Article

Mutation in a 17D-204 Vaccine Substrain-Specific Envelope Protein Epitope Alters the Pathogenesis of Yellow Fever Virus in Mice

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

The heterogeneous nature of the yellow fever (YF) 17D-204 vaccine virus population was exploited in this study to isolate virus variants able to escape neutralization by the 17D-204 vaccine-specific MAb 864. The conformational change on the virus surface that resulted in the loss of the MAb 864-defined epitope was effected in each variant by a single amino acid mutation in the envelope (E) protein at either position E-305 or E-325. Interestingly, both positions were mutated during attenuation of the 17D-204 vaccine substrain from the wildtype Asibi strain. The mutations in several of the variants represented reversion to the wildtype Asibi virus sequence consistent with loss of a 17D-204 substrain-specific epitope. The majority of the variant viruses were shown to have altered mouse neurovirulence phenotypes, ranging from complete avirulence through to increased virulence. The avirulent variants are the first flavivirus MAb-neutralization-resistant variants to be attenuated for neurovirulence in the adult mouse model. Overall, the results indicate that the E protein epitope recognized by MAb 864 defines a functionally important region that encodes major molecular determinants of YF virus pathogenesis in vivo.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... One exception is the 17D-204 vaccine substrain-specific epitope identified using mAb 864, which elicits high neutralizing activity allowing for mAb neutralization resistant mutants to be generated. Using these techniques mAb resistance was mapped to the epitope that include residues E-305 and E-325 on EDIII 18 . ...
... Eight of the 20 residues reside in the E protein and are located in all three domains in the N-terminal ectodomain plus the transmembrane domain. In addition, E-325 is part of the 17D-204 vaccine substrain-specific epitope 18 . ...
... A previous study used plaque-purified mutants from 17D-204 to identify E-173 in this WT epitope 18 , one of the eight residues that distinguishes Asibi from 17D. EDIII, an Ig-like domain, has been implicated in receptor binding with regions of the upper lateral surface shown to be important to a 17D-204 substrain-specific epitope, mouse neurovirulence, and rate of viral clearance in mice 18,37,46 . The 17D-204 specific vaccine epitope was mapped to E-305 and E-325 within EDIII with mAb 864 and was shown to be distinct from the vaccine-specific mAb 411 used here through direct competition assays 18,47 . ...
Article
Full-text available
The envelope (E) protein of flaviviruses is functionally associated with viral tissue tropism and pathogenicity. For yellow fever virus (YFV), viscerotropic disease primarily involving the liver is pathognomonic for wild-type (WT) infection. In contrast, the live-attenuated vaccine (LAV) strain 17D does not cause viscerotropic disease and reversion to virulence is associated with neurotropic disease. The relationship between structure-function of the E protein for WT strain Asibi and its LAV derivative 17D strain is poorly understood; however, changes to WT and vaccine epitopes have been associated with changes in virulence. Here, a panel of Asibi and 17D infectious clone mutants were generated with single-site mutations at the one membrane residue and each of the eight E protein amino acid substitutions that distinguish the two strains. The mutants were characterized with respect to WT-specific and vaccine-specific monoclonal antibodies (mAbs) binding to virus plus binding of virus to brain, liver, and lung membrane receptor preparations (MRPs) generated from AG129 mice. This approach shows that amino acids in the YFV E protein domains (ED) I and II contain the WT E protein epitope, which overlap with those that mediate YFV binding to mouse liver. Furthermore, amino acids in EDIII associated with the vaccine epitope overlap with those that facilitate YFV binding mouse brain MRPs. Taken together, these data suggest that the YFV E protein is a key determinant in the phenotype of WT and 17D vaccine strains of YFV.
... Because the timing of viral exposure is known for vacinees, individuals experiencing post-vaccinal SAEs are likely candidates for anti-YF antibody therapy. One theoretical limitation of single MAb therapy for flaviruses is the high mutation rate of flaviviral ssRNA genomes, which could result in generation of MAb escape mutants (Ryman et al., 1998). Neutralization escape variants of WNV have been selected both in vitro and in vivo following single dose MAb treatment (Zhang et al., 2009(Zhang et al., , 2010. ...
... MAb 864 is substrain specific and reacts only with YFV 17D-204 vaccine, neutralizes virus infectivity, and has been shown to protect mice from virus challenge when administered to 3e4 week-old immunocompetent mice as mouse ascitic fluid 24 h before YF-17D challenge via the intracerebral route Gould et al., 1986). Unlike mMAb 2C9, mMAb 864 identifies a neutralization epitope in DIII of the E protein (Ryman et al., 1998); thus we predicted that combined therapy using 864-cIgG and 2C9-cIgG should increase therapeutic efficacy compared to 2C9-cIgG alone. ...
... As expected for a YFV type-specific epitope, these residues are identical in the parental YFV Asibi strain and 17D-204. A substrainspecific epitope in DIII (shown in blue) of the 17D-204 E protein was identified in which mutation of either AA F305 or S325 resulted in loss of binding m864 (Ryman et al., 1998). These positions are occupied by S305 and P325 in the parental YFV Asibi strain and m864 does not bind wild type YFV . ...
Article
The yellow fever virus (YFV) vaccine 17D-204 is considered safe and effective, yet rare severe adverse events (SAEs), some resulting in death, have been documented following vaccination. Individuals exhibiting post-vaccinal SAEs are ideal candidates for antiviral monoclonal antibody (MAb) therapy; the time until appearance of clinical signs post-exposure is usually short and patients are quickly hospitalized. We previously developed a murine-human chimeric monoclonal antibody (cMAb), 2C9-cIgG, reactive with both virulent YFV and 17D-204, and demonstrated its ability to prevent and treat YF disease in both AG129 mouse and hamster models of infection. To counteract possible selection of 17D-204 variants that escape neutralization by treatment with a single MAb (2C9-cIgG), we developed a second cMAb, 864-cIgG, for use in combination with 2C9-cIgG in post-vaccinal therapy. MAb 864-cIgG recognizes/neutralizes only YFV 17D-204 vaccine substrain and binds to domain III (DIII) of the viral envelope protein, which is different from the YFV type-specific binding site of 2C9-cIgG in DII. Although it neutralized 17D-204 in vitro, administration of 864-cIgG had no protective capacity in the interferon receptor-deficient AG129 mouse model of 17D-204 infection. The data presented here show that although DIII-specific 864-cIgG neutralizes virus infectivity in vitro, it does not have the ability to abrogate disease in vivo. Therefore, combination of 864-cIgG with 2C9-cIgG for treatment of YF vaccination SAEs does not appear to provide an improvement on 2C9-cIgG therapy alone.
... This syndrome is sometimes observed as a complication after vaccination of humans with the YFV 17D strain (9). Strains of YFV are known to differ in their neurovirulence potentials (2,8,18,31,32), and this property can be enhanced by serial passage of the virus in brain tissue (7,25,34,36). Mutations in numerous regions of the viral genome have been identified in such neuroadapted viruses, although the viral E envelope protein is believed to contain the most important determinants of this virulence (3,33). ...
... Mutations in numerous regions of the viral genome have been identified in such neuroadapted viruses, although the viral E envelope protein is believed to contain the most important determinants of this virulence (3,33). Mutations in the flavivirus E protein are likely to contribute enhanced virulence through effects on virus entry into host cells and spread to critical target tissues (11,20,22,23,27,28,31,32), although the exact mechanisms involved in this process have not been defined. ...
... The results of the current experiments confirm that for YFV, a region within domain III of the E protein governs neuroinvasiveness as assessed in the SCID mouse model. This contrasts with other studies that have implicated molecular determinants within domains I, II, and III, as well as the stem-anchor region of the E protein, in the virulence of flaviviruses (1,11,19,20,27,28,30,31,32,35). ...
Article
Full-text available
A molecular clone of yellow fever virus (YFV) strain 17D was used to identify critical determinants of mouse neuroinvasiveness previously localized to domain III of the neuroadapted SPYF-MN virus envelope protein. Three candidate virulence substitutions (305F-->V, 326K-->E, and 380R-->T) were individually evaluated for their roles in this phenotype in a SCID mouse model. The virus containing a glutamic acid residue at position 326 of the envelope protein (326E) caused rapidly lethal encephalitis, with a mortality rate and average survival time resembling those of the parental SPYF-MN virus. Determinants at positions 380 (380T) and 305 (305V) did not independently affect neuroinvasiveness. Testing a panel of viruses with various amino acid substitutions at position 326 revealed that attenuation of neuroinvasiveness required a positively charged residue (lysine or arginine) at this position. Molecular-modeling studies suggest that residues 326 and 380 contribute to charge clusters on the lateral surface of domain III that constitute putative heparin binding sites, as confirmed by studies of heparin inhibition of plaque formation. The neuroinvasiveness of YFVs in the SCID model correlated inversely with sensitivity to heparin. These findings establish that residue 326 in domain III of the E protein is a critical determinant of YFV neuroinvasiveness in the SCID mouse model. Together with modeling of domain III from virulent YFV strains, the data suggest that heparin binding activity involving lysine at position 326 may be a modulator of YFV virulence phenotypes.
