5,476
Views
6
CrossRef citations to date
0
Altmetric
Review

Glycosylation of viral proteins: Implication in virus–host interaction and virulence

, , , , , & show all
Pages 670-683 | Received 14 Jan 2022, Accepted 28 Mar 2022, Published online: 18 Apr 2022

References

  • Jones KE, Patel NG, Levy MA, et al. Global trends in emerging infectious diseases. Nature. 2008;451:990–993.
  • Bagdonaite I, Wandall HH. Global aspects of viral glycosylation. Glycobiology. 2018;28:443–467.
  • Evans DeWald L, Starr C, Butters T, et al. Iminosugars: a host-targeted approach to combat Flaviviridae infections. Antiviral Res. 2020;184:104881.
  • Mathys L, François KO, Quandte M, et al. Deletion of the highly conserved N-glycan at Asn260 of HIV-1 gp120 affects folding and lysosomal degradation of gp120, and results in loss of viral infectivity. PLoS One. 2014;9:e101181.
  • Kim JM, Yun SI, Song BH, et al. A single N-linked glycosylation site in the Japanese encephalitis virus prM protein is critical for cell type-specific prM protein biogenesis, virus particle release, and pathogenicity in mice. J Virol. 2008;82:7846–7862.
  • Hanna SL, Pierson TC, Sanchez MD, et al. N-Linked glycosylation of west Nile virus envelope proteins influences particle assembly and infectivity. J Virol. 2005;79:13262–13274.
  • York IA, Stevens J, Iv A. Influenza virus N-linked glycosylation and innate immunity. Biosci Rep. 2019;39(1):BSR20171505.
  • Bonhomme CJ, Capul AA, Lauron EJ, et al. Glycosylation modulates arenavirus glycoprotein expression and function. Virology. 2011;409:223–233.
  • Zhu X, Liu Y, Guo J, et al. Effects of N-linked glycan on Lassa virus envelope glycoprotein cleavage, infectivity, and immune response. Virol Sin. 2021;36:774–783.
  • Yang Q, Hughes TA, Kelkar A, et al. Inhibition of SARS-CoV-2 viral entry upon blocking N- and O-glycan elaboration. Elife. 2020;9. DOI:10.7554/eLife.61552.
  • Mondotte JA, Lozach PY, Amara A, et al. Essential role of Dengue virus envelope protein N glycosylation at asparagine-67 during viral propagation. J Virol. 2007;81:7136–7148.
  • Gwon YD, Zusinaite E, Merits A, et al. N-glycosylation in the pre-membrane protein is essential for the Zika virus life cycle. Viruses. 2020;12(9):925.
  • Lennemann NJ, Rhein BA, Ndungo E, et al. Comprehensive functional analysis of N-linked glycans on Ebola virus GP1. mBio. 2014;5:e00862–13.
  • Vigerust DJ, Shepherd VL. Virus glycosylation: role in virulence and immune interactions. Trends Microbiol. 2007;15:211–218.
  • Gubler DJ. Dengue/dengue haemorrhagic fever: history and current status. Novartis Found Symp. 2006;27771-3: 3–16–3251–3. discussion -22.
  • Halstead SB. Observations related to pathogenesis of dengue hemorrhagic fever. VI. Hypotheses and discussion. Yale J Biol Med. 1970;42:350–362.
  • Rodenhuis-Zybert IA, Wilschut J, Smit JM. Dengue virus life cycle: viral and host factors modulating infectivity. Cell Mol Life Sci. 2010;67:2773–2786.
  • Ma L, Jones CT, Groesch TD, et al. Solution structure of Dengue virus capsid protein reveals another fold. Proc Natl Acad Sci U S A. 2004;101:3414–3419.
  • Jones CT, Ma L, Burgner JW, et al. Flavivirus capsid is a dimeric alpha-helical protein. J Virol. 2003;77:7143–7149.
  • Chang CJ, Luh HW, Wang SH, et al. The heterogeneous nuclear ribonucleoprotein K (hnRNP K) interacts with Dengue virus core protein. DNA Cell Biol. 2001;20:569–577.
