moieties. We also discuss the recent developments in antiviral therapies that target these processes and interactions, focusing specifically on the use of carbohydrate-binding agents derived from plants, commonly known as lectins, to inhibit the progression of infection.

Keywords: Dengue Virus, Mannose Receptor, Dengue Hemorrhagic Fever, Dengue Infection, DENV Infection

Introduction

Dengue virus (DENV) is a member of the family Flaviviridae and has four distinct serotypes, designated DENV1–4. Fifty million people are estimated to become infected with dengue virus annually, and approximately 500,000–1,000,000 of these infections lead to dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS), with mortality rates of 5 %–30 % [1]. With no licensed vaccine or specific antiviral treatments available to prevent dengue infection, it is considered a major public health problem in subtropical and tropical regions.

Dengue virus infection is characterized by headache, biphasic fever, prostration, rash, pain in various parts of the body, leukopenia, and lymphadenopathy [2]. In severe cases, the infection can progress to DHF, which occurs when infection with one serotype causes individuals to suffer more-severe disease after subsequent infection with a different serotype. DHF is a severe febrile disease characterized by abnormalities of hemostasis and increased vascular permeability, which in some instances results in hypovolemic shock syndrome, DSS, when immune cells are enhanced by preexisting non-neutralizing antibodies directed against dengue viral proteins, although the exact mechanism is yet to be resolved [3, 4]. However, host and viral factors, such as the genetics of the infecting viral strain, have been implicated in contributing to disease severity [5].

DENV, like other flaviviruses, has three structural proteins. These structural proteins are originally encoded as a polyprotein by a single long open reading frame (ORF), which is later cleaved co- and posttranslationally by both cellular and viral proteases to produce the C, M, and E proteins from the amino terminus of the polyprotein. The virus also expresses seven nonstructural proteins derived from the carboxyl terminal sequence of the polyprotein, NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5 [6, 7]. The positive-stranded RNA viral genome is packaged in a lipid bilayer [8]. The functions of the proteins are summarized in Table 1. The ORF is flanked by 5′ and the 3′ untranslated regions, which comprise the conserved cis-acting RNA secondary structural elements required for mRNAs translation and replication [9, 10].

Table 1.

Structural and nonstructural proteins of dengue virus

Protein Size Subunits Functions References
C 100 amino acids None Facilitates capsid assembly [11]
prM/M 21 kDa/8 kDa None prM interacts with and stabilizes the E protein. [12]
E 53 kDa None

Forms outer protein envelope

Receptor-binding capability

[12, 13]
NS1 48 kDa None

Cleaves the NS1–NS2A junction

Involved in the early steps of viral replication

[1416]
NS2 22 kDa NS2A, NS2B

NS2A plays a role in RNA synthesis and virion maturation and as an activator of correct NS1 processing

NS2B acts as an activator of NS3, allowing its proper function

[7, 17]
NS3 618 amino acids None

Acts as a protease when combined with NS2B

Functions as an RNA helicase and RTPase/NTPase

[18, 19]
NS4 16 kDa NS4A, NS4B Plays a role in membrane curvature and facilitates protein–protein interactions [2022]
NS5 104 kDa None Essential for the replication and transcription of the viral genome [23, 24]

Glycosylation of DENV glycoproteins

DENV does not encode its own glycosyl transferase enzymes, so it requires the host cell to provide most or all of those necessary for the complete synthesis of the viral glycoproteins, including the glycosylation process. To date, N-glycosylation sites have been identified in only three of the 10 DENV proteins (prM/M, E, and NS1) [8]. The biosynthesis of these viral glycoproteins requires the transfer of a Glc3Man9GlcNAc2 oligosaccharide precursor to the budding protein, after which the terminal glucose residues on the core unit are trimmed [25]. This involves the sequential trimming of three glucose residues attached to the oligosaccharide, which is catalyzed by the endoplasmic reticulum (ER) α-glucosidase I and II enzymes [26]. α-Glucosidase I cleaves the outer α-1,2-linked glucose residue, whereas α-glucosidase II cleaves the two inner α-1,3-linked glucose residues, leaving the proteins monoglycosylated. This action allows the glycoproteins to bind to the ER chaperones calnexin and/or calreticulin to be properly folded downstream. The reglycosylation process is initiated on the incompletely folded proteins by UDP-glucosyltransferase 1, which acts as a sensor of correct protein folding. Once correctly folded, the proteins are then released from the re- and deglycosylation cycle and transported to the Golgi complex for further processing [27].

Glycosylation of E, prM/M, and NS1 glycoproteins

The E glycoprotein monomer has three structural and functional domains. Domain I is the hinge region, which contains the amino terminus; domain II is responsible for the stabilization of the dimer and contains the fusion peptide; and domain III is an immunoglobulin (Ig)-like domain containing putative receptor-binding motifs [28, 29]. The glycoprotein has one or more potential N-linked glycosylation motifs in the form N–X–T/S, where X is any amino acid except proline [30]. Studies have shown that the numbers and locations of the motifs vary significantly among the DENV subtypes [31, 32]. Glycosylation motifs have commonly been observed in domain I at the residue asparagine 153 (N153) or N154, but the addition of a glycan to these residues is not an absolute requirement for infectivity. Although glycosylation is not essential at these residues, several studies have shown that mutations that occur at N-linked glycosylation sites in the E glycoprotein can affect virus-mediated membrane fusion and neurovirulence [3335]. Glycosylation mutants have been described for glycoproteins E. Differences in amino acids were noted between the four wild-type DENV serotypes at amino acid 154, which is glutamic acid in both DENV 1 [36] and DENV 3 [37], and aspartic acid in both DENV 2 [38] and DENV 4 [39]. Another N-linked glycosylation motif of DENV occurs at N67, which mediates the viral infection of dendritic cells bearing DC-SIGN receptors and is essential for viral assembly and exit [40, 41]. Removal of the N67 glycan has been shown to severely affect viral fitness and to reduce cell infectivity [42].

The glycosylation of the DENV prM/M glycoprotein has not been extensively studied. The prM protein is fused to the ectodomain of the E glycoprotein [43] with a glycosylation modification at asparagine N69. Other potential N-linked glycosylation sites have also been identified at residues 7, 31, and 52. The glycosylation of prM may allow it to act as a chaperone for E protein folding in the ER, permit its association with the membrane, and allow it to act in the assembly of the E protein [44].

The glycosylation of NS1 begins when the monomer is altered within the ER lumen by the addition of high-mannose-type glycans at both N-linked glycosylation sites and its subsequent rapid dimerization [45, 46]. Pryor and Wright [47] demonstrated that when secreted NS1 was treated with endoglycosidase H, complex glycans attached at N130 and high-mannose glycans at N207, so both N130 and N207 are N-linked glycosylation sites. Further alterations of the carbohydrate moieties in the NS1 dimer occur in the Golgi apparatus before the protein is transported to the cell surface and released from the infected cell. Interestingly, the presence of a polymannose-type sugar on the dimer may protect one of the N-glycans from further maturation in the Golgi, without which the stability of the dimer and the secretion of the glycoprotein would be reduced [48]. The glycosylation of NS1 is essential for viral processes ranging from replication to virulence, but it is unclear at this point how glycosylation affects these processes [15, 49].