... Introduction of virus into the murine central nervous system (CNS) causes an acute encephalitis, the outcome of which can be influenced by the dose of virus and the age and strain of the mouse (15). Different strains of yellow fever virus (YFV) can be distinguished by their level of mouse neurovirulence (5,15,28,(52)(53)(54), and this property can be enhanced by serial passage of the virus in mouse brain (37,56,59). This neuroadaptation has also been observed with other flaviviruses (7,11,25). ...
... The role of these various mutations in altering those functional properties of the flavivirus E protein which increase neurovirulence is not well understood. It is assumed that receptor binding and subsequent events associated with virus entry, including low-pH-induced conformational changes and fusion with intracellular membranes, are principally involved (6,32,34,35,45,46,52,54). In contrast to the E protein, neurovirulence determinants in the nonstructural and untranslated regions have been less well studied (8,14,25,31,38,43,47). ...
... Positions 326 and 380 lie within the putative receptor binding domain at two distinct regions which have been proposed as critical for the function of this portion of the E protein (6). A YF17D substrain-specific epitope is known to involve the adjacent residue 325, whose substitution is associated with alteration in mouse neurovirulence (54). Residue 380 is included within the RGD motif of mosquito-borne flaviviruses, in which mutations have been shown to affect the efficiency of virus spread in cell culture and neuroinvasion in mice (29,60). ...
Article
Full-text available
A neuroadapted strain of yellow fever virus (YFV) 17D derived from a multiply mouse brain-passaged virus (Porterfield YF17D) was additionally passaged in SCID and normal mice. The virulence properties of this virus (SPYF) could be distinguished from nonneuroadapted virus (YF5.2iv, 17D infectious clone) by decreased average survival time in SCID mice after peripheral inoculation, decreased average survival time in normal adult mice after intracerebral inoculation, and occurrence of neuroinvasiveness in normal mice. SPYF exhibited more efficient growth in peripheral tissues of SCID mice than YF5.2iv, resulting in a more rapid accumulation of virus burden, but with low-titer viremia, at the time of fatal encephalitis. In cell culture, SPYF was less efficient in replication than YF5.2iv in all cell lines tested. The complete nucleotide sequence of SPYF revealed 29 nucleotide substitutions relative to YF5.2iv, and these were distributed throughout the genome. There were a total of 13 predicted amino acid substitutions, some of which correspond to known differences among the Asibi, French viscerotropic virus, French neurotropic vaccine, and YF17D vaccine strains. The envelope (E) protein contained five substitutions, within all three functional domains. Substitutions were also present in regions encoding the NS1, NS2A, NS4A, and NS5 proteins and in the 3' untranslated region (UTR). Construction of YFV harboring all of the identified coding nucleotide substitutions and those in the 3' UTR yielded a virus whose cell culture and pathogenic properties, particularly neurovirulence and neuroinvasiveness for SCID mice, generally resembled those of the original SPYF isolate. These findings implicate the E protein and possibly other regions of the genome as virulence determinants during pathogenesis of neuroadapted YF17D virus in mice. The determinants affect replication efficiency in both neural and extraneural tissues of the mouse and confer some limited host-range differences in cultured cells of nonmurine origin.
... A recent study found that substitution at TMUV E-304 can alter its virulence, and the substitution of basic residues can markedly increase the affinity of the virus for GAGs (Yang et al., 2021). A YFV variant that escapes neutralization by a monoclonal antibody (mAb) exhibited a conformational change in the E protein, and the E-305 residue was identified to be one of the epitopes bound by the mAb and one of the determinants of YFV pathogenesis in vivo (Ryman et al., 1998). A panel of WNV mutants containing all possible amino acid substitutions at E-332 revealed that the change in E-332 had a great influence on the antigenicity of the virus (Plante et al., 2016). ...
... With artificial modifications or by natural selection, certain amino acids may be mutated, and these mutations can cause the phenotype of the virus to change (Ramond et al., 2019). This variation may manifest as a change in the infectivity of the virus in vitro, virulence in vivo, or the recognition of neutralizing antibodies (NAbs) (Ryman et al., 1998). In domain III of flavivirus E protein, a number of decisive sites involved in NAb recognition or virus infectivity have been previously identified (Mandl et al., 2000;Matsui et al., 2010). ...
Article
Full-text available
Flavivirus envelope protein (E) plays an important role in cellular infection, especially in virulence and antigenicity. E domain III of Tembusu virus (TMUV) is highly conserved among flaviviruses and contains four loop regions. However, the functions of the loop regions of TMUV E domain III in the viral life cycle have not yet been discovered. In this study, using a reverse genetics system, we performed site-directed mutagenesis on loops I, II, III, and IV of TMUV E domain III. Mutant 6 (S388A.G389A.K390A) showed better proliferation than the wild-type virus, while mutants 1–5 exhibited decreased in vitro infectivity, as determined by immunofluorescence assay (IFA). Based on a TMUV replicon system, the mutations exhibited no apparent effect on TMUV RNA replication. Subcellular fractionation assays and packaging system assays indicated that mutations in loops II–IV (T332A, T332S, S365A.S366A.T367A, and S388A.G389A.K390A, respectively) disrupted virion assembly. Moreover, loops I–IV played an important role in virus binding and entry, while mutant 6 (S388A.G389A.K390A) exhibited robust activity in virus entry. Taken together, our findings indicated the critical role of the loop regions in TMUV E domain III in the virus entry and assembly process.
... A316 influences binding to glycosaminoglycans (residues 325-326 and 380) or other cell receptors, modulates the efficacy of virus spread, neuroinvasiveness and neurovirulence -diminishes infection rates in Aedes aegypti mosquitoes (YFV), not studied in Culex mosquitoes [192] YFV, DENV S305F [181,[193][194][195] TBEV D308K [197] JEV A315V [198] YFV S325P, E326K/R [193,195,196] YFV, MVEV R380T [193,194,199,200] NS1 N-glycosylation site DENV N130A + N208A N130 + N207 ablation of the first glycosylation site (N130) decreases replication, viral production and neurovirulence and diminishes vector competence of Culex tarsalis mosquitoes for WNV [177,198] YFV N130A + N208A Decreased replication and neurovirulence (1st glycosylation site) [116,201,202] 3 UTR Deletion of nucleotides JEV −27nt not investigated on WNV Attenuates or increases (TBEV) neurovirulence [203] DENV −4 nt [204,205] TBEV −206 nt [206] It would also be a good starting point to compare the virulence and pathogenesis of the WNV with those of other flaviviruses of interest, such as the dengue and yellow fever viruses [97]. It is noteworthy that the study of flavivirus molecular determinants has been driven by the identification in the field or by artificial selection of mutants with properties of interest, such as attenuation, neuroadaptation, and escape from neutralization. ...
... A316 influences binding to glycosaminoglycans (residues 325-326 and 380) or other cell receptors, modulates the efficacy of virus spread, neuroinvasiveness and neurovirulence -diminishes infection rates in Aedes aegypti mosquitoes (YFV), not studied in Culex mosquitoes [192] YFV, DENV S305F [181,[193][194][195] TBEV D308K [197] JEV A315V [198] YFV S325P, E326K/R [193,195,196] YFV, MVEV R380T [193,194,199,200] NS1 N-glycosylation site DENV N130A + N208A N130 + N207 ablation of the first glycosylation site (N130) decreases replication, viral production and neurovirulence and diminishes vector competence of Culex tarsalis mosquitoes for WNV [177,198] YFV N130A + N208A Decreased replication and neurovirulence (1st glycosylation site) [116,201,202] 3 UTR Deletion of nucleotides JEV −27nt not investigated on WNV Attenuates or increases (TBEV) neurovirulence [203] DENV −4 nt [204,205] TBEV −206 nt [206] It would also be a good starting point to compare the virulence and pathogenesis of the WNV with those of other flaviviruses of interest, such as the dengue and yellow fever viruses [97]. It is noteworthy that the study of flavivirus molecular determinants has been driven by the identification in the field or by artificial selection of mutants with properties of interest, such as attenuation, neuroadaptation, and escape from neutralization. ...
Article
Full-text available
West Nile virus (WNV), like the dengue virus (DENV) and yellow fever virus (YFV), are major arboviruses belonging to the Flavivirus genus. WNV is emerging or endemic in many countries around the world, affecting humans and other vertebrates. Since 1999, it has been considered to be a major public and veterinary health problem, causing diverse pathologies, ranging from a mild febrile state to severe neurological damage and death. WNV is transmitted in a bird–mosquito–bird cycle, and can occasionally infect humans and horses, both highly susceptible to the virus but considered dead-end hosts. Many studies have investigated the molecular determinants of WNV virulence, mainly with the ultimate objective of guiding vaccine development. Several vaccines are used in horses in different parts of the world, but there are no licensed WNV vaccines for humans, suggesting the need for greater understanding of the molecular determinants of virulence and antigenicity in different hosts. Owing to technical and economic considerations, WNV virulence factors have essentially been studied in rodent models, and the results cannot always be transported to mosquito vectors or to avian hosts. In this review, the known molecular determinants of WNV virulence, according to invertebrate (mosquitoes) or vertebrate hosts (mammalian and avian), are presented and discussed. This overview will highlight the differences and similarities found between WNV hosts and models, to provide a foundation for the prediction and anticipation of WNV re-emergence and its risk of global spread.