  • Yap SSL, Nguyen-Khuong T, Rudd PM, et al. Dengue virus glycosylation: what do we know? Front Microbiol. 2017;8:1415.
  • Alen MM, Dallmeier K, Balzarini J, et al. Crucial role of the N-glycans on the viral E-envelope glycoprotein in DC-SIGN-mediated Dengue virus infection. Antiviral Res. 2012;96:280–287.
  • Pokidysheva E, Zhang Y, Battisti AJ, et al. Cryo-EM reconstruction of Dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell. 2006;124:485–493.
  • Courageot MP, Frenkiel MP, Dos Santos CD, et al. Alpha-Glucosidase inhibitors reduce Dengue virus production by affecting the initial steps of virion morphogenesis in the endoplasmic reticulum. J Virol. 2000;74:564–572.
  • Pierson TC, Diamond MS. The emergence of Zika virus and its new clinical syndromes. Nature. 2018;560:573–581.
  • Oehler E, Watrin L, Larre P, et al. Zika virus infection complicated by Guillain-Barre syndrome–case report, French Polynesia, December 2013. Euro Surveill 2014; 19.
  • Muñoz LS, Parra B, Pardo CA. Neurological implications of Zika virus infection in adults. J Infect Dis. 2017;216:S897–S905.
  • Carbaugh DL, Baric RS, Lazear HM. Envelope protein glycosylation mediates Zika virus pathogenesis. J Virol. 2019;93(12):e00113-19.
  • Wen D, Li S, Dong F, et al. N-glycosylation of viral E protein is the determinant for vector midgut invasion by flaviviruses. mBio. 2018;9.
  • Fontes-Garfias CR, Shan C, Luo H, et al. Functional analysis of glycosylation of Zika virus envelope protein. Cell Rep. 2017;21:1180–1190.
  • Guo Y, Bao L, Xu Y, et al. The ablation of envelope protein glycosylation enhances the neurovirulence of ZIKV and cell apoptosis in newborn mice. J Immunol Res. 2021;2021:5317662.
  • Maharaj PD, Langevin SA, Bolling BG, et al. N-Linked glycosylation of the West Nile virus envelope protein is not a requisite for avian virulence or vector competence. PLoS Negl Trop Dis. 2019;13:e0007473.
  • Ebel GD, Carricaburu J, Young D, et al. Genetic and phenotypic variation of West Nile virus in New York, 2000-2003. Am J Trop Med Hyg. 2004;71:493–500.
  • Adams SC, Broom AK, Sammels LM, et al. Glycosylation and antigenic variation among Kunjin virus isolates. Virology. 1995;206:49–56.
  • Berthet FX, Zeller HG, Drouet MT, et al. Extensive nucleotide changes and deletions within the envelope glycoprotein gene of Euro-African West Nile viruses. J Gen Virol. 1997;78(Pt 9):2293–2297.
  • Martina BE, Koraka P, van den Doel P, et al. DC-SIGN enhances infection of cells with glycosylated West Nile virus in vitro and virus replication in human dendritic cells induces production of IFN-alpha and TNF-alpha. Virus Res. 2008;135:64–71.
  • Moudy RM, Payne AF, Dodson BL, et al. Requirement of glycosylation of West Nile virus envelope protein for infection of, but not spread within, Culex quinquefasciatus mosquito vectors. Am J Trop Med Hyg. 2011;85:374–378.
  • Moudy RM, Zhang B, Shi PY, et al. West Nile virus envelope protein glycosylation is required for efficient viral transmission by Culex vectors. Virology. 2009;387:222–228.
  • Shirato K, Miyoshi H, Goto A, et al. Viral envelope protein glycosylation is a molecular determinant of the neuroinvasiveness of the New York strain of West Nile virus. J Gen Virol. 2004;85:3637–3645.
  • Zhang Y, Chen P, Cao R, et al. Mutation of putative N-linked glycosylation sites in Japanese encephalitis virus premembrane and envelope proteins enhances humoral immunity in BALB/C mice after DNA vaccination. Virol J. 2011;8:138.