Overview of glycans: virus–glycan interactions

Carbohydrate chains, also known as glycans, are one of the four basic components of cells. They are remarkably structurally diverse [50] and usually exist as covalent linkages with saccharides that are conjugated to proteins (glycoproteins) or to lipids (glycolipids) on the cell surface. The glycans of mammals are well conserved, although species-specific variations occur. Because of these variations, glycans that are involved in pathogen–receptor interactions may determine the susceptibility of specific organisms to infectious pathogens [51]. Different types of glycans can be produced during glycosylation depending on the type of residues that are attached to the cellular proteins or lipids. The glycosylation of proteins involves N-glycans, O-glycans, and glycosaminoglycans, frequently known as proteoglycans. N-glycans are formed when they bind to a specific subset of N residues in proteins, located in the N–X–S/T motif, whereas O-glycans attach to subsets of serine and threonine residues [52, 53]. The linear glycosaminoglycans are also serine- and threonine-linked but are usually highly sulfated [54]. Lipid glycosylation is also a common modification in the secretory pathway that produces glycolipids, also known as glycosphingolipids, which include the sialic-acid-bearing gangliosides [55].

The propagation of a virus and disease progression depend on the direct interactions between the virus and the host cell receptors. Different viruses may have preferences for differ glycan moieties for their attachment. These can be charged glycan moieties, such as sialic acid, which are readily recognized by orthoreoviruses [56], rotaviruses [57], and influenza viruses [58]; heparan sulfate, recognized by herpes viruses [59] and parvoviruses [60]; or neutral glycans, such as histo-blood group antigens, which are bound by rotaviruses [61, 62] and noroviruses [63, 64]. The significant diversity in the recognition of specific glycans within a particular viral species, which arises from genetic differences, dictates the cell tropism, host specificity and adaptation, interspecies transmission, and pathogenesis of the virus.

Glycan interactions with DENV glycoproteins

Four major types of receptors in mammals are believed to be targeted by DENV. These are 1) carbohydrate molecules, 2) carbohydrate-binding proteins (also known as lectins), 3) factors related to protein folding, such as chaperones [65, 66] and heat shock proteins [67], and 4) other proteins, such as a high-affinity laminin receptor [68] and a CD14-associated protein [69]. However, in this review, we are focusing on molecules containing carbohydrate moieties (Table 2), so only carbohydrate and carbohydrate-binding protein receptors will be discussed.

Table 2.

Carbohydrate receptors targeted by DENV on mammalian and insect cells

Molecule Type Cell type Serotype References
DC-SIGN C-type lectin Monocyte-derived dendritic cells DENV 1, 2, 3, and 4 [71]
Heparan sulfate Glycosaminoglycans

Vero

CHO K1

DENV 2 [72]
nLc4Cer Glycosphingolipid

K562

BHK-21

LLC-MK2

DENV 2 [73, 74]
Mannose receptor Protein

NIH3T3

Monocytes

Macrophages

DENV 1, 2, 3, and 4 [75]
High-affinity laminin receptor Protein

HepG2

PS clone D

DENV 1, 2 and 3 [68, 76]
CLEC5A C-type lectin Macrophages DENV 1, 2, 3, and 4 [77]
L-3 Glycosphingolipid AP-61 DENV 2 [74]
40- and 45-kDa glycoproteins Glycoprotein C6/36 cells DENV 4 [7880]

Carbohydrate molecules as receptors

This group of receptors includes sulfated glycosaminoglycans (GAGs) and glycosphingolipids, which can act as coreceptor molecules, enhancing the efficiency of viral entry. Heparan sulfate is among the sulfated GAGs that are involved in the initial attachment of DENV to the cell surface, when the E glycoprotein binds to the molecule. Recently, Okamoto et al. [81] showed that a specific heparan sulfate proteoglycan, called syndecan-2, is a membrane heparan sulfate proteoglycan utilized by DENV as a receptor. Heparan sulfate is a linear repeating copolymer with variably sulfated uronic acid and glucosamine carbohydrate residues, and it is highly negatively charged [82]. It is found not only on the cell surface but also in the extracellular matrix [72].

Another type of carbohydrate molecule that has been reported recently is neolactotetraosylceramide (nLc4Cer), a glycosphingolipid with no sulfation. This carbohydrate molecule may be a coreceptor of DENV and assist in the attachment of the virus to the host cell [73]. DENV interacts with this glycosphingolipid at the nonreducing terminus of Galβ1-4GlcNAcβ, which is expressed on the surface of susceptible cells, such as human erythroleukemia K562 and baby hamster kidney BHK-21 cells.

Carbohydrate-binding proteins

These proteins are commonly grouped with the C-lectins, which are expressed on dendritic cells and macrophages located under human skin. They are known to play an important role in the initial contact with DENV, after it is introduced by a mosquito bite. The best-characterized lectin involved in the interaction between the virus and dendritic cells is dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN). DC-SIGN is classified as a member of the calcium-dependent C-type lectin family and has affinity for the high-mannose glycans found on different pathogens, including Ebola virus [83], hepatitis C virus [84], human immunodeficiency virus (HIV) [85], and several parasites, yeasts, and bacteria [86]. The interaction between DC-SIGN and DENV occurs through the high-mannose N-linked glycans found on the E glycoprotein [40, 87]. This interaction has been confirmed in studies by Navarro-Sanchez et al. [70] and Tassaneetrithep et al. [71] in which both groups successfully inhibited DENV infection by introducing soluble DC-SIGN and antibodies directed against DC-SIGN. However, the internalization of DC-SIGN is not essential for DENV infection [88], so the lectin may not function as a specific receptor but may allow the attachment and concentration of the virus on the cell surface.

This phenomenon can be explained by studying the attachment of the E glycoprotein to the carbohydrate moiety of DC-SIGN. When the E glycoprotein binds to the carbohydrate-recognition domain of DC-SIGN, there is no conformational change in the E glycoprotein of the mature virus, even though such changes are observed when full-length DC-SIGN molecules bind to the glycoprotein. However, this binding leaves a single E glycoprotein in the asymmetric unit vacant, and the putative receptor-binding domain III present in each E glycoprotein is free to bind the receptor at the icosahedral fivefold and threefold axes [40]. Because DC-SIGN is in an oligomeric state on the surface of the cell, occurring as a tetramer when interacting with the virus, the binding of DC-SIGN could promote viral clustering on the cell surface and facilitate its binding to the receptor. It has been proposed that the interaction of carbohydrate moieties with DC-SIGN mimics normal cellular processes and therefore protects the receptor-binding domain of the E protein from the host’s immune surveillance and neutralization.

Lectin receptors on macrophages

Macrophages bear other C-type lectin receptors, the mannose receptor and C-type lectin domain family 5 member A (CLEC5A). The mannose receptor has been shown to bind DENV, Japanese encephalitis virus, and tick-borne meningoencephalitis virus by a mechanism similar to the mechanism of DC-SIGN binding. However, the ligand specificity of the mannose receptor differs from that of DC-SIGN in that its ligands are terminal mannose, fucose, and N-acetyl glucosamine rather than high-mannose oligosaccharides and fucosylated glycans. It has been hypothesized that the mannose receptor is more than just an attachment molecule for DENV because it is internalized into cells during infection and is found mainly in the endocytic pathway, unlike DC-SIGN, which is mainly restricted to the plasma membrane [75].