... 56 Moreover, mutations at E-173 and E-305 are integral components of neutralizing epitopes mapped by monoclonal antibody escape mutants. 104,105 The loss of a 17D-204 substrain-specific epitope from the 17D virus, selected for neutralization escape, resulted in dramatic changes in neurovirulence ranging from avirulence to increased virulence. 104 Although less well studied, mutations in other locations, particularly in the nonstructural proteins, are also being considered as determinants of YFV virulence. ...
... 104,105 The loss of a 17D-204 substrain-specific epitope from the 17D virus, selected for neutralization escape, resulted in dramatic changes in neurovirulence ranging from avirulence to increased virulence. 104 Although less well studied, mutations in other locations, particularly in the nonstructural proteins, are also being considered as determinants of YFV virulence. 101,106,107 ...
Article
Yellow fever (YF) is a viral disease, endemic to tropical regions of Africa and the Americas, which principally affects humans and nonhuman primates and is transmitted via the bite of infected mosquitoes. Yellow fever virus (YFV) can cause devastating epidemics of potentially fatal, hemorrhagic disease. Despite mass vaccination campaigns to prevent and control these outbreaks, the risk of major YF epidemics, especially in densely populated, poor urban settings, both in Africa and South America, has greatly increased. Consequently, YF is considered an emerging, or reemerging disease of considerable importance. This article comprehensively reviews the history, microbiology, epidemiology, clinical presentation, diagnosis, and treatment of YFV, as well as the vaccines produced to combat YF.
... This ectodomain forms a homodimer, with each dimer subunit organized into three domains, designated I, II, and III. Comparisons of E proteins from wild-type viruses with those of attenuated or escape mutant viruses have identified a number of residues in domain III that may be responsible for receptor recognition (4,8,(12)(13)(14). We examined the solution properties of domain III of the E protein from Langat virus (a tick-borne flavivirus) and the ability of this domain to function as an antagonist for virus infectivity. ...
... Thus, the structural and chemical properties of region 1 may be similar among vector-specific flaviviruses, allowing this region to form vectorspecific interactions with cell surface receptors. Recent studies showed that mutations in region 1 were associated with virus attenuation (8,13,14). Region 2, formed by residues 324, 355, 376, 378, 381, 382, and 394, made extensive contacts between the ␤-sheet termini of domain III (Fig. 4). The majority of these residues were partially buried within the domain III structure and thus likely shielded from intermolecular interac-tions. ...
Article
Full-text available
The molecular determinants responsible for flavivirus host cell binding and tissue tropism are largely unknown, although domain III of the envelope protein has been implicated in these functions. We examined the solution properties and antagonist activity of Langat virus domain III. Our results suggest that domain III adopts a stably folded structure that can mediate binding of tick-borne flaviviruses but not mosquito-borne flaviviruses to their target cells. Three clusters of phylogenetically conserved residues are identified that may be responsible for the vector-specific antagonist activity of domain III.
... A live-attenuated vaccine (YFV strain 17D and related substrains), which induces humoral immunity against the currently known six genotypes of YFV, was developed in the 1930s through serial passage of wild-type YFV on different cell substrates (Monath, 2004). Neutralizing antibodies are predominantly directed against the envelope glycoprotein E of the virus (Brandriss et al., 1986;Gould et al., 1986;Pincus et al., 1992), which for other flaviviruses has been shown to govern the host cell receptor binding and endosomal pHdependent membrane fusion (Lindenbach and Rice, 2001 protein amino acids (aa) E-71, E-72, E-155, E-158, E-173, E-240, E-305 and E-325 have been mapped on the monomeric E protein using mouse monoclonal antibodies, but no molecular studies of human antibody responses to YFV have been performed to date (Lobigs et al., 1987;Ryman et al., 1997aRyman et al., , 1997bRyman et al., , 1998. ...
... The ectodomain of the E protein folds into three distinct domains, a central domain (I), a dimerization and fusion domain (II) and an IgG-like domain responsible for interaction with the unknown cellular receptor (III). Mapping of neutralizing epitopes on the YFV-E protein using mouse monoclonal antibodies has previously revealed critical amino acid residues in domain II (aa E-71/72, E-173), domain III (E-305/325) and domain I (aa E-155/158) (Lobigs et al., 1987;Ryman et al., 1997aRyman et al., , 1997bRyman et al., , 1998. ...
Article
Human monoclonal antibody fragments neutralizing wild-type and vaccine strains of yellow fever (YF) virus (genotypes West Africa I + II, East/Central Africa, 17D-204-WHO) were generated by repertoire cloning from YF patients. Analysis of virus escape variants identified amino acid (aa) 71 in domain II of the envelope glycoprotein (E) as the most critical residue for neutralization, with aa 153-155 in domain I contributing to the epitope. These data confirm the previous mapping of YFV neutralizing epitopes using mouse monoclonal antibodies but suggest that a conformational epitope could be formed by amino acids from domains I and II opposing each other in the dimeric form of the E protein. While the sera of the YF patients showed up to 10-fold reduced neutralizing activity against the 17D escape variants, sera from 17D vaccinees retained their neutralizing titers. Mutations in this major neutralizing epitope of YFV thus do not seem to carry the risk of immune escape in persons immunized with the YFV-17D vaccine.
... Investigating the antigenic and genetic homogeneity of the HAdV-4 and HAdV-7 vaccines has not been conducted prior to this study. Heterogeneity of other licensed live virus vaccines, including live-attenuated yellow fever virus, Polio virus and mumps virus, have been described and shown to influence phenotypic outcomes [14][15][16][17][18][19]. ...
Article
Full-text available
The FDA-approved Adenovirus Type 4 and Type 7 Vaccine, Live, Oral is highly effective and essential in preventing acute respiratory diseases (ARDs) in U.S. military recruits. Our study revealed the presence of a previously undetected mutation, not found in the wild-type human adenovirus type 4 (HAdV-4) component of the licensed vaccine, which contains an amino acid substitution (P388T) in the pre-terminal protein (pTP). This study demonstrated that replication of the T388 HAdV-4 vaccine mutant virus is favored over the wild type in WI-38 cells, the cell type utilized in vaccine manufacturing. However, results from serial human stool specimens of vaccine recipients support differential genome replication in the gastrointestinal tract (GI), demonstrated by the steady decline of the percentage of mutant T388 vaccine virus. Since vaccine efficacy depends upon GI replication and the subsequent immune response, the mutation can potentially impact vaccine efficacy.
... A complicated passaging process was required for the acquisition of YFV17D, and during this process, changes in 32 amino acids changed the entire viral protein [96]. Among these differences, residues 325 and 380 located in domain III were shown to be related to virulence in mice [56,57,97]. Site-directed mutations at residues 325 and 380 of wild-type YFV were used to determine the effect of the mutation site on the binding ability of the virus to attachment factors (GAGs); the two substitutions significantly reduced sensitivity to heparin inhibition, implying a role in viral attachment [98]. ...
Article
Full-text available
Flaviviruses are enveloped viruses that infect multiple hosts. Envelope proteins are the outermost proteins in the structure of flaviviruses and mediate viral infection. Studies indicate that flaviviruses mainly use envelope proteins to bind to cell attachment receptors and endocytic receptors for the entry step. Here, we present current findings regarding key envelope protein amino acids that participate in the flavivirus early infection process. Among these sites, most are located in special positions of the protein structure, such as the α-helix in the stem region and the hinge region between domains I and II, motifs that potentially affect the interaction between different domains. Some of these sites are located in positions involved in conformational changes in envelope proteins. In summary, we summarize and discuss the key envelope protein residues that affect the entry process of flaviviruses, including the process of their discovery and the mechanisms that affect early infection.
... After independent subcultures in chick embryo tissue, it was generated the strains of 17DD (YF17DD) used as a vaccine in South America, and 17D-204, used in the rest of the world [3,4]. Genome sequencing studies revealed mutations in non-coding regions, and amino acid differences in structural and non-structural proteins, including non-conservative variations in E299, E305, E331 and E380 within ectodomain III of envelope protein, the cell binding region [5][6][7][8]. Further, it was shown that YF vaccines present much lower quasispecies diversity, when compared with virulent strains [9,10]. ...