  • Liang JJ, Chou MW, Lin YL. DC-SIGN binding contributed by an extra N-linked glycosylation on Japanese Encephalitis Virus envelope protein reduces the ability of viral brain invasion. Front Cell Infect Microbiol. 2018;8:239.
  • Helle F, Vieyres G, Elkrief L, et al. Role of N-linked glycans in the functions of hepatitis C virus envelope proteins incorporated into infectious virions. J Virol. 2010;84:11905–11915.
  • Goffard A, Callens N, Bartosch B, et al. Role of N-linked glycans in the functions of hepatitis C virus envelope glycoproteins. J Virol. 2005;79:8400–8409.
  • Falkowska E, Kajumo F, Garcia E, et al. Hepatitis C virus envelope glycoprotein E2 glycans modulate entry, CD81 binding, and neutralization. J Virol. 2007;81:8072–8079.
  • Helle F, Goffard A, Morel V, et al. The neutralizing activity of anti-hepatitis C virus antibodies is modulated by specific glycans on the E2 envelope protein. J Virol. 2007;81:8101–8111.
  • Beasley DW, Whiteman MC, Zhang S, et al. Envelope protein glycosylation status influences mouse neuroinvasion phenotype of genetic lineage 1 West Nile virus strains. J Virol. 2005;79:8339–8347.
  • Bryant JE, Calvert AE, Mesesan K, et al. 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.
  • Guirakhoo F, Hunt AR, Lewis JG, et al. Selection and partial characterization of dengue 2 virus mutants that induce fusion at elevated pH. Virology. 1993;194:219–223.
  • Kawano H, Rostapshov V, Rosen L, et al. Genetic determinants of dengue type 4 virus neurovirulence for mice. J Virol. 1993;67:6567–6575.
  • Lee E, Leang SK, Davidson A, et al. Both E protein glycans adversely affect dengue virus infectivity but are beneficial for virion release. J Virol. 2010;84:5171–5180.
  • Mossenta M, Marchese S, Poggianella M, et al. Role of N-glycosylation on Zika virus E protein secretion, viral assembly and infectivity. Biochem Biophys Res Commun. 2017;492:579–586.
  • Auwerx J, François KO, Covens K, et al. Glycan deletions in the HIV-1 gp120 V1/V2 domain compromise viral infectivity, sensitize the mutant virus strains to carbohydrate-binding agents and represent a specific target for therapeutic intervention. Virology. 2008;382:10–19.
  • Mathys L, Balzarini J. Several N-glycans on the HIV envelope glycoprotein gp120 preferentially locate near disulphide bridges and are required for efficient infectivity and virus transmission. PLoS One. 2015;10:e0130621.
  • François KO, Balzarini J. The highly conserved glycan at asparagine 260 of HIV-1 gp120 is indispensable for viral entry. J Biol Chem. 2011;286:42900–42910.
  • Quiñones-Kochs MI, Buonocore L, Rose JK. Role of N-linked glycans in a human immunodeficiency virus envelope glycoprotein: effects on protein function and the neutralizing antibody response. J Virol. 2002;76:4199–4211.
  • Schulze IT. Effects of glycosylation on the properties and functions of influenza virus hemagglutinin. J Infect Dis. 1997;176 Suppl 1:S24–8.
  • Roberts PC, Garten W, Klenk HD. Role of conserved glycosylation sites in maturation and transport of influenza a virus hemagglutinin. J Virol. 1993;67:3048–3060.
  • Ohuchi R, Ohuchi M, Garten W, et al. Oligosaccharides in the stem region maintain the influenza virus hemagglutinin in the metastable form required for fusion activity. J Virol. 1997;71:3719–3725.
  • Tsuchiya E, Sugawara K, Hongo S, et al. Role of overlapping glycosylation sequons in antigenic properties, intracellular transport and biological activities of influenza A/H2N2 virus haemagglutinin. J Gen Virol. 2002;83:3067–3074.
  • Gambaryan AS, Marinina VP, Tuzikov AB, et al. Effects of host-dependent glycosylation of hemagglutinin on receptor-binding properties on H1N1 human influenza a virus grown in MDCK cells and in embryonated eggs. Virology. 1998;247:170–177.