CLEC5A, also called myeloid DAPI2-associating lectin (MDL-1), is classified as a type II transmembrane receptor and is found on the surface of macrophages and monocytes [89]. The receptor contains a C-type lectin-like domain in its C-terminal extracellular region and has only four amino acids in its predicted N-terminal cytoplasmic region. The receptor is recognized as a potential DENV receptor. The binding of DENV to CLEC5A contributes significantly to the mortality associated with DENV infection by triggering excessive macrophage activation. Chen et al. [77] demonstrated that the survival rate in mice increased when CLEC5A was blocked. Both human and mouse CLEC5A are reported to bind DENV and the interaction is inhibited by mannose and fucose in vitro.

Viral glycosylation processes as therapeutic targets

Understanding the mechanisms that contribute to the invasion of host cells, as discussed above, may clarify how specific agents that interrupt or modify viral replication or expression can be used to limit the damage caused during infection. The synthesis of N-glycans may be interrupted at several stages by the inhibition of specific enzymes. In the rough ER, tunicamycin inhibits the transfer of the tri-mannose core to asparagine residues. The removal of terminal glucose residues may be blocked by nojirimycin or castanospermine [90], and the phosphatase-mediated conversion of the dolichylpyrophosphate released by glycosidic cleavage is inhibited by bacitracin. Brefeldin A inhibits the transport of the resultant high-mannose N-glycans to the Golgi apparatus, and further processing by mannosidase enzymes may be affected by deoxymannojirimycin or swainsonine [91, 92]. Deoxynojirimycin is an inhibitor of ER α-glucosidase and disrupts the trimming of terminal glucose, thus affecting the subsequent folding pathways of DENV glycoproteins prM and E [44, 93].

Targeting virus–carbohydrate receptor interactions with plant lectins

Plant lectins, also known as carbohydrate-binding agents (CBAs), have recently emerged as potent agents for treatment of DENV infection, targeting the initial attachment of the virus to cells. Several CBAs with unique properties and specificities (Table 3) have been identified and used to block viruses. Most of these lectins bind specifically to mannose moieties, ranging from terminal mannosyl residues to high-mannose glycans, which are targeted by a wide range of viruses [94106].

Table 3.

Properties and specificities of carbohydrate-binding agents derived from plants used to target viruses

Acronym Source of lectin Common name Major specificity Virus References
HHA Hippeastrum hybrid Amaryllis bulbs α1,3-mannose and/or α1,6-mannose SIV, HCV, DENV1–4 [9496]
GNA Galanthus nivalis Snowdrop High-mannose structures, multiple terminal mannose α1,3-mannose HCV, HIV-1, DENV1–4 [94, 96]
ConA Canavalia ensiformis Jack bean Terminal mannosyl residues DENV2 [97]
NTL Narcissus tazetta var. chinensis Chinese daffodil Similar to GNA RSV, various strains of influenza virus A and B [98, 99]
NPA Narcissus pseudonarcissus Daffodil or Lent lily α-d-mannose HIV-1 [100]
CA Cymbidium hybrid Cymbidium (orchid) Mannose-specific, d-mannose HIV-1 and 2, CMV, influenza A, HCV [94, 101]
EHA Epipactis helleborine Broad-leaved helleborine Mannose-specific, d-mannose HIV-1 and 2, CMV, influenza A [101]
TDL Typhonium divaricatum (L.) Decne Rodent tuber Mannose-specific HSV-2 [102]
SGM2 Smilax glabra Sarsaparilla Mannose and/or mannan HSV-1, RSV [103]
PpeL Parkia pendula Acacia (male) Terminal mannosyl residues, glucose CMV, herpes virus 6 [104]
PCL Polygonatum cyrtonema Giant Chinese Solomon’s seal Mannose and sialic acid HIV-1 and HIV-2 [105]
BanLec Musa acuminate Banana High-mannose structures HIV-1 [106]
WGA Triticum vulgaris Wheatgerm GlcNAc oligomers, N-acetyl lactosamine, some sialic acid residues DENV2 [107]
UDA Urtica dioica Stinging nettle root GlcNAc oligomers, Galβ1,4-GlcNAcβ1 CMV, coronaviruses, SIV, DENV1–4 [95, 96, 101, 107]
JFL Artocarpus heterophyllus Jackfruit N-acetyl-α-d-galactosamine HSV-2, VZV, CMV [108]

SIV = simian immunodeficiency virus; CMV = cytomegalovirus; DENV = dengue virus; HIV = human immunodeficiency virus; RSV = respiratory syncytial virus; HSV = herpes simplex virus; VZV = varicella-zoster virus

The antiviral activities of these CBAs against DENV have recently been explored, particularly the lectins of Hippeastrum hybrid (HHA), Galanthus nivalis (GNA), and Urtica dioica (UDA), which were initially found to inhibit the interaction between HIV and DC-SIGN-expressing cells [100]. The same CBAs have also been shown to dose-dependently inhibit the interaction between all four serotypes of DENV and DC-SIGN in Raji/DC-SIGN+ cells and monocyte-derived dendritic cells [96]. Binding assays have shown that these CBAs do not interact with cellular membrane proteins but instead interact directly with the viral glycosylated envelope proteins [109]. Concanavalin A (ConA) and wheat-germ agglutinin reduce the development of plaque induced by DENV in BHK cells [97]. A competition assay using mannose showed that the inhibitory effect of ConA results from its binding to α-mannose residues on the viral protein. However, these lectins do not completely inhibit DENV when introduced individually to infected cells. Therefore, further studies of the effects of combinations of CBAs are required.

Concluding remarks

The importance of DENV glycosylation and its interaction with carbohydrate receptors warrants further investigation to develop an efficient treatment for DENV-related diseases. Available drugs that disrupt protein glycosylation will result in the incomplete maturation of DENV. In cases of severe dengue infection, where DENV replication can be enhanced by an antibody-dependent enhancement mechanism [110], these drugs are anticipated to be effective in limiting viral replication, thus reducing the viral load, which may lessen the severity of the disease. The aforementioned drugs are designed to inhibit human glycosylation enzymes, but whether the same drugs can elicit similar effects on the glycosylation enzymes of DENV vectors must be explored further. To date, no studies have specifically explored the DENV protein glycosylation processes in mosquitoes. However, Mason [111] demonstrated the ability of mosquito cell lines to release the mature and glycosylated E glycoprotein and NS1 of Japanese encephalitis virus, another member of the family Flaviviridae, Therefore, it is predicted that DENV also undergoes glycosylation in the mosquito, and glycosylation inhibitors are anticipated to be effective to some degree.

Although drugs that disturb the glycosylation process during the formation of viral glycoproteins may seem useful in this context, the same drugs may also disturb the glycosylation of cellular glycoproteins. However, CBAs or lectins act directly on the viral interaction at the cell surface and need not fuse with the cell to exert their antiviral activities. Therefore, it is anticipated that CBAs will not interfere with the synthesis of glycans on cell-surface glycoproteins. The potential utility of lectins as antiviral agents seems promising. The antiviral potency of lectins against each virus may differ because their three-dimensional conformations differ, or because the availability of glycan conformations with the proper carbohydrate moieties differs across viral proteins. Several challenges must still be addressed, such as the high cost of the purification and mass production of lectins, their storage and stability, bioavailability, and cellular toxicity. However, these issues may be resolved by designing synthetic CBAs that are structurally similar to the natural molecules but stable and nontoxic. Several synthetic molecules are already available [112, 113], providing a basis for the further exploration of potentially therapeutic CBAs. Drug delivery using especially designed capsules or nanocarriers may also be a useful strategy to combat bioavailability problems. Based on these factors, CBAs should be considered a valuable class of antiviral agents that warrants further investigation and eventual application in the clinical setting.