Article
Full-text available
The yellow fever vaccine (YF17DD) is highly effective with a single injection conferring protection for at least 10 years. The YF17DD induces polyvalent responses, with a TH1/TH2 CD4+ profile, robust T CD8+ responses, and synthesis of interferon-gamma (IFN-γ), culminating in high titers of neutralizing antibodies. Furthermore, C-type lectin domain containing 5A (CLEC5A) has been implicated in innate outcomes in other flaviviral infections. Here, we conducted a follow-up study in volunteers immunized with YF17DD, investigating the humoral response, cellular phenotypes, gene expression, and single nucleotide polymorphisms (SNPs) of IFNG and CLEC5A, to clarify the role of these factors in early response after vaccination. Activation of CLEC5A+ monocytes occurred five days after vaccination (DAV). Following, seven DAV data showed activation of CD4+ and CD8+T cells together with early positive correlations between type II IFN and genes of innate antiviral response (STAT1, STAT2, IRF7, IRF9, OAS1, and RNASEL) as well as antibody levels. Furthermore, individuals with genotypes rs2430561 AT/AA, rs2069718 AG/AA (IFNG), and rs13237944 AC/AA (CLEC5A), exhibited higher expression of IFNG and CLEC5A, respectively. Together, we demonstrated that early IFN-γ and CLEC5A responses, associated with rs2430561, rs2069718, and rs13237944 genotypes, may be key mechanisms in the long-lasting immunity elicited by YF17DD.
... Whilst JEV-169 mAb is not known to recognize any previously reported epitopes, the loss of binding caused by the G184A mutation indicates the recognition of the antigenic structure between the G 0 and H 0 beta-strands of EDI. It is noteworthy that two neutralizing antibodies also recognize homologous regions in other flaviviruses including anti-dengue virus type 4 5H2 chimpanzee mAb and anti-yellow fever virus 117 mAb (Lai et al., 2007;Men et al., 2004;Ryman et al., 1998). The protective efficacy of JEV-169 mAb is in agreement with the immune protection mediated by the passive transfer of 5H2 mAb in the intracerebral challenge model using neonatal BALB/c mice (Lai et al., 2007). ...
Article
Full-text available
Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus endemic in the Asia Pacific region. Despite use of several highly effective vaccines, it is estimated that up to 44,000 new cases of Japanese encephalitis (JE) occur every year including 14,000 deaths and 24,000 survivors with permanent sequelae. Humoral immunity induced by vaccination is critical for effective protection. Potently neutralizing antibodies reactive with the JEV envelope (E) protein are important since protective immune responses induced by both live-attenuated and inactivated JE vaccines target the E protein. Our understanding of how vaccine-induced humoral immunity protects vaccinees from morbidity and mortality is, however, limited and largely obtained from in vitro studies. With the exception of neurovirulence mouse models, very few platforms are available for evaluating the protective efficacy of neutralizing antibodies against JEV in vivo. Swine are a major amplifying host in the natural JEV transmission cycle and develop multiple pathological outcomes similar to humans infected with JEV. In this study, prophylactic passive immunization was performed in a miniature swine model, using two vaccination-induced monoclonal antibodies (mAb), JEV-31 and JEV-169. These were selected as representatives for antibodies reactive with the major antigenic structures in the E protein of JEV and related flaviviruses. JEV-31 recognizes the lateral ridge of E protein domain III (EDIII) whilst JEV-169 has a broad footprint of binding involving residues throughout domains I (EDI) and II (EDII) of the E protein. Detection of neutralizing antibodies in the serum of immunized animals mimics the presence of neutralizing antibodies in vaccinated individuals. Passive immunization with both mAbs significantly reduced the severity of diseases that resemble the symptoms of human JE including fever, viremia, viral shedding, systemic infection, and neuroinvasion. In contrast to the uniformed decrease of viral loads in lymphoid and central nervous systems, distinct kinetics in the onset of fever and viremia between animals receiving JEV-31 and JEV-169 suggest potential differences in immune protection mechanisms between anti-EDI and anti-EDIII neutralizing antibodies elicited by vaccination. Our data demonstrate the feasibility of using swine models in characterizing the protective humoral immunity against JEV and increase our understanding of how clonal populations of anti-E mAbs derived from JE vaccination protect against infection in vivo.
... DII contains a highly conserved 16 amino acid fusion peptide motif (DRGWGNGCGLFGKGSI) (Figure 2, highlighted pink) [50] and can be found in a parallel orientation to the virion's lipid envelope in the pre-fusion state (See Figure 2A, lower panel). DIII harbors epitopes essential for neutralization of YFV [51,52]. However, antibody repertoire cloning from YFV patients also identified key neutralizing epitope residues in domains I (residue 71) and II (residues , which likely combine to form a conformational epitope [53]. ...
Article
Full-text available
Yellow fever virus (YFV) represents a re-emerging zoonotic pathogen, transmitted by mosquito vectors to humans from primate reservoirs. Sporadic outbreaks of YFV occur in endemic tropical regions, causing a viral hemorrhagic fever (VHF) associated with high mortality rates. Despite a highly effective vaccine, no antiviral treatments currently exist. Therefore, YFV represents a neglected tropical disease and is chronically understudied, with many aspects of YFV biology incompletely defined including host range, host-virus interactions and correlates of host immunity and pathogenicity. In this article, we review the current state of YFV research, focusing on the viral lifecycle, host responses to infection, species tropism and the success and associated limitations of the YFV-17D vaccine. In addition, we highlight the current lack of available treatments and use publicly available sequence and structural data to assess global patterns of YFV sequence diversity and identify potential drug targets. Finally, we discuss how technological advances, including real-time epidemiological monitoring of outbreaks using next-generation sequencing and CRISPR/Cas9 modification of vector species, could be utilized in future battles against this re-emerging pathogen which continues to cause devastating disease.
... This switch in entry mechanism relies essentially on the 12 mutations differentiating YFV 17D E protein from that of its parental (Asibi) strain [23]. Other alterations of YFV E protein have been reported to impact viral tropism and virulence [23,27,35,[168][169][170][171]. ...
Article
Full-text available
As revealed by the recent resurgence of yellow fever virus (YFV) activity in the tropical regions of Africa and South America, YFV control measures need urgent rethinking. Over the last decade, most reported outbreaks occurred in, or eventually reached, areas with low vaccination coverage but that are suitable for virus transmission, with an unprecedented risk of expansion to densely populated territories in Africa, South America and Asia. As reflected in the World Health Organization’s initiative launched in 2017, it is high time to strengthen epidemiological surveillance to monitor accurately viral dissemination, and redefine vaccination recommendation areas. Vector-control and immunisation measures need to be adapted and vaccine manufacturing must be reconciled with an increasing demand. We will have to face more yellow fever (YF) cases in the upcoming years. Hence, improving disease management through the development of efficient treatments will prove most beneficial. Undoubtedly, these developments will require in-depth descriptions of YFV biology at molecular, physiological and ecological levels. This second section of a two-part review describes the current state of knowledge and gaps regarding the molecular biology of YFV, along with an overview of the tools that can be used to manage the disease at the individual, local and global levels.
... The E protein is involved in receptor binding and membrane fusion [11][12][13] . Nonsynonymous (NS) mutations in this protein have been reported to impact viral tropism and virulence [14][15][16][17][18] . ...
Article
Full-text available
Yellow fever virus (Flavivirus genus) is an arthropod-borne pathogen, which can infect humans, causing a severe viscerotropic disease with a high mortality rate. Adapted viral strains allow the reproduction of yellow fever disease in hamsters with features similar to the human disease. Here, we used the Infectious Subgenomic Amplicons reverse genetics method to produce an equivalent to the hamster-virulent strain, Yellow Fever Ap7, by introducing a set of four synonymous and six nonsynonymous mutations into a single subgenomic amplicon, derived from the sequence of the Asibi strain. The resulting strain, Yellow Fever Ap7M, induced a disease similar to that described for Ap7 in terms of symptoms, weight evolution, viral loads in the liver and lethality. Using the same methodology, we produced mutant strains derived from either Ap7M or Asibi viruses and investigated the role of each of Ap7M nonsynonymous mutations in its in vivo phenotype. This allowed identifying key components of the virulence mechanism in hamsters. In Ap7M virus, the reversion of either E/Q27H or E/D155A mutations led to an important reduction of both virulence and in vivo replicative fitness. In addition, the introduction of the single D155A Ap7M mutation within the E protein of the Asibi virus was sufficient to drastically modify its phenotype in hamsters toward both a greater replication efficiency and virulence. Finally, inspection of the Asibi strain E protein structure combined to in vivo testing revealed the importance of an exposed α-helix in domain I, containing residues 154 and 155, for Ap7M virulence in hamsters.
... Domain III of the flavivirus E protein has been suggested to have an important role in binding to cell surface receptors. Mutations in EDIII have also been associated with attenuation of virulence in dengue [54] as well as in other flaviviruses, such as yellow fever virus and tickborne encephalitis [55,56], suggesting a fitness disadvantage for evolution or escape in this region. Additional factors could determine the impact of an escape mutation detected from in vitro studies or infection in vivo. ...