  • Ohuchi M, Ohuchi R, Feldmann A, et al. Regulation of receptor binding affinity of influenza virus hemagglutinin by its carbohydrate moiety. J Virol. 1997;71:8377–8384.
  • Alymova IV, York LA, Air GM, et al. Glycosylation changes in the globular head of H3N2 influenza hemagglutinin modulate receptor binding without affecting virus virulence. Sci Rep. 2016;6:36216.
  • Deshpande KL, Fried VA, Ando M, et al. Glycosylation affects cleavage of an H5N2 influenza virus hemagglutinin and regulates virulence. Proc Natl Acad Sci U S a. 1987;84:36–40.
  • Ohuchi M, Orlich M, Ohuchi R, et al. Mutations at the cleavage site of the hemagglutinin after the pathogenicity of influenza virus A/chick/Penn/83 (H5N2). Virology. 1989;168:274–280.
  • Reading PC, Pickett DL, Tate MD, et al. Loss of a single N-linked glycan from the hemagglutinin of influenza virus is associated with resistance to collectins and increased virulence in mice. Respir Res. 2009;10:117.
  • Wagner R, Wolff T, Herwig A, et al. Interdependence of hemagglutinin glycosylation and neuraminidase as regulators of influenza virus growth: a study by reverse genetics. J Virol. 2000;74:6316–6323.
  • Li S, Schulman J, Itamura S, et al. Glycosylation of neuraminidase determines the neurovirulence of influenza A/WSN/33 virus. J Virol. 1993;67:6667–6673.
  • Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–574.
  • Walls AC, Park YJ, Tortorici MA, et al. Function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020;181:281–92.e6.
  • Wrapp D, Wang N, Corbett KS, et al. Cryo-EM structure of the 2019-nCov spike in the prefusion conformation. Science. 2020;367:1260–1263.
  • Shajahan A, Archer-Hartmann S, Supekar NT, et al. Comprehensive characterization of N- and O- glycosylation of SARS-CoV-2 human receptor angiotensin converting enzyme 2. Glycobiology. 2021;31:410–424.
  • Lan J, Ge J, Yu J, et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 2020;581:215–220.
  • Tooze SA, Tooze J, Warren G. Site of addition of N-acetyl-galactosamine to the E1 glycoprotein of mouse hepatitis virus-A59. J Cell Biol. 1988;106:1475–1487.
  • Holmes KV, Doller EW, Sturman LS. Tunicamycin resistant glycosylation of coronavirus glycoprotein: demonstration of a novel type of viral glycoprotein. Virology. 1981;115:334–344.
  • Yamada Y, Liu DX. Proteolytic activation of the spike protein at a novel RRRR/S motif is implicated in furin-dependent entry, syncytium formation, and infectivity of coronavirus infectious bronchitis virus in cultured cells. J Virol. 2009;83:8744–8758.
  • Dawood AA, Altobje MA. Inhibition of N-linked glycosylation by tunicamycin may contribute to the treatment of SARS-CoV-2. Microb Pathog. 2020;149:104586.
  • Delmas B, Laude H. Assembly of coronavirus spike protein into trimers and its role in epitope expression. J Virol. 1990;64:5367–5375.
  • Voss D, Pfefferle S, Drosten C, et al. Studies on membrane topology, N-glycosylation and functionality of SARS-CoV membrane protein. Virol J. 2009;6:79.
  • Lennemann NJ, Walkner M, Berkebile AR, et al. The role of conserved N-linked glycans on Ebola Virus glycoprotein 2. J Infect Dis. 2015;212 Suppl 2:S204–9.
  • Kwak JE, Kim YI, Park SJ, et al. Development of a SFTSV DNA vaccine that confers complete protection against lethal infection in ferrets. Nat Commun. 2019;10:3836.
  • Wu Y, Zhu Y, Gao F, et al. Structures of phlebovirus glycoprotein Gn and identification of a neutralizing antibody epitope. Proc Natl Acad Sci U S a. 2017;114:E7564–e73.