Acknowledgments

This work is part of the Neuroinfectious Diseases in Brunei Darussalam Region project UBD/BRC (1), funded by the Brunei Research Council, Department of Economic Planning and Development, Brunei Darussalam, and supported by Universiti Brunei Darussalam. F. Idris is a recipient of a Graduate Research Scholarship, Universiti Brunei Darussalam.

Funding

This work was funded by the Brunei Research Council, Department of Economic Planning and Development, Brunei Darussalam (UBD/BRC(1)).

Conflicts of interest

Idris F declares that he has no conflict of interest. Muharram SH declares that she has no conflict of interest. Diah S declares that she has no conflict of interest.

Ethical approval

This study contained no experiments involving human participants or animals.

References

  • 1.Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century. Trends Microbiol. 2002;10:100–103. doi: 10.1016/S0966-842X(01)02288-0. [DOI] [PubMed] [Google Scholar]
  • 2.Oishi K, Saito M, Mapua CA, Natividad FF. Dengue illness: clinical features and pathogenesis. J Infect Chemother. 2007;13:125–133. doi: 10.1007/s10156-007-0516-9. [DOI] [PubMed] [Google Scholar]
  • 3.Halstead SB. Neutralization and antibody-dependent enhancement of dengue viruses. Adv Virus Res. 2003;60:421–467. doi: 10.1016/S0065-3527(03)60011-4. [DOI] [PubMed] [Google Scholar]
  • 4.Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S, Limpitikul W, Puttikhunt C, Edwards C, Duangchinda T, Supasa S, Chawansuntati K, Malasit P, Mongkolsapaya J, Screaton G. Cross-reacting antibodies enhance dengue virus infection in humans. Science. 2010;328:745–748. doi: 10.1126/science.1185181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.OhAinle M, Balmaseda A, Macalalad AR, Tellez Y, Zody MC, Saborio S, Nuñez A, Lennon NJ, Birren BW, Gordon A, Henn MR, Harris E. Dynamics of dengue disease severity determined by the interplay between viral genetics and serotype-specific immunity. Sci Transl Med. 2011;3:114ra128. doi: 10.1126/scitranslmed.3003084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Noisakran S, Dechtawewat T, Rinkaewkan P, Puttikhunt C, Kanjanahaluethai A, Kasinrerk W, Sittisombut N, Malasit P. Characterization of dengue virus NS1 stably expressed in 293T cell lines. J Virol Methods. 2007;142:67–80. doi: 10.1016/j.jviromet.2007.01.008. [DOI] [PubMed] [Google Scholar]
  • 7.Stevens AJ, Gahan ME, Mahalingam S, Keller PA. The medicinal chemistry of dengue fever. J Med Chem. 2009;52:7911–7926. doi: 10.1021/jm900652e. [DOI] [PubMed] [Google Scholar]
  • 8.Lindenbach BD, Rich CM. Flaviviridae: the viruses and their replication. In: Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA, Roizman B, Straus SE, editors. Fields virology. 4. Philadelphia: Lippincott Williams & Wilkins; 2001. pp. 991–1041. [Google Scholar]
  • 9.Holden KL, Harris E. Enhancement of dengue virus translation: role of the 3′ untranslated region and the terminal 3′ stem-loop domain. Virology. 2004;329:119–133. doi: 10.1016/j.virol.2004.08.004. [DOI] [PubMed] [Google Scholar]
  • 10.Chiu WW, Kinney RM, Dreher TW. Control of translation by the 5′- and 3′-terminal regions of the dengue virus genome. J Virol. 2005;79:8303–8315. doi: 10.1128/JVI.79.13.8303-8315.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ma L, Jones CT, Groesch TD, Kuhn RJ, Post CB. Solution structure of dengue virus capsid protein reveals another fold. Proc Natl Acad Sci USA. 2004;101:3414–3419. doi: 10.1073/pnas.0305892101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang W, Chipman PR, Corver J, Johnson PR, Zhang Y, Mukhopadhyay S, Baker TS, Strauss JH, Rossmann MG, Kuhn RJ. Visualization of membrane protein domains by cryo-electron microscopy of dengue virus. Nat Struct Biol. 2003;10:907–912. doi: 10.1038/nsb990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kuhn RJ, Zhang W, Rossmann MG, Pletnev SV, Corver J, Lenches E, Jones CT, Mukhopadhyay S, Chipman PR, Strauss EG, Baker TS, Strauss JH. Structure of dengue virus. Implications for flavivirus organization, maturation and fusion. Cell. 2002;108:717–725. doi: 10.1016/S0092-8674(02)00660-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Falgout B, Markoff L. Evidence that flavivirus NS1-NS2A cleavage is mediated by a membrane-bound host protease in the endoplasmic reticulum. J Virol. 1995;69:7232–7243. doi: 10.1128/jvi.69.11.7232-7243.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Muylaert IR, Chambers TJ, Galler R, Rice CM. Mutagenesis of the N-linked glycosylation sites of the yellow fever virus NS1 protein: effects on virus replication and mouse neurovirulence. Virology. 1996;222:159–168. doi: 10.1006/viro.1996.0406. [DOI] [PubMed] [Google Scholar]
  • 16.Avirutnan P, Zhang L, Punyadee N, Manuyakorn A, Puttikhunt C, Kasinrerk W, Malasit P, Atkinson JP, Diamond MS. Secreted NS1 of dengue virus attaches to the surface of cells via interactions with heparan sulfate and chondroitin sulfate E. PLoS Pathog. 2007;3:e183. doi: 10.1371/journal.ppat.0030183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Xie X, Gayen S, Kang C, Shi PY. Membrane topology and function of dengue virus NS2A protein. J Virol. 2013;87:4609–4622. doi: 10.1128/JVI.02424-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bazan JF, Fletterick RJ. Detection of a trypsin-like serine protease domain in flaviviruses and pestiviruses. Virology. 1989;171:637–639. doi: 10.1016/0042-6822(89)90639-9. [DOI] [PubMed] [Google Scholar]
  • 19.Gorbolenya AE, Donchenko AP, Koonin EV, Blinov VM. N-terminal domains of putative helicases of flavi- and pestiviruses may be serine proteases. Nucleic Acids Res. 1989;17:3889–3897. doi: 10.1093/nar/17.10.3889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lindenbach BD, Rice CM. Genetic interaction of flavivirus non-structural proteins NS1 and NS4A as a determinant of replicase function. J Virol. 1999;73:4611–4621. doi: 10.1128/jvi.73.6.4611-4621.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.McMahon HT, Gallop JL. Membrane curvature and mechanisms of dynamic cell membrane remodelling. Nature. 2005;438:590–596. doi: 10.1038/nature04396. [DOI] [PubMed] [Google Scholar]