Article
Full-text available
Dengue virus (DENV) infection imposes enormous health and economic burden worldwide with no approved treatment. Several small molecules, including lovastatin, celgosivir, balapiravir and chloroquine have been tested for potential anti-dengue activity in clinical trials; none of these have demonstrated a protective effect. Recently, based on identification and characterization of cross-serotype neutralizing antibodies, there is increasing attention on the potential for dengue immunotherapy. Here, we tested the ability of VIS513, an engineered cross-neutralizing humanized antibody targeting the DENV E protein domain III, to overcome antibody-enhanced infection and high but brief viremia, which are commonly encountered in dengue patients, in various in vitro and in vivo models. We observed that VIS513 efficiently neutralizes DENV at clinically relevant viral loads or in the presence of enhancing levels of DENV immune sera. Single therapeutic administration of VIS513 in mouse models of primary infection or lethal secondary antibody-enhanced infection, reduces DENV titers and protects from lethal infection. Finally, VIS513 administration does not readily lead to resistance, either in cell culture systems or in animal models of dengue infection. The findings suggest that rapid viral reduction during acute DENV infection with a monoclonal antibody is feasible.
... Ab is able to distinguish between small differences in the primary amino acid sequence of proteins, in addition to differences in charge, optical configuration and steric conformation. The single amino acid mutations in the protein, although fairly distant from the contact residues, appeared to induce subtle conformational changes leading to the changes of the interaction with antibody 27,28 . Therefore, it is proposed that few different amino acids of MreB1/4 at S. eriocheiris and S. mirum alter the partial conformation of the proteins, thereby forming common and specific motifs of MreB1/4 between S. eriocheiris and S. mirum. ...
Article
Full-text available
A new species of spiroplasma, Spiroplasma eriocheiris (S. eriocheiris), was identified as a lethal pathogen of tremor disease (TD) in Chinese mitten crab recently. In order to acquire appropriate biological and diagnostic tools for characterizing this newly discovered pathogen, 5 monoclonal antibodies (mAbs) and a polyclonal antibody (pAb) against S. eriocheiris were produced. Among the mAbs, 6F5, 7C8 and 12H5 lead to the deformation of S. eriocheiris. A peptide sequence, YMRDMQSGLPRY was identified as a mimic motif of MreB that is the cell shape determining protein of S. eriocheiris interacting with 3 mAbs. Furthermore, a double antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) for detection of S. eriocheiris was established using the mAb and pAb we prepared. It detected as low as 0.1 mu g/mL of S. eriocheiris. No cross-reaction was observed with three other common bacteria (Pseudomonas aeruginosa, Escherichia coli, and Bacillus subtilis) and the hemolymph samples of healthy Eriocheir sinensis. Collectively, our results indicated that the mAbs and pAb we prepared could be used in the analysis of S. eriocheiris membrane proteins mimotope and development of a diagnostic kit for S. eriocheiris infections.
... The emergence of neutralization-resistant variants has been reported in vitro for YFV (e.g. Daffis et al., 2005;Lobigs et al., 1987;Ryman et al., 1998) and in animal models for other flaviviruses (Lai et al., 2007;Zhang et al., 2009) suggesting that use of MAbs in therapeutic applications will most probably require the preparation of multivalent antibody pools. A single study using purified neutralizing mouse antibodies for therapy of a human YFV infection late in the disease course reported no significant effects (Colebunders et al., 2002). ...
... Interestingly, E-240 has already been shown to play a role in YFV neurovirulence in mice (Ryman et al., 1998). This aa residue has also been shown to be located within a T-helper cell epitope for Murray Valley encephalitis virus (Mathews et al., 1991). ...
Article
Full-text available
Yellow fever virus (YFV) is a mosquito-transmitted, enveloped, positive stranded RNA virus belonging to the genus flavivirus, which causes hemorrhagic fever in humans in Africa and South America. The YFV is responsible for 200 000 clinical infections per year including 40 000 deaths. Despite the presence of a highly effective YF vaccine called 17D vaccine, this disease is now strongly re-emerging and has to be considered as a public health problem. The present live attenuated 17D vaccine has two major drawbacks: 1) the ancient production method by inoculating viable embryonated eggs which limits the vaccine production capacity and, therefore, impairs attempts to control the disease and may contribute to vaccine supply shortage. 2) this vaccine is a non clonal vaccine which is constituted of heterogenous virion sub-populations. Furthermore, recent reports of several cases of viscerotropic and neurotropic disease associated with 17D vaccination have raised the obvious question of vaccine safety. Taken together, these data show that it appears essential to design a new clonal vaccine which could be based on infectious cDNA clone and produced in animal cell culture. For this purpose, the knowledge of YFV neutralizing epitopes is essential. Because YFV immunity is mainly antibody-mediated, we wanted to isolate human neutralizing antibodies specific for YFV and use them as a tool to characterize the neutralizing epitopes of YFV. The phage display technology provides one of the most convenient systems to isolate such neutralizing recombinant antibody fragments. We generated YF patient-derived antibody phage libraries which were screened against purified virions of the YFV-204-WHO vaccine strain. This step led to the isolation of several single-chain antibody fragments (scFv) which recognized conformational and pH sensitive epitopes in the envelope E protein. Three genetically closely-related and competing scFvs were found to be able to neutralize in vitro the 17D vaccine strain and five wild-type African strains of YFV. To map their epitopes, neutralization escape variants of the YFV-17D-204-WHO were generated using one high-affinity scFv (scFv-7A). Amino acids (aa) E-153, E-154 and E-155 in domain I and aa E-71 in domain II of the E protein were shown to be the critical components of one complex neutralizing epitope. These aa do not form a contiguous epitope on the monomeric E protein, but are in close vicinity in the dimeric form the E protein is predicted to adopt, based on the crystal structures of related flaviviruses. The neutralizing epitope is thus predicted to be formed by contribution of aa from domain I and II of opposing E monomers. The nature of this epitope was supported by the analysis of one wild-type YFV strain (Senegal 90) which is naturally resistant to neutralization by scFv-7A. Microneutralization assays using sera from YFV-infected patients and 17D-immunized travelers confirm the importance of E-71 in YFV neutralization but also showed that those escape variants, originally present in the vaccine lot, do not carry a risk of neutralization escape in persons who are immunized with the 17D vaccine. The potential neutralization mechanism by which these scFvs act, particularly by preventing the fusion process, and their potential use as a therapeutical tool are discussed. The structural complexity of the epitope identified in this work has implications for understanding the mechanism of antibody-mediated neutralization of YFV and these data may be useful for the design of a new recombinant yellow fever vaccine based on a cDNA-derived infectious clone.
... Aside from dengue type 2 virus, for which a requirement for heparan sulfate binding has been demonstrated (2), no specific flavivirus cellular receptors have been definitively identified, but some reports indicate the presence of various cell surface proteins with specific binding affinities for different flaviviruses (10,11,19,27). An involvement of the lateral surface of domain III in cell attachment is suggested by several lines of indirect evidence (12,22), including the immunoglobulin-like fold of this domain, which is characteristically found in many proteins with specific binding functions, the high density of charged surface residues on the lateral surface, the presence of an RGD motif in some mosquito-borne flaviviruses (which is known in other cases to be recognized by members of the integrin protein familiy), and the identification of mutations in this region in host range mutants (14) and mutants with altered virulence properties (1,3,4,8,9,13,26). In this study we introduced mutations at four positions (residues 308 to 311) of the upper-lateral surface of domain III of the TBE virus protein E and investigated their influence on biological properties of the resulting mutant viruses. ...
Article
Full-text available
The impact of a specific region of the envelope protein E of tick-borne encephalitis (TBE) virus on the biology of this virus was investigated by a site-directed mutagenesis approach. The four amino acid residues that were analyzed in detail (E308 to E311) are located on the upper-lateral surface of domain III according to the X-ray structure of the TBE virus protein E and are part of an area that is considered to be a potential receptor binding determinant of flaviviruses. Mutants containing single amino acid substitutions, as well as combinations of mutations, were constructed and analyzed for their virulence in mice, growth properties in cultured cells, and genetic stability. The most significant attenuation in mice was achieved by mutagenesis of threonine 310. Combining this mutation with deletion mutations in the 3'-noncoding region yielded mutants that were highly attenuated. The biological effects of mutation Thr 310 to Lys, however, could be reversed to a large degree by a mutation at a neighboring position (Lys 311 to Glu) that arose spontaneously during infection of a mouse. Mutagenesis of the other positions provided evidence for the functional importance of residue 308 (Asp) and its charge interaction with residue 311 (Lys), whereas residue 309 could be altered or even deleted without any notable consequences. Deletion of residue 309 was accompanied by a spontaneous second-site mutation (Phe to Tyr) at position 332, which in the three-dimensional structure of protein E is spatially close to residue 309. The information obtained in this study is relevant for the development of specific attenuated flavivirus strains that may serve as future live vaccines.
... Residue E 315 lies along the distal surface of domain III, a region which has been previously implicated in the process of virion attachment to host cells (44). In YFV and JEV, mutations in the vicinity of E 315 are associated with altered virus tropism and changes in virulence (22,23,39,49). Residue E 439 lies within a predicted alpha-helical segment of the stem-anchor region whose structural integrity is required for stability of the prM-E heterodimer, based on studies with TBEV (2). ...