  • Halldorsson S, Behrens AJ, Harlos K, et al. Structure of a phleboviral envelope glycoprotein reveals a consolidated model of membrane fusion. Proc Natl Acad Sci U S a. 2016;113:7154–7159.
  • Hofmann H, Li X, Zhang X, et al. Severe fever with thrombocytopenia virus glycoproteins are targeted by neutralizing antibodies and can use DC-SIGN as a receptor for pH-dependent entry into human and animal cell lines. J Virol. 2013;87:4384–4394.
  • Sun Y, Qi Y, Liu C, et al. Nonmuscle myosin heavy chain IIA is a critical factor contributing to the efficiency of early infection of severe fever with thrombocytopenia syndrome virus. J Virol. 2014;88:237–248.
  • Schmaljohn CS, Schmaljohn AL, Dalrymple JM. Hantaan virus M RNA: coding strategy, nucleotide sequence, and gene order. Virology. 1987;157:31–39.
  • Shi X, Elliott RM. Analysis of N-linked glycosylation of hantaan virus glycoproteins and the role of oligosaccharide side chains in protein folding and intracellular trafficking. J Virol. 2004;78:5414–5422.
  • Phoenix I, Nishiyama S, Lokugamage N, et al. N-glycans on the Rift Valley fever virus envelope glycoproteins Gn and Gc redundantly support viral infection via DC-SIGN. Viruses. 2016;8(5):149.
  • Said A, Elmanzalawy M, Ma G, et al. An equine herpesvirus type 1 (EHV-1) vector expressing Rift Valley fever virus (RVFV) Gn and Gc induces neutralizing antibodies in sheep. Virol J. 2017;14:154.
  • Wright D, Allen ER, Clark MHA, et al. Naturally acquired Rift Valley fever virus neutralizing antibodies predominantly target the Gn glycoprotein. iScience. 2020;23:101669.
  • Lam SK. Nipah virus–a potential agent of bioterrorism? Antiviral Res. 2003;57:113–119.
  • Tan CT, Wong KT. Nipah encephalitis outbreak in Malaysia. Ann Acad Med Singap. 2003;32:112–117.
  • Aguilar HC, Matreyek KA, Filone CM, et al. N-Glycans on Nipah virus fusion protein protect against neutralization but reduce membrane fusion and viral entry. J Virol. 2006;80:4878–4889.
  • Biering SB, Huang A, Vu AT, et al. N-Glycans on the Nipah virus attachment glycoprotein modulate fusion and viral entry as they protect against antibody neutralization. J Virol. 2012;86:11991–12002.
  • Steffen DL, Xu K, Nikolov DB, et al. Henipavirus mediated membrane fusion, virus entry and targeted therapeutics. Viruses. 2012;4:280–308.
  • Martínez I, Melero JA. Binding of human respiratory syncytial virus to cells: implication of sulfated cell surface proteoglycans. J Gen Virol. 2000;81:2715–2722.
  • Leemans A, Boeren M, Van der Gucht W, et al. Characterization of the role of N-glycosylation sites in the respiratory syncytial virus fusion protein in virus replication, syncytium formation and antigenicity. Virus Res. 2019;266:58–68.
  • Leemans A, Boeren M, Van der Gucht W, et al. Removal of the N-glycosylation sequon at position N116 located in p27 of the respiratory Syncytial virus fusion protein elicits enhanced antibody responses after DNA immunization. Viruses. 2018;10.
  • Wright KE, Spiro RC, Burns JW, et al. Post-Translational processing of the glycoproteins of lymphocytic choriomeningitis virus. Virology. 1990;177:175–183.
  • Wright KE, Salvato MS, Buchmeier MJ. Neutralizing epitopes of lymphocytic choriomeningitis virus are conformational and require both glycosylation and disulfide bonds for expression. Virology. 1989;171:417–426.
  • Bonhomme CJ, Knopp KA, Bederka LH, et al. LCMV glycosylation modulates viral fitness and cell tropism. PLoS One. 2013;8:e53273.
  • Branco LM, Garry RF. Characterization of the Lassa virus GP1 ectodomain shedding: implications for improved diagnostic platforms. Virol J. 2009;6:147.