  • 22.Jao CC, Hedge BG, Gallop JL, Hegde PB, McMahon HT, Haworth IS, Langen R. Roles of amphipathic helices and the bin/amphiphysin/rvs (BAR) domain of endophilin in membrane curvature generation. J Biol Chem. 2010;285:20164–20170. doi: 10.1074/jbc.M110.127811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Egloff MP, Benarroch D, Selisko B, Romette JL, Canard B. An RNA cap (nucleoside-2′-O-)-methyltransferase in the flavivirus RNA polymerase NS5: crystal structure and functional characterization. EMBO J. 2002;21:2757–2768. doi: 10.1093/emboj/21.11.2757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Selisko B, Dutartre H, Guillemot JC, Debarnot C, Benarroch D, Khromykh A, Desprès P, Eglof MP, Canard B. Comparative mechanistic studies of de novo RNA synthesis by flavivirus RNA-dependent RNA polymerases. Virology. 2006;351:145–158. doi: 10.1016/j.virol.2006.03.026. [DOI] [PubMed] [Google Scholar]
  • 25.Ruddock LW, Molinari M. N-glycan processing in ER quality control. J Cell Sci. 2006;119:4373–4380. doi: 10.1242/jcs.03225. [DOI] [PubMed] [Google Scholar]
  • 26.Hebert DN, Foellmer B, Helenius A. Glucose trimming and reglucosylation determine glycoprotein association with calnexin in the endoplasmic reticulum. Cell. 1995;81:425–433. doi: 10.1016/0092-8674(95)90395-X. [DOI] [PubMed] [Google Scholar]
  • 27.Hammond C, Braakman I, Helenius A. Role of N-linked, glucose trimming, and calnexin in glycoprotein folding and quality control. Proc Natl Acad Sci USA. 1994;91:913–917. doi: 10.1073/pnas.91.3.913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Modis Y, Ogata S, Clements D, Harrison SC. A ligand-binding pocket in the dengue virus envelope glycoprotein. Proc Natl Acad Sci USA. 2003;100:6986–6991. doi: 10.1073/pnas.0832193100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Modis Y, Ogata S, Clements D, Harrison SC. Variable surface epitopes in the crystal structure of dengue virus type 3 envelope glycoprotein. J Virol. 2005;79:1223–1231. doi: 10.1128/JVI.79.2.1223-1231.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gavel Y, von Heijne G. Sequence differences between glycosylated and non-glycosylated Asn-X-Thr/Ser acceptor sites: implications for protein engineering. Protein Eng. 1990;3:433–442. doi: 10.1093/protein/3.5.433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Johnson AJ, Guirakhoo F, Roehrig JT. The envelope glycoproteins of dengue 1 and dengue 2 viruses grown in mosquito cells differ in their utilization of potential glycosylation sites. Virology. 1994;203:241–249. doi: 10.1006/viro.1994.1481. [DOI] [PubMed] [Google Scholar]
  • 32.Ishak H, Takegami T, Kamimura K, Funada H. Comparative sequences of two type 1 dengue virus strains possessing different growth characteristics in vitro. Microbiol Immunol. 2001;45:327–331. doi: 10.1111/j.1348-0421.2001.tb02627.x. [DOI] [PubMed] [Google Scholar]
  • 33.Guirakhoo F, Hunt AR, Lewis JG, Roehrig JT. Selection and partial characterization of dengue 2 virus mutants that induce fusion at elevated pH. Virology. 1993;194:219–223. doi: 10.1006/viro.1993.1252. [DOI] [PubMed] [Google Scholar]
  • 34.Kawano H, Rostapshov V, Rosen L, Lai CJ. Genetic determinants of dengue type 4 virus neurovirulence for mice. J Virol. 1993;67:6567–6575. doi: 10.1128/jvi.67.11.6567-6575.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sanchez IJ, Ruiz BH. A single nucleotide change in the E protein gene of dengue virus 2 Mexican strain affects neurovirulence in mice. J Gen Virol. 1996;77:2541–2545. doi: 10.1099/0022-1317-77-10-2541. [DOI] [PubMed] [Google Scholar]
  • 36.Chu MC, O’Rourke EJ, Trent DW. Genetic relatedness among structural protein genes of dengue 1 virus strains. J Gen Virol. 1989;70:1701–1712. doi: 10.1099/0022-1317-70-7-1701. [DOI] [PubMed] [Google Scholar]
  • 37.Osatomi K, Sumiyoshi H. Complete nucleotide sequence of dengue type 3 virus genome RNA. Virology. 1990;176:643–647. doi: 10.1016/0042-6822(90)90037-R. [DOI] [PubMed] [Google Scholar]
  • 38.Deubel V, Kinney RM, Trent DW. Nucleotide sequence and deduced amino-acid sequence of the nonstructural proteins of dengue type 2 virus, Jamaica genotype: comparative analysis of the full-length genome. Virology. 1988;165:234–244. doi: 10.1016/0042-6822(88)90677-0. [DOI] [PubMed] [Google Scholar]
  • 39.Zhao B, Mackow E, Buckler-White A, Markoff L, Chanock RM, Lai CJ, Makino Y. Cloning full length dengue type 4 viral DNA sequences: analysis of genes coding for structural proteins. Virology. 1986;156:77–88. doi: 10.1016/0042-6822(86)90169-8. [DOI] [PubMed] [Google Scholar]
  • 40.Pokidysheva E, Zhang Y, Battisti AJ, Bator-Kelly CM, Chipman PR, Xiao C, Gregio GG, Hendrickson WA, Kuhn RJ, Rossmann MG. Cryo-EM reconstruction of dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell. 2006;124:485–493. doi: 10.1016/j.cell.2005.11.042. [DOI] [PubMed] [Google Scholar]
  • 41.Mondotte JA, Lozach PY, Amara A, Gamarnik AV. Essential role of dengue virus envelope protein N glycosylation at asparagine-67 during viral propagation. J Virol. 2007;81:7136–7148. doi: 10.1128/JVI.00116-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bryant JE, Calvert AE, Mesesan K, Crabtree MB, Volpe KE, Silengo S, Kinney RM, Huang CYH, Miller BR, Roehrig JT. Glycosylation of the dengue 2 virus E protein at N67 is critical for virus growth in vitro but not for growth in intrathoracically inoculated Aedes aegypti mosquitoes. Virology. 2007;366:415–423. doi: 10.1016/j.virol.2007.05.007. [DOI] [PubMed] [Google Scholar]
  • 43.Li L, Lok SM, Yu IM, Zhang Y, Kuhn R, Chen J, Rossmann MG. The flavivirus precursor membrane-envelope protein complex: structure and maturation. Science. 2008;319:1830–1834. doi: 10.1126/science.1153263. [DOI] [PubMed] [Google Scholar]
  • 44.Courageot MP, Frenkiel MP, Dos Santos CD, Deubel V, Després P. α-Glucosidase inhibitors reduce dengue virus production by affecting the initial steps of virion morphogenesis in the endoplasmic reticulum. J Virol. 2000;74:564–572. doi: 10.1128/JVI.74.1.564-572.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Winkler G, Randolph VB, Cleaves GR, Ryan TE, Stollar V. Evidence that the mature form of the flavivirus non-structural protein NS1 is a dimer. Virology. 1988;162:187–196. doi: 10.1016/0042-6822(88)90408-4. [DOI] [PubMed] [Google Scholar]