Article
Full-text available
A yellow fever virus (YFV)/Japanese encephalitis virus (JEV) chimera in which the structural proteins prM and E of YFV 17D are replaced with those of the JEV SA14-14-2 vaccine strain is under evaluation as a candidate vaccine against Japanese encephalitis. The chimera (YFV/JEV SA14-14-2, or ChimeriVax-JE) is less neurovirulent than is YFV 17D vaccine in mouse and nonhuman primate models (F. Guirakhoo et al., Virology 257:363-372, 1999; T. P. Monath et al., Vaccine 17:1869-1882, 1999). Attenuation depends on the presence of the JEV SA14-14-2 E protein, as shown by the high neurovirulence of an analogous YFV/JEV Nakayama chimera derived from the wild JEV Nakayama strain (T. J. Chambers, A. Nestorowicz, P. W. Mason, and C. M. Rice, J. Virol. 73:3095-3101, 1999). Ten amino acid differences exist between the E proteins of ChimeriVax-JE and the YFV/JEV Nakayama virus, four of which are predicted to be neurovirulence determinants based on various sequence comparisons. To identify residues that are involved in attenuation, a series of intratypic YFV/JEV chimeras containing either single or multiple amino acid substitutions were engineered and tested for mouse neurovirulence. Reversions in at least three distinct clusters were required to restore the neurovirulence typical of the YFV/JEV Nakayama virus. Different combinations of cluster-specific reversions could confer neurovirulence; however, residue 138 of the E protein (E(138)) exhibited a dominant effect. No single amino acid reversion produced a phenotype significantly different from that of the ChimeriVax-JE parent. Together with the known genetic stability of the virus during prolonged cell culture and mouse brain passage, these findings support the candidacy of this experimental vaccine as a novel live-attenuated viral vaccine against Japanese encephalitis.
... Additional mutagenesis targets could be considered from the outcome of other Flavivirus studies. Mutations in the vicinity of E315 are associated with altered virus tropism and changes in virulence [31][32][33] . Position E244 might not play a significant role as it is either a glycine (G) or glutamic acid (E) in several virulent JE strains analyzed 34,36 . ...
Article
By combining molecular-biological techniques with our increased understanding of the effect of gene sequence modification on viral function, yellow fever 17D, a positive-strand RNA virus vaccine, has been manipulated to induce a protective immune response against viruses of the same family (e.g. Japanese encephalitis and dengue viruses). Triggered by the emergence of West Nile virus infections in the New World afflicting humans, horses and birds, the success of this recombinant technology has prompted the rapid development of a live-virus attenuated candidate vaccine against West Nile virus.
... Collectively, data from many studies have indicated that amino acid substitutions in the E protein can influence virulence properties (25,31,40,41,49,50; reviewed in reference 38). The crystal structure of the soluble fragment of the tickborne encephalitis virus E protein is widely regarded as a model common to all flaviviruses (46). ...
Article
Full-text available
Serial passage of yellow fever 17D virus (YF5.2iv, derived from an infectious molecular clone) on mouse neuroblastoma (NB41A3) cells established a persistent noncytopathic infection associated with a variant virus. This virus (NB15a) was dramatically reduced in plaque formation and exhibited impaired replication kinetics on all cell lines examined compared to the parental virus. Nucleotide sequence analysis of NB15a revealed a substitution in domain III of the envelope (E) protein at residue 360, where an aspartic acid residue was replaced by glycine. Single mutations were also found within the NS2A and NS3 proteins. Engineering of YF5.2iv virus to contain the E(360) substitution yielded a virus (G360 mutant) whose plaque size and growth efficiency in cell culture resembled those of NB15a. Compared with YF5.2iv, both NB15a and G360 were markedly restricted for spread through Vero cell monolayers and mildly restricted in C6/36 cells. On NB41A3 cells, spread of the viruses was similar, but all three were generally inefficient compared with spread in other cell lines. Compared to YF5.2iv virus, NB15a was uniformly impaired in its ability to penetrate different cell lines, but a difference in cell surface binding was detected only on NB41A3 cells, where NB15a appeared less efficient. Despite its small plaque size, impaired growth, and decreased penetration efficiency, NB15a did not differ from YF5.2iv in mouse neurovirulence testing, based on mortality rates and average survival times after intracerebral inoculation of young adult mice. The data indicate that persistence of yellow fever virus in NB41A3 cells is associated with a mutation in the receptor binding domain of the E protein that impairs the virus entry process in cell culture. However, the phenotypic changes which occur in the virus as a result of the persistent infection in vitro do not correlate with attenuation during pathogenesis in the mouse central nervous system.
... In particular, structural domain III of E has been proposed elsewhere as a putative receptor-binding domain (13). Studies with several flaviviruses-including JE, yellow fever, dengue, and Murray Valley encephalitis viruses-have identified epitopes recognized by neutralizing antibodies within this domain (4,14,15,17). Although neutralizing epitopes have been identified in other domains of the E protein, antibodies binding to domain III are reported elsewhere to be the most efficient at blocking virus attachment to cells, supporting the proposed role of this domain in receptor binding (6). ...
Article
Full-text available
Using a panel of neutralizing monoclonal antibodies, we have mapped epitopes in domain III of the envelope protein of the New York strain of West Nile virus. The ability of monoclonal antibodies that recognize these epitopes to neutralize virus appeared to differ between lineage I and II West Nile virus strains, and epitopes were located on the upper surface of domain III at residues E307, E330, and E332.
... The E protein of tick-borne encephalitis has been crystallized; 118 domain III is believed to contain the putative-receptor binding domain 118 since most neutralizing mAbs bind to epitopes in this region. [119][120][121][122][123][124] In WNV infection, mutations that enable escape from neutralizing antibodies map to amino acid residues in domain III of the E protein. 119,125 Similarly, other flaviviruses (dengue, louping ill, yellow fever, and tick-borne encephalitis) efficiently escape antibody neutralization with mutations that map to domain III of the E protein. ...
Article
After a virus infects an animal, antiviral responses are generated that attempt to prevent dissemination. Interferons, antibody, complement, T and natural killer cells all contribute to the control and eradication of viral infections. Most flaviviruses, with the exception of some of the encephalitic viruses, cause acute disease and do not establish persistent infection. The outcome of flavivirus infection in an animal is determined by a balance between the speed of viral replication and spread, and the immune system response. Although many of the mechanistic details require further elucidation, flaviviruses have evolved specific tactics to evade the innate and adaptive immune response. A more thorough understanding of these principles could lead to improved models for viral pathogenesis and to strategies for the development of novel antiviral agents.
... A number of studies with monoclonal antibodies have identified E protein epitopes that are vaccine specific (17D-204 and 17DD substrains and FNV), wild-type specific, and 17D-204 substrain specific (Gould et al., 1985(Gould et al., , 1989Schlesinger et al., 1983;Sil et al., 2000). Ryman et al. (1997bRyman et al. ( , 1998) mapped one of the wild-type epitopes to E173 and a 17D-204 substrain-specific epitope to E305 and E325. Such epitopes have been predicted to be involved in the virulence phenotype of YF virus, but to date only the 17D-204 substrain-specific epitope has been found to alter the phenotype of the virus . ...
Article
Full-text available
It will be apparent to the reader that there is much to learn about the pathogenesis of YF. The role of specific genes and molecular determinants of neurotropism and viscerotropism has been defined only partially. The availability of infectious clones and a small animal (hamster) model should allow dissection of virulence factors, which can then be tested in the more difficult monkey model. The marked differences between wild-type YF strains should be evaluated by evaluating the relationships between virulence and genome sequence. The role of cytokine dysregulation and endothelial injury in YF will be elucidated as access to patients and of patients to more sophisticated medical care improves. The number of cases of YF in unvaccinated travelers hospitalized after return from the tropics has unfortunately increased, but such cases afford unique opportunities to study the pathogenesis of renal failure, coagulopathy, vascular instability, and shock, as well as new treatment modalities. At the cellular level, there are also important opportunities for research on YF virus-cell receptor interactions, the control of apoptotic cell death, and the predilection for cells of the midzone of the liver lobule. The role of dendritic cells in the early stage of YF infection is deserving of study. Finally, the role of the immune response to infection, particularly cellular immunity, is poorly characterized, and the suggestion that immune clearance may aggravate the condition of the host during the period of intoxication should be evaluated in appropriate animal models.
... Therefore full understanding of the molecular determinants for attenuation and virulence is of importance. Alan Barrett (University of Texas Medical Branch, Galveston, USA) showed that none of the 20 aa substitutions in YF-17D could be linked to neuro-and hepatotropism, however the E and NS4B protein may be important in determining the virus phenotype [18,19]. ...