  • Burri DJ, JR da P, Kunz S, et al. Envelope glycoprotein of arenaviruses. Viruses. 2012;4:2162–2181.
  • Bowen MD, Rollin PE, Ksiazek TG, et al. Genetic diversity among Lassa virus strains. J Virol. 2000;74:6992–7004.
  • Lenz O, Ter Meulen J, Klenk HD, et al. The Lassa virus glycoprotein precursor GP-C is proteolytically processed by subtilase SKI-1/S1P. Proc Natl Acad Sci U S A. 2001;98:12701–12705.
  • Rojek JM, Pasqual G, Sanchez AB, et al. Targeting the proteolytic processing of the viral glycoprotein precursor is a promising novel antiviral strategy against arenaviruses. J Virol. 2010;84:573–584.
  • Shrivastava-Ranjan P, Bergeron É, Chakrabarti AK, et al. 25-hydroxycholesterol inhibition of Lassa virus infection through aberrant GP1 glycosylation. mBio. 2016;7(6):e01808–16.
  • Zhu S, Wan W, Zhang Y, et al. Comprehensive interactome analysis reveals that STT3B is required for N-glycosylation of Lassa virus glycoprotein. J Virol. 2019;93(23):e01443–19.
  • Dobrica MO, Lazar C, Branza-Nichita N. N-glycosylation and N-glycan processing in HBV biology and pathogenesis. Cells. 2020;9(6):1404.
  • Bruss V, Vieluf K. Functions of the internal pre-S domain of the large surface protein in hepatitis B virus particle morphogenesis. J Virol. 1995;69:6652–6657.
  • Lazar C, Durantel D, Macovei A, et al. Treatment of hepatitis B virus-infected cells with alpha-glucosidase inhibitors results in production of virions with altered molecular composition and infectivity. Antiviral Res. 2007;76:30–37.
  • Spear PG, Manoj S, Yoon M, et al. Different receptors binding to distinct interfaces on herpes simplex virus gD can trigger events leading to cell fusion and viral entry. Virology. 2006;344:17–24.
  • Arii J, Wang J, Morimoto T, et al. A single-amino-acid substitution in herpes simplex virus 1 envelope glycoprotein B at a site required for binding to the paired immunoglobulin-like type 2 receptor alpha (PILRalpha) abrogates PILRalpha-dependent viral entry and reduces pathogenesis. J Virol. 2010;84:10773–10783.
  • Lu H, Cherepanova NA, Gilmore R, et al. Targeting STT3A-oligosaccharyltransferase with NGI-1 causes herpes simplex virus 1 dysfunction. Faseb J. 2019;33:6801–6812.
  • Antoine TE, Park PJ, Shukla D. Glycoprotein targeted therapeutics: a new era of anti-herpes simplex virus-1 therapeutics. Rev Med Virol. 2013;23:194–208.
  • Eisenberg RJ, Atanasiu D, Cairns TM, et al. Herpes virus fusion and entry: a story with many characters. Viruses. 2012;4:800–832.
  • Akhtar J, Shukla D. Viral entry mechanisms: cellular and viral mediators of herpes simplex virus entry. Febs J. 2009;276:7228–7236.
  • Herold BC, Visalli RJ, Susmarski N, et al. Glycoprotein C-independent binding of herpes simplex virus to cells requires cell surface heparan sulphate and glycoprotein B. J Gen Virol. 1994;75(Pt 6):1211–1222.
  • Herold BC, WuDunn D, Soltys N, et al. Glycoprotein C of herpes simplex virus type 1 plays a principal role in the adsorption of virus to cells and in infectivity. J Virol. 1991;65:1090–1098.
  • Pertel PE, Fridberg A, Parish ML, et al. Cell fusion induced by herpes simplex virus glycoproteins gB, gD, and gH-gL requires a gD receptor but not necessarily heparan sulfate. Virology. 2001;279:313–324.
  • Hansen JE, Lund O, Engelbrecht J, et al. Prediction of O-glycosylation of mammalian proteins: specificity patterns of UDP-GalNac:polypeptide N-acetylgalactosamin-yltransferase. Biochem J. 1995;308(Pt 3):801–813.