  • 46.Pryor MJ, Wright PJ. The effects of site-directed mutagenesis on the dimerization and secretion of the NS1 protein specified by dengue virus. Virology. 1993;194:769–780. doi: 10.1006/viro.1993.1318. [DOI] [PubMed] [Google Scholar]
  • 47.Pryor MJ, Wright PJ. Glycosylation mutants of dengue virus NS1 protein. J Gen Virol. 1994;75:1183–1187. doi: 10.1099/0022-1317-75-5-1183. [DOI] [PubMed] [Google Scholar]
  • 48.Flamand M, Megret F, Mathieu M, Lepault J, Rey FA, Deubel V. Dengue virus type 1 nonstructural glycoprotein NS1 is secreted from mammalian cells as a soluble hexamer in a glycosylation-dependent fashion. J Virol. 1999;73:6104–6110. doi: 10.1128/jvi.73.7.6104-6110.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Somnuke P, Hauhart RE, Atkinson JP, Diamond MS, Avirutnam P. N-linked glycosylation of dengue virus NS1 protein modulates secretion, cell-surface expression, hexamer stability, and interactions with human complement. Virology. 2011;413:253–264. doi: 10.1016/j.virol.2011.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ohtsubo K, Marth JD. Glycosylation in cellular mechanisms of health and disease. Cell. 2006;126:855–867. doi: 10.1016/j.cell.2006.08.019. [DOI] [PubMed] [Google Scholar]
  • 51.Gagneux P, Varki A. Evolutionary considerations in relating oligosaccharide diversity to biological function. Glycobiology. 1999;9:747–755. doi: 10.1093/glycob/9.8.747. [DOI] [PubMed] [Google Scholar]
  • 52.Schachter H. The joys of HexNAc. The synthesis and function of N- and O-glycan branches. Glycoconj J. 2000;17:465–483. doi: 10.1023/A:1011010206774. [DOI] [PubMed] [Google Scholar]
  • 53.Yan A, Lennarz WJ. Unravelling the mechanism of protein N-glycosylation. J Biol Chem. 2005;280:3121–3124. doi: 10.1074/jbc.R400036200. [DOI] [PubMed] [Google Scholar]
  • 54.Esko JD, Selleck SB. Order out of chaos: assembly of ligand binding sites in heparan sulfate. Annu Rev Biochem. 2002;71:435–471. doi: 10.1146/annurev.biochem.71.110601.135458. [DOI] [PubMed] [Google Scholar]
  • 55.Maccioni HJ, Giraudo CG, Danniotti JL. Understanding the stepwise synthesis of glycolipids. Neurochem Res. 2002;27:629–636. doi: 10.1023/A:1020271932760. [DOI] [PubMed] [Google Scholar]
  • 56.Reiter DM, Frierson JM, Halvorson EE, Kobayashi T, Dermody TS, Stehle T. Crystal structure of reovirus attachment protein σ1 in complex with sialylated oligosaccharides. PLoS Pathog. 2011;7:e1002166. doi: 10.1371/journal.ppat.1002166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dormitzer PR, Sun ZY, Wagner G, Harrison SC. The rhesus rotavirus VP4 sialic acid binding domain has a galectin fold with a novel carbohydrate binding site. EMBO J. 2002;21:885–897. doi: 10.1093/emboj/21.5.885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Connor RJ, Kawaoka Y, Webster RG, Paulson JC. Receptor specificity in human, avian, and equine H2 and H3 influenza virus isolates. Virology. 1994;205:17–23. doi: 10.1006/viro.1994.1615. [DOI] [PubMed] [Google Scholar]
  • 59.Shieh MT, WuDunn D, Montgomery RI, Esko JD, Spearm PG. Cell surface receptors for herpes simplex virus are heparan sulfate proteoglycans. J Cell Biol. 1992;116:1273–1281. doi: 10.1083/jcb.116.5.1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hueffer K, Parrishm CR. Parvovirus host range, cell tropism and evolution. Curr Opin Microbiol. 2003;6:392–398. doi: 10.1016/S1369-5274(03)00083-3. [DOI] [PubMed] [Google Scholar]
  • 61.Huang P, Xia M, Tan M, Zhong W, Wei C, Wang L, Morrow A, Jiang X. Spike protein VP8* of human rotavirus recognizes histo-blood group antigens in a type-specific manner. J Virol. 2012;86:4833–4843. doi: 10.1128/JVI.05507-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hu L, Crawford SE, Czako R, Cortes-Penfield NW, Smith DF, Le Pendu J, Estes MK, Prasad BV. Cell attachment protein VP8* of a human rotavirus specifically interacts with A-type histo-blood group antigen. Nature. 2012;485:256–259. doi: 10.1038/nature10996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Marionneau S, Ruvoen N, Le Moullac-Vaidye B, Clement M, Cailleau-Thomas A, Ruiz-Palacois G, Huang P, Jiang X, Le Pendu J. Norwalk virus binds to histo-blood group antigens present on gastroduodenal epithelial cells of secretor individuals. Gastroenterology. 2002;122:1967–1977. doi: 10.1053/gast.2002.33661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Huang P, Farkas T, Marionneau S, Zhong W, Ruvoen-Clouet N, Morrow AL, Altaye M, Pickering LK, Newburg DS, Le Pendu J, Jiang X. Noroviruses bind to human ABO, Lewis, and secretor histo-blood group antigens: identification of 4 distinct strain-specific patterns. J Infect Dis. 2003;188:19–31. doi: 10.1086/375742. [DOI] [PubMed] [Google Scholar]
  • 65.Cabrera-Hernandez A, Thepparit C, Suksanpaisan L, Smith DR. Dengue virus entry into liver (HepG2) cells is independent of hsp90 and hsp70. J Med Virol. 2007;79:386–392. doi: 10.1002/jmv.20786. [DOI] [PubMed] [Google Scholar]
  • 66.Upanan S, Kuadkitkan A, Smith DR. Identification of dengue virus binding proteins using affinity chromatography. J Virol Methods. 2008;151:325–328. doi: 10.1016/j.jviromet.2008.05.001. [DOI] [PubMed] [Google Scholar]
  • 67.Reyes-Del Valle J, Chávez-Salinas S, Medina F, Del Angel RM. Heat shock protein 90 and heat shock protein 70 are components of dengue virus receptor complex in human cells. J Virol. 2005;79:4557–4567. doi: 10.1128/JVI.79.8.4557-4567.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Tio PH, Jong WW, Cardosa MJ. Two dimensional VOPBA reveals laminin receptor (LAMR1) interaction with dengue virus serotypes 1, 2 and 3. Virol J. 2005;2:25. doi: 10.1186/1743-422X-2-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chen YC, Wang SY, King CC. Bacterial lipopolysaccharide inhibits dengue virus infection of primary human monocytes/macrophages by blockade of virus entry via a CD14-dependent mechanism. J Virol. 1999;73:2650–2657. doi: 10.1128/jvi.73.4.2650-2657.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Navarro-Sanchez E, Altmeyer R, Amara A, Schwartz O, Fieschi F, Virelizier JL, Arenzana-Seisdedos F, Desprès P. Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Rep. 2003;4:723–728. doi: 10.1038/sj.embor.embor866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tassaneetrithep B, Burgess TH, Granelli-Piperno A, Trumpfheller C, Finke J, Sun W, Eller MA, Pattanapanyasat K, Sarasombath S, Birx DL, Steinman RM, Schlesinger S, Marovich MA. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med. 2003;197:823–829. doi: 10.1084/jem.20021840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chen Y, Maguire T, Hileman RE, Fromm JR, Esko JD, Linhardt RJ, Marks RM. Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nat Med. 1997;3:866–871. doi: 10.1038/nm0897-866. [DOI] [PubMed] [Google Scholar]