... For example, a lysine to asparagine substitution in the YF 17D E protein was associated with fatal vaccine-associated encephalitis (Jennings et al., 1994). Residue 303 is adjacent to an antibody epitope which distinguishes wild type from vaccine strains of YFV and is associated with neutralization-escape mutations (phenylalanine 305 to valine or serine) (Ryman et al., 1998) that modulate neurovirulence properties conferred by a determinant in a distal region of domain II (residue 240). In addition, mutagenesis studies of the corresponding region of the TBE virus E protein demonstrated that lysine 311 (equivalent to YFV lysine 303 in the E protein alignment) modulates the effects of adjacent substitutions on virus growth efficiency, plaque size, and mouse neuroinvasiveness (Mandl et al., 2000). ...
Article
A yellow fever (YFV) 17D virus variant, which causes persistent infection of mouse neuroblastoma cells associated with defective cell penetration and small plaque size, yielded plaque-revertant viruses from cells transfected with viral transcripts encoding the adaptive mutation (Gly360 in the E protein). Reconstruction of a plaque-purified revertant which contained Gly360 and additional substitutions (Asn for Lys303 and Val for Ala261) yielded a virus whose infectious center size, growth efficiency, and cell penetration rate similar to the parental YF5.2iv virus, whereas viruses with Asn303 or Val261 alone with Gly360 yielded either a small-plaque virus or a parental revertant. These data indicate that the YFV E protein is subject to suppression of mutations in domain III that are deleterious for viral entry and spread by a second-site mutation in domain II. Position 261 lies within the hydrophobic ligand-binding pocket at the domain I-II interface, a site believed to be involved in the hinge-like conformational change of domain II during activation of membrane fusion-activity. Results of this study provide genetic data consistent with findings on flavivirus structure and implicate domain III in functions beyond simply cell surface attachment.
Article
Yellow fever virus (YFV) is the prototype member of the genus Flavivirus, which contains more than 60 positive-sense, single-stranded RNA viruses, many of which are considered public health threats. YF disease is controlled by a live attenuated vaccine, 17D, which was generated empirically through serial passage of the wild-type (WT) strain Asibi in chicken tissue. The vaccine, which has been used for over 80 years, is considered to be one of the safest and most effective live attenuated vaccines. It has been shown that the humoral immune response is essential to a positive disease outcome during infection. As such, the neutralizing antibody response and its correlation to long-term protection are a critical measure of 17D efficacy. The primary target of these antibodies is the envelope (E) protein, which is the major component of the virion. Monoclonal antibodies can distinguish WT strain Asibi and vaccine strain 17D by many different measures, including physical binding, hemagglutination inhibition, neutralization, and passive protection. This makes the WT-vaccine pair ideal candidates to study the structure-function relationship of the E protein in the attenuation and immunogenicity of flaviviruses. In this study, we provide an overview of structure-function of YFV E protein and its involvement in protective immunity.
Preprint
Full-text available
As revealed by the recent resurgence of yellow fever virus (YFV) activity in the tropical regions of Africa and South America, YFV control measures need urgent rethinking. Over the last decade, most reported outbreaks occurred in, or eventually reached, areas of low vaccination coverage but suitable for virus transmission, with an unprecedented risk of expansion to densely populated territories in Africa, South America and Asia. As reflected in the World Health Organization’s initiative launched in 2017, it is high time to strengthen epidemiological surveillance to monitor accurately, viral dissemination and redefine vaccination recommendation areas. Vector-control and immunisation measures need to be adapted and vaccine manufacturing must be reconciled with an increasing demand. We will have to face more YF cases in the upcoming years hence, improving disease management through the development of efficient treatments will prove most beneficial. Undoubtedly, these developments will require in-depth descriptions of YFV biology at molecular, physiological and ecological levels. This second section of the two-part review describes the current state of knowledge and gaps regarding the molecular biology of YFV, along with an overview of the tools that can be used to manage the disease at the individual, local and global levels.
Preprint
Full-text available
Yellow fever virus ( Flavivirus genus) is an arthropod-borne pathogen which can infect humans, causing a severe viscerotropic disease with a high mortality rate. Adapted viral strains allow the reproduction of yellow fever disease in hamsters with features similar to the human disease. Here, we used the Infectious Subgenomic Amplicons reverse genetics method to produce an equivalent to the hamster-virulent strain, Yellow Fever Ap7 , by introducing a set of 4 synonymous and 6 non-synonymous mutations into a single subgenomic amplicon, derived from the sequence of the Asibi strain. The resulting strain, Yellow Fever Ap7M , induced a disease similar to that described for Ap7 in terms of symptoms, weight evolution, viral loads in the liver and lethality. Using the same methodology, we produced mutant strains derived from either Ap7M or Asibi viruses and investigated the role of each of Ap7M non-synonymous mutations in its in vivo phenotype. This allowed identifying key components of the virulence mechanism in hamsters. In Ap7M virus, the reversion of either E/Q27H or E/D155A mutations, led to an important reduction of both virulence and in vivo replicative fitness. In addition, the introduction of the single D155A Ap7M mutation within the E protein of the Asibi virus was sufficient to drastically modify its phenotype in hamsters towards both a greater replication efficiency and virulence. Finally, inspection of the Asibi strain E protein structure combined to in vivo testing revealed the importance of an exposed α-helix in domain I, containing residues 154 and 155, for Ap7M virulence in hamsters.
Article
The worldwide use of yellow fever (YF) live attenuated vaccines came recently under close scrutiny as rare but serious adverse events have been reported. The population identified at major risk for these safety issues were extreme ages and immunocompromised subjects. Study NCT01426243 conducted by the French National Agency for AIDS research is an ongoing interventional study to evaluate the safety of the vaccine and the specific immune responses in HIV-infected patients following 17D-204 vaccination. As a preliminary study, we characterized the molecular diversity from E gene of the single 17D-204 vaccine batch used in this clinical study. Eight vials of lyophilized 17D-204 vaccine (Stamaril(®), Sanofi-Pasteur, Lyon, France) of the E lot 5499 batch were reconstituted for viral quantification, cloning and sequencing of C/prM/E region. The average rate of virions per vial was 8.68±0.07log10 genome equivalents with a low coefficient of variation (0.81%). 246 sequences of the C/prM/E region (29-33 per vials) were generated and analyzed for the eight vials, 25 (10%) being defective and excluded from analyses. 95% of sequences had at least one nucleotide mutation. The mutations were observed on 662 variant sites distributed through all over the 1995 nucleotides sequence and were mainly non-synonymous (66%). Genome variability between vaccine vials was highly homogeneous with a nucleotide distance ranging from 0.29% to 0.41%. Average p-distances observed for each vial were also homogeneous, ranging from 0.0015 to 0.0031. This study showed a homogenous YF virus RNA quantity in vaccine vials within a single lot and a low clonal diversity inter and intra vaccine vials. These results are consistent with a recent study showing that the main mechanism of attenuation resulted in the loss of diversity in the YF virus quasi-species. Copyright © 2015. Published by Elsevier Ltd.
Article
Full-text available
The Colombian 17D vaccine contains at least four phenotypes denominated small (0.3 - 1.2 mm), medium (1.3 - 2.1 mm) large (2.2 - 3.0 mm) and extra-large (>3.1 mm). These phenotypes composition and percentage distribution vary between different production lots and between ampoules from a particular lot. Each variant was cloned by diluting the vaccine and its virulent effect was analysed in young mice. The small plaque phenotype was slightly under represented in the lots analysed and showed similar virulence to the neurotropic French wild strain (LD50 >10-6), whilst the medium- sized phe-notype predominated and was the most attenuated (LD50:10-4). The large and extra-large phenotypes showed intermediate virulence (LD50:10-5) regarding the others. Sequence analysis of variants within the region between the 3'NS5 end and the beginning of 3'NCR showed the closeness between those variants having some degree of virulence and between the attenuated variant and the Colombian vaccine. The 17D vaccine's heterogeneity constitutes evidence of RNA virus quasi-specie structure and shows how adverse yellow fever vaccine reaction can be associated with applying vaccines produced from attenuated viral strains.
Chapter
Flaviviruses are complex immunogens that elicit antibodies of varying specificity and with a spectrum of functional properties. Flavivirus virions are covered by a dense array of envelope (E) proteins that mediate steps of the virus entry pathway and are a primary target of neutralizing antibodies. The development of virus-specific antibodies is a critical aspect of protection against flavivirus infection and a major goal of ongoing efforts to produce vaccines against flaviviruses of clinical importance, such as the West Nile virus. In this chapter, we will review current models that describe how antibodies engage flaviviruses and block infection. Recent insight into the relationships that govern where antibodies bind virions and how this impacts the potency and mechanisms of neutralization of antibodies have been driven in part by insights from the structural biology of flaviviruses. The factors that define antibody potency will be discussed with a focus on the stoichiometric requirements for neutralization.