  • Peng T, Ponce de Leon M, Novotny MJ, et al. Structural and antigenic analysis of a truncated form of the herpes simplex virus glycoprotein gH-gL complex. J Virol. 1998;72:6092–6103.
  • Kornfeld R, Kornfeld S. Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem. 1985;54:631–664.
  • Hutchinson L, Browne H, Wargent V, et al. A novel herpes simplex virus glycoprotein, gL, forms a complex with glycoprotein H (gH) and affects normal folding and surface expression of gH. J Virol. 1992;66:2240–2250.
  • Nordén R, Halim A, Nyström K, et al. O-Linked glycosylation of the mucin domain of the herpes simplex virus type 1-specific glycoprotein gC-1 is temporally regulated in a seed-and-spread manner. J Biol Chem. 2015;290:5078–5091.
  • Serafini-Cessi F, Dall’-Olio F, Malagolini N, et al. Comparative study on O-linked oligosaccharides of glycoprotein D of herpes simplex virus types 1 and 2. J Gen Virol. 1988;69(Pt 4):869–877.
  • Isaacs CE, Xu W, Pullarkat RK, et al. Retinoic acid reduces the yield of herpes simplex virus in Vero cells and alters the N-glycosylation of viral envelope proteins. Antiviral Res. 2000;47:29–40.
  • Serafini-Cessi F, Dall’-Olio F, Scannavini M, et al. Processing of herpes simplex virus-1 glycans in cells defective in glycosyl transferases of the Golgi system: relationship to cell fusion and virion egress. Virology. 1983;131:59–70.
  • Bagdonaite I, Nordén R, Joshi HJ, et al. A strategy for O-glycoproteomics of enveloped viruses–the O-glycoproteome of herpes simplex virus type 1. PLoS Pathog. 2015;11:e1004784.
  • Yao Z, Jackson W, Forghani B, et al. Varicella-Zoster virus glycoprotein gpI/gpiv receptor: expression, complex formation, and antigenicity within the vaccinia virus-T7 RNA polymerase transfection system. J Virol. 1993;67:305–314.
  • Maresová L, Kutinová L, Ludvíková V, et al. Characterization of interaction of gH and gL glycoproteins of varicella-zoster virus: their processing and trafficking. J Gen Virol. 2000;81:1545–1552.
  • Yamagishi Y, Sadaoka T, Yoshii H, et al. Varicella-Zoster virus glycoprotein M homolog is glycosylated, is expressed on the viral envelope, and functions in virus cell-to-cell spread. J Virol. 2008;82:795–804.
  • Mo C, Lee J, Sommer M, et al. The requirement of varicella zoster virus glycoprotein E (gE) for viral replication and effects of glycoprotein I on gE in melanoma cells. Virology. 2002;304:176–186.
  • Montalvo EA, Parmley RT, Grose C. Structural analysis of the varicella-zoster virus gp98-gp62 complex: posttranslational addition of N-linked and O-linked oligosaccharide moieties. J Virol. 1985;53:761–770.
  • Yamada K, Noguchi K, Nishizono A. Efficient N-glycosylation at position 37, but not at position 146, in the street rabies virus glycoprotein reduces pathogenicity. Virus Res. 2014;179:169–176.
  • Zhou J, Sun XY, Frazer IH. Glycosylation of human papillomavirus type 16 L1 protein. Virology. 1993;194:210–218.
  • Schowalter RM, Smith SE, Dutch RE. Characterization of human metapneumovirus F protein-promoted membrane fusion: critical roles for proteolytic processing and low pH. J Virol. 2006;80:10931–10941.
  • Apte-Sengupta S, Sirohi D, Kuhn RJ. Coupling of replication and assembly in flaviviruses. Curr Opin Virol. 2014;9:134–142.
  • Lopez-Denman AJ, Mackenzie JM. The IMPORTance of the nucleus during flavivirus replication. Viruses. 2017;9(1):14.