  • 73.Aoki C, Hidari KIPJ, Itonari S, Yamada A, Takahashi N, Kasama T, Hasebe F, Islam MA, Hatano K, Matsuoka K, Taki T, Guo CT, Takahashi T, Sakano Y, Suzuki T, Miyamoto D, Sugita M, Terunuma D, Morita K, Suzuki Y. Identification and characterization of carbohydrate molecules in mammalian cells recognized by dengue virus type 2. J Biochem. 2006;139:607–614. doi: 10.1093/jb/mvj067. [DOI] [PubMed] [Google Scholar]
  • 74.Wichit S, Jittmittraphap A, Hidari KI, Thaisomboonsuk B, Petmitr S, Aoki C, Itonori S, Morita K, Suzuki T, Suzuki Y, Jampangern W. Dengue virus type 2 recognizes the carbohydrate moiety of neutral glycosphingolipids in mammalian and mosquito cells. Microbiol Immunol. 2011;55:135–140. doi: 10.1111/j.1348-0421.2010.00293.x. [DOI] [PubMed] [Google Scholar]
  • 75.Miller JL, Dewet BJ, Martinez-Pomares L, Radcliffe CM, Dwek RA, Rudd PM, Gordon S. The mannose receptor mediates dengue virus infection of macrophages. PLoS Pathog. 2008;4:e17. doi: 10.1371/journal.ppat.0040017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Thepparit C, Smith DR. Serotype-specific entry of dengue virus into liver cells: identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor as a dengue virus serotype 1 receptor. J Virol. 2004;78:12647–12656. doi: 10.1128/JVI.78.22.12647-12656.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chen ST, Lin YL, Huang MT, Wu MF, Cheng SC, Lei HY, Lee CK, Chiou TW, Wong CH, Hsieh SL. CLEC5A is critical for dengue-virus-induced lethal disease. Nature. 2008;453:672–676. doi: 10.1038/nature07013. [DOI] [PubMed] [Google Scholar]
  • 78.Salas-Benito JS, del Angel RM. Identification of two surface proteins from C6/36 cells that bind dengue type 4 virus. J Virol. 1997;71:7746–7752. doi: 10.1128/jvi.71.10.7246-7252.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yazi Mendoza M, Salas-Benito JS, Lanz-Mendoza H, Hernández-Martínez S, del Angel RM. A putative receptor for dengue virus in mosquito tissues: localization of a 45-kDa glycoprotein. Am J Trop Med Hyg. 2002;67:76–84. doi: 10.4269/ajtmh.2002.67.76. [DOI] [PubMed] [Google Scholar]
  • 80.Reyes-del Valle J, del Angel RM. Isolation of putative dengue virus receptor molecules by affinity chromatography using a recombinant E protein ligand. J Virol Methods. 2004;116:95–102. doi: 10.1016/j.jviromet.2003.10.014. [DOI] [PubMed] [Google Scholar]
  • 81.Okamoto K, Kinoshita H, Parquet Mdel C, Raekiansyah M, Kimura D, Yui K, Islam MA, Hasebe F, Morita K. Dengue virus strain DEN2 16681 utilizes a specific glycochain of syndecan-2 proteoglycan receptor. J Gen Virol. 2012;93:761–770. doi: 10.1099/vir.0.037853-0. [DOI] [PubMed] [Google Scholar]
  • 82.Griffin CC, Linhardt RJ, Van Gorp CL, Toida T, Hileman RE, Schubert RL, Brown SE. Isolation and characterization of heparan sulfate from crude porcine intestinal mucosal peptidoglycan heparin. Carbohydr Res. 1995;276:183–197. doi: 10.1016/0008-6215(95)00166-Q. [DOI] [PubMed] [Google Scholar]
  • 83.Marzi A, Möller P, Hanna SL, Harrer T, Eisemann J, Steinkasserer A, Becker S, Baribaud F, Pöhlmann S. Analysis of the interaction of Ebola virus glycoprotein with DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) and its homologue DC-SIGNR. J Infect Dis. 2007;196:S237–S246. doi: 10.1086/520607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Pöhlmann S, Zhang J, Baribaud F, Chen Z, Leslie GJ, Lin G, Granelli-Piperno A, Doms RW, Rice CM, McKeating JA. Hepatitis C virus glycoproteins interact with DC-SIGN and DC-SIGNR. J Virol. 2003;77:4070–4080. doi: 10.1128/JVI.77.7.4070-4080.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Geijtenbeek TB, Kwon DS, Torensma R, van Vliet SJ, van Duijnhiven GC, Middel J, Cornelissen IL, Mottet HS, KewaRahmani VN, Littman DR, Fiqdor CG, van Kooyk Y. DC-SIGN, a dendritic cell-specific HIV-1 binding protein that enhances trans-infection of T cells. Cell. 2000;100:587–597. doi: 10.1016/S0092-8674(00)80694-7. [DOI] [PubMed] [Google Scholar]
  • 86.van Kooyk Geijtenbeek TB. DC-SIGN: escape mechanism for pathogens. Nat Rev Immunol. 2003;3:697–709. doi: 10.1038/nri1182. [DOI] [PubMed] [Google Scholar]
  • 87.Hacker K, White L, de Silva AM. N-linked glycans on dengue viruses grown in mammalian and insect cells. J Gen Virol. 2009;90:2097–2106. doi: 10.1099/vir.0.012120-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lozach PY, Burleigh L, Staropoli I, Navarro-Sanchez E, Harriague J, Virelizier JL, Rey FA, Després P, Arenzana-Seisdedos F, Amara A. Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals. J Biol Chem. 2005;280:23698–23708. doi: 10.1074/jbc.M504337200. [DOI] [PubMed] [Google Scholar]
  • 89.Bakker AB, Baker E, Sutherland GR, Phillips JH, Lanier LL. Myeloid DAP12-associating lectin (MDL)-1 is a cell surface receptor involved in the activation of myeloid cells. Proc Natl Acad Sci USA. 1999;96:9792–9796. doi: 10.1073/pnas.96.17.9792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Elbein AD. Glycosidase inhibitors: inhibitors of N-linked oligosaccharide processing. FASEB J. 1991;5:3055–3063. doi: 10.1096/fasebj.5.15.1743438. [DOI] [PubMed] [Google Scholar]
  • 91.Cenci di Bello I, Fleet G, Namgoong SK, Tadano K, Winchester B. Structure–activity relationship of swainsonine. Inhibition of human α-mannosidases by swainsonine analogues. Biochem J. 1989;259:855–861. doi: 10.1042/bj2590855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Winchester B, Barker C, Baines S, Jacob GS, Namgoong SK, Fleet G. Inhibition of α-l-fucosidase by derivatives of deoxyfuconojirimycin and deoxymannojirimycin. Biochem J. 1990;265:277–282. doi: 10.1042/bj2650277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wu SF, Lee CJ, Liao CL, Dwek RA, Zitzmann N, Lin YL. Antiviral effects of an iminosugar derivative on flavivirus infections. J Virol. 2002;76:3596–3604. doi: 10.1128/JVI.76.8.3596-3604.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Bertaux C, Daelemans D, Meertens L, Cormier EG, Reinus JF, Peumans WJ, Van Damme EJM, Igarashi Y, Oki T, Schols D, Dragic T, Balzarini J. Entry of hepatitis C virus and human immunodeficiency virus is selectively inhibited by carbohydrate-binding agents but not by polyanions. Virology. 2007;366:40–50. doi: 10.1016/j.virol.2007.04.008. [DOI] [PubMed] [Google Scholar]
  • 95.François KO, Auwerx J, Schols D, Balzarini J. Simian immunodeficiency virus is susceptible to inhibition by carbohydrate-binding agents in a manner similar to that of HIV: implications for further preclinical drug development. Mol Pharmacol. 2008;74:330–337. doi: 10.1124/mol.108.047621. [DOI] [PubMed] [Google Scholar]
  • 96.Alen MMF, Kaptein SJF, De Burghgraeve T, Balzarini J, Neyts J, Schols D. Antiviral activity of carbohydrates-binding agents and the role of DC-SIGN in dengue virus infection. Virology. 2009;387:67–75. doi: 10.1016/j.virol.2009.01.043. [DOI] [PubMed] [Google Scholar]
  • 97.Hung SL, Lee PL, Chen LK, Kao CL, King CC. Analysis of the steps involved in Dengue virus entry into host cells. Virology. 1999;257:156–167. doi: 10.1006/viro.1999.9633. [DOI] [PubMed] [Google Scholar]
  • 98.Ooi LSM, Ho WS, Ngai KLK, Tian L, Chan PKS, Sun SSM, Ooi VEC. Narcissus tazetta lectin shows strong inhibitory effects against respiratory syncytial virus, influenza A (H1N1, H3N2, H5N1) and B viruses. J Biosci. 2010;35:95–103. doi: 10.1007/s12038-010-0012-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gao ZM, Zheng B, Wang WY, Li Q, Yuan QP. Cloning and functional characterization of a GNA-like lectin from Chinese narcissus (Narcissus tazetta var. Chinensis Roem) Physiol Plant. 2011;142:193–204. doi: 10.1111/j.1399-3054.2011.01449.x. [DOI] [PubMed] [Google Scholar]
  • 100.Balzarini J, Van Herrewege Y, Vermeire K, Vanham G, Schols D. Carbohydrate-binding agents efficiently prevent dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN)-directed HIV-1 transmission to T lymphocytes. Mol Pharmacol. 2007;71:3–11. doi: 10.1124/mol.106.030155. [DOI] [PubMed] [Google Scholar]
  • 101.Balzarini J, Neyts J, Schols D, Hosoya M, van Damme E, Peumans W, de Clercq E. The mannose-specific plant lectins from Cymbidium hybrid and Epipactis helleborine and the (N-acetylglucosamine)n-specific plant lectin from Urtica dioica are potent and selective inhibitors of human immunodeficiency virus and cytomegalovirus replication in vitro. Antivir Res. 1992;18:191–207. doi: 10.1016/0166-3542(92)90038-7. [DOI] [PubMed] [Google Scholar]
  • 102.Luo Y, Xu X, Liu J, Li J, Sun Y, Liu Z, Liu J, van Damme E, Balzarini J, Bao J. A novel mannose-binding tuber lectin from Typhonium divaricatum (L.) Decne (family Araceae) with antiviral activity against HSV-II and anti-proliferative effect on human cancer cell lines. J Biochem Mol Biol. 2007;40:358–367. doi: 10.5483/BMBRep.2007.40.3.358. [DOI] [PubMed] [Google Scholar]
  • 103.Ooi LSM, Sun SSM, Wang H, Ooi VEC. New mannose-binding lectin isolated from the rhizome of sarsaparilla Smilax glabra Roxb. (Liliaceae) J Agric Food Chem. 2004;52:6091–6095. doi: 10.1021/jf030837o. [DOI] [PubMed] [Google Scholar]
  • 104.Favacho ARM, Cintra EA, Coelho LCBB, Linhares MIS. In vitro activity evaluation of Parkia pendula seed lectin against human cytomegalovirus and herpes virus 6. Biologicals. 2007;35:189–194. doi: 10.1016/j.biologicals.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 105.An J, Liu J, Wu C, Li J, Dai L, van Damme E, Balzarini J, de Clercq E, Chen F, Bao J. Anti-HIV I/II activity and molecular cloning of a novel mannose/sialic acid-binding lectin from rhizome of Polygonatum cystonema Hua. Acta Biochim Biophys Sin. 2006;38:70–78. doi: 10.1111/j.1745-7270.2006.00140.x. [DOI] [PubMed] [Google Scholar]
  • 106.Swanson MD, Winter HC, Goldstein IJ, Markovitz DM. A lectin isolated from bananas is a potent inhibitor of HIV replication. J Biol Chem. 2010;285:8646–8655. doi: 10.1074/jbc.M109.034926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Van der Meer FJUM, de Haan CAM, Schuurman NMP, Haijema BJ, Verheije MH, Bosch BJ, Balzarini J, Egberink HF. The carbohydrate-binding plant lectins and the non-peptidic antibiotic pradimicin A target the glycans of the coronavirus envelope glycoproteins. J Antimicrob Chemother. 2007;60:741–749. doi: 10.1093/jac/dkm301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wetprasit N, Threesangsri W, Klamklai N, Chulavatnatol M. Jackfruit lectin: properties of mitogenicity and the inhibition of herpesvirus infection. Jpn J Infect Dis. 2000;53:156–161. [PubMed] [Google Scholar]
  • 109.Alen MMF, De Burghgraeve T, Kaptein SJF, Balzarini J, Neyts J, Schols D. Broad antiviral activity of carbohydrate-binding agents against the four serotypes of dengue virus in monocyte-derived dendritic cells. PLoS One. 2011;6:e21658. doi: 10.1371/journal.pone.0021658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Guzman MG, Alvarez M, Halstead SB. Secondary infection as a risk factor for dengue hemorrhagic fever/dengue shock syndrome: an historical perspective and role of antibody-dependent enhancement of infection. Arch Virol. 2013;158:1445–1459. doi: 10.1007/s00705-013-1645-3. [DOI] [PubMed] [Google Scholar]
  • 111.Mason P. Maturation of Japanese encephalitis virus glycoproteins produced by infected mammalian and mosquito cells. Virology. 1989;169:354–364. doi: 10.1016/0042-6822(89)90161-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Striegler S, Dittel M. A sugar discriminating binuclear copper (II) complex. J Am Chem Soc. 2003;125:11518–11524. doi: 10.1021/ja035561f. [DOI] [PubMed] [Google Scholar]
  • 113.Mazik M, Cavqa H, Jones PG. Molecular recognition of carbohydrates with artificial receptors: mimicking the binding motifs found in the crystal structures of protein–carbohydrate complexes. J Am Chem Soc. 2005;127:9045–9052. doi: 10.1021/ja043037i. [DOI] [PubMed] [Google Scholar]

Articles from Archives of Virology are provided here courtesy of Nature Publishing Group

RESOURCES