Article
This study of the yellow fever French neurotropic vaccine strain from the Institut Pasteur (FNV-IP) demonstrates that this viral genome is not as stable as that of the 17D-204 vaccine virus. FNV-IP was plaque-purified three times and then passaged eight times in Vero cells. Viral populations from the second and eighth passage post purification were sequenced and compared to the published sequences of FNV-IP. The passage-2 viral population had 31 nucleotide and nine amino acid changes compared to the parental virus while the passage-8 virus had six additional nucleotide changes encoding a single amino acid substitution. The plaque-purified virus also had two sequence deletions in the 3′-noncoding region. The plaque purification resulted in selection of a passage-2 virus that had a mouse LD50 of 20 pfu/ml, 67-fold greater than parental FNV-IP which had an LD50 of 0.3 pfu/ml. Subsequent passage in Vero cells resulted in a passage-8 virus which had increased neurovirulence with an LD50 of 3.2 pfu/ml. The only amino acid difference between the passage-2 and passage-8 viruses was at amino acid 638 of NS5 which lies within domain V of the RNA-dependent-RNA polymerase. Overall, these data indicate that FNV-IP virus has an inherently less stable genome than 17D vaccine virus and a variable viral population.
Article
Full-text available
Many RNA viruses encode error-prone polymerases which introduce mutations into B and T cell epitopes, providing a mechanism for immunological escape. When regions of hypervariability are found within immunodominant epitopes with no known function, they are referred to as "decoy epitopes," which often deceptively imprint the host's immune response. In this work, a decoy epitope was identified in the foot-and-mouth disease virus (FMDV) serotype O VP1 G-H loop after multiple sequence alignment of 118 isolates. A series of chimeric cyclic peptides resembling the type O G-H loop were prepared, each bearing a defined "B cell xenoepitope" from another virus in place of the native decoy epitope. These sequences were derived from porcine respiratory and reproductive syndrome virus (PRRSV), from HIV, or from a presumptively tolerogenic sequence from murine albumin and were subsequently used as immunogens in BALB/c mice. Cross-reactive antibody responses against all peptides were compared to a wild-type peptide and ovalbumin (OVA). A broadened antibody response was generated in animals inoculated with the PRRSV chimeric peptide, in which virus binding of serum antibodies was also observed. A B cell epitope mapping experiment did not reveal recognition of any contiguous linear epitopes, raising the possibility that the refocused response was directed to a conformational epitope. Taken together, these results indicate that xenoepitope substitution is a novel method for immune refocusing against decoy epitopes of RNA viruses such as FMDV as part of the rational design of next-generation vaccines.
Article
Dengue virus (DENV) is the causative agent of dengue fever (DF), the most prevalent arthropod-borne viral disease in the world and therefore is considered an emerging global health threat. The four DENV serotypes (DENV-1, DENV-2, DENV-3 and DENV-4) that infect humans are distinguished from one another by unique antigenic determinants (epitopes) on the DENV envelope (E) protein. The E protein is the primary antigenic site of the DENV and is responsible for inducing neutralising antibody (Ab) and cell mediated immune response in DENV infected hosts. The DENV E protein also mediates attachment of virions to host cell receptors and entry of virions into host cells by membrane fusion. The study of epitopes on DENV E protein is necessary for understanding viral function and for the design of unique polyvalent vaccines capable of inducing a neutralising antibody response against each DENV serotype. Reverse genetics using infectious cDNA clones has enabled the construction of functional intertypic DENV, where the E protein of one DENV serotype is put in the genetic background of a different DENV serotype. In addition, observations from our laboratory indicate that chimeric E proteins, consisting of E protein structural domains from different DENV serotypes can fold into functional proteins. This suggests that there is potential to engineer viruses with intertypic DENV E proteins as potential DENV vaccine candidates, which is the long term goal of studies within our research group. However, if a chimeric E protein was to be constructed containing epitopes involved in antibody mediated neutralisation of each DENV serotype, then knowledge of the location of these epitopes on the E protein of each DENV serotype would be essential. Prior to this study, monoclonal antibodies (MAbs) had been used to identify epitopes involved in antibody mediated neutralisation on the E protein of all DENV serotypes, except DENV-4. The primary objective of this study was to identify epitopes on the DENV-4 E protein involved in neutralisation by antibodies. In order to achieve this objective, a panel of 14 MAbs was generated against DENV-4 in BALB/c mice and characterised using various serological and functional assays. The identification of DENV-4 specific neutralising MAbs in the panel was essential for subsequent experiments aimed at determining antigenic domains, structural domains or specific epitopes (peptides or amino acids) involved in the neutralisation of DENV-4. The majority of MAbs (11/14) generated against DENV-4 recognised the E protein. The remaining three MAbs reacted with the non-structural (NS) 1 protein. The majority of MAbs against the E protein were DENV or Flavivirus group reactive, but four MAbs were DENV-4 specific. All MAbs against the E protein recognised conformationally dependent epitopes and were able to capture DENV-4 in an enzyme linked immuno-adsorbent assay (ELISA). Eighty percent (9/11) of the anti-E MAbs produced for this study neutralised infection of cells by DENV-4 in vitro. Three of the neutralising MAbs (F1G2, 18F5 and 13H8) were DENV-4 specific and also demonstrated the strongest neutralisation activity of the panel, reducing DENV-4 infectivity by 100-1000 fold. The amount of virus neutralised by the MAbs was not related to the avidity of the MAbs. The DENV-4 specific MAbs F1G2, 18F5 and 13H8 were used to identify epitopes involved in neutralisation of DENV-4. The MAbs that effectively captured DENV-4 were used in competitive binding assays (CBAs) to determine spatial relationships between epitopes and therefore define antigenic domains on the DENV-4 E protein. The CBAs indicated that the epitopes recognised by the panel of MAbs segregated into two distinct domains (D4E1 and D4E2) and both contained epitopes involved in neutralisation. CBAs incorporating human serum from DENV-4 infected patients suggested that the MAbs recognised the same, or spatially related, epitopes in domain D4E2 as antibodies from humans who had experienced natural dengue infections, indicating the clinical relevance of such epitopes for the development of DENV vaccines. The reactivity of the capture MAbs with low pH treated DENV-4 was also evaluated in an attempt to identify epitopes that might be more accessible during low pH-mediated virus fusion. Only one of the MAbs (13H8) recognised an acid resistant epitope. Initial attempts to identify epitopes on the DENV-4 E protein involved in neutralisation followed the traditional epitope mapping approach of selecting subpopulations of DENV-4 which escaped neutralisation by MAbs. These attempts were unsuccessful so a variety of strategies for mapping epitopes were used including DENV-4 variant analysis and site directed mutagenesis of the DENV-4 E protein, MAb screening of chimeric DENV-3/4 E proteins and MAb screening of a bacterial peptide display library. DENV-4 variants including DENV-4 isolates from different geographical locations or chemically mutagenised DENV-4 were screened with neutralising MAbs to identify neutralisation escape mutant (n.e.m.) viruses. Site directed mutagenesis of the DENV-4 E protein confirmed whether amino acid changes identified in DENV-4 n.e.m.s were essential for the binding of neutralising MAbs to an epitope. The MAb screening of DENV-4 variants identified n.e.m.s with amino acid changes at residues E95, E96, E156, E157, E203, E329 and E402 of the DENV-4 E protein. Site directed mutagenesis of the DENV-4 E protein identified two epitopes recognised by the DENV-4 specific neutralising MAbs F1G2 and 18F5 at specific amino acid residues within domains II and III of the DENV-4 E protein. No specific epitopes were identified for the MAb 13H8; however this MAb did recognise domain I and II of the DENV-4 E protein, when screened against DENV-3/4 chimeric DENV E proteins. The first epitope, which was recognised by the MAb F1G2, contained residue E95 which was located in domain II of the DENV-4 E protein. The aspartate (Asp) to alanine (Ala) change at E95 prevented the binding of F1G2 to the DENV-4 E protein. The binding of F1G2 to the E95 residue was confirmed using the pFlitrX bacterial peptide display library, which demonstrated binding of F1G2 to a peptide homologous with residues E99-E104. No peptides recognised by 13H8 and 18F5 were identified by this method. The MAb F1G2 also bound to the domain III region (E300-E495) of the DENV-4 E protein when screened against DENV-3/4 chimeric DENV E proteins. This implied that F1G2 may be recognising a discontinuous epitope consisting of domains II and III. The second epitope, which was recognised by MAb 18F5, contained residue E329 which was located in domain III of the DENV-4 E protein. The alanine (Ala) to threonine (Thr) change at E329 prevented the binding of 18F5 to the DENV-4 E protein. MAb 18F5 also bound to the domain III region (E300-E495) of the DENV-4 E protein when screened against DENV-3/4 chimeric E proteins, thus confirming the E329 epitope. The potential mechanisms by which the DENV-4 specific MAbs neutralise virus infection were evaluated by the virus overlay protein binding assay (VOPBA). The binding of MAb 18F5 to a domain III (E329) epitope of the DENV-4 E protein and the binding of MAb F1G2 to domain II (E95, E99-E104) and domain III epitopes (chimeric E protein) of the DENV-4 E protein, prevented the attachment of DENV-4 to a 40 kDa C6/36 cell protein. In contrast the binding of MAb 13H8 to domains I and II of the DENV-4 E protein did not prevent attachment of DENV-4 to the same protein. This was preliminary evidence that the binding of domain III epitopes by the MAbs F1G2 and 18F5 may be important in preventing virus attachment. The binding of