  • Yu L, Nomaguchi M, Padmanabhan R, et al. Specific requirements for elements of the 5’ and 3’ terminal regions in flavivirus RNA synthesis and viral replication. Virology. 2008;374:170–185.
  • Putnak JR, Charles PC, Padmanabhan R, et al. Functional and antigenic domains of the dengue-2 virus nonstructural glycoprotein NS-1. Virology. 1988;163:93–103.
  • Winkler G, Maxwell SE, Ruemmler C, et al. Newly synthesized dengue-2 virus nonstructural protein NS1 is a soluble protein but becomes partially hydrophobic and membrane-associated after dimerization. Virology. 1989;171:302–305.
  • Wang C, Puerta-Guardo H, Biering SB, et al. Endocytosis of flavivirus NS1 is required for NS1-mediated endothelial hyperpermeability and is abolished by a single N-glycosylation site mutation. PLoS Pathog. 2019;15:e1007938.
  • Fan J, Liu Y, Yuan Z. Critical role of Dengue Virus NS1 protein in viral replication. Virol Sin. 2014;29:162–169.
  • Tajima S, Takasaki T, Kurane I. Characterization of Asn130-to-Ala mutant of dengue type 1 virus NS1 protein. Virus Genes. 2008;36:323–329.
  • Pryor MJ, Gualano RC, Lin B, et al. Growth restriction of dengue virus type 2 by site-specific mutagenesis of virus-encoded glycoproteins. J Gen Virol. 1998;79(Pt 11):2631–2639.
  • Pletnev AG, Bray M, Lai CJ. Chimeric tick-borne encephalitis and dengue type 4 viruses: effects of mutations on neurovirulence in mice. J Virol. 1993;67:4956–4963.
  • Crabtree MB, Kinney RM, Miller BR. Deglycosylation of the NS1 protein of dengue 2 virus, strain 16681: construction and characterization of mutant viruses. Arch Virol. 2005;150:771–786.
  • Puerta-Guardo H, Glasner DR, Espinosa DA, et al. Flavivirus NS1 triggers tissue-specific vascular endothelial dysfunction reflecting disease tropism. Cell Rep. 2019;26:1598–613.e8.
  • Muylaert IR, Chambers TJ, Galler R, et al. 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.
  • Whiteman MC, Li L, Wicker JA, et al. Development and characterization of non-glycosylated E and NS1 mutant viruses as a potential candidate vaccine for West Nile virus. Vaccine. 2010;28:1075–1083.
  • Kuno G, Chang GJ. Characterization of Sepik and Entebbe bat viruses closely related to yellow fever virus. Am J Trop Med Hyg. 2006;75:1165–1170.
  • Mandl CW, Heinz FX, Stöckl E, et al. Genome sequence of tick-borne encephalitis virus (Western subtype) and comparative analysis of nonstructural proteins with other flaviviruses. Virology. 1989;173:291–301.
  • Whiteman MC, Wicker JA, Kinney RM, et al. Multiple amino acid changes at the first glycosylation motif in NS1 protein of West Nile virus are necessary for complete attenuation for mouse neuroinvasiveness. Vaccine. 2011;29:9702–9710.
  • Gadlage MJ, Sparks JS, Beachboard DC, et al. Murine hepatitis virus nonstructural protein 4 regulates virus-induced membrane modifications and replication complex function. J Virol. 2010;84:280–290.
  • Fung TS, Liu DX. Post-Translational modifications of coronavirus proteins: roles and function. Future Virol. 2018;13:405–430.
  • Diaz Y, Chemello ME, Pena F, et al. Expression of nonstructural rotavirus protein NSP4 mimics Ca2+ homeostasis changes induced by rotavirus infection in cultured cells. J Virol. 2008;82:11331–11343.
  • Gouttenoire J, Castet V, Montserret R, et al. Identification of a novel determinant for membrane association in hepatitis C virus nonstructural protein 4B. J Virol. 2009;83:6257–6268.
  • Gouttenoire J, Montserret R, Kennel A, et al. An amphipathic alpha-helix at the C terminus of hepatitis C virus nonstructural protein 4B mediates membrane association. J Virol. 2009;83:11378–11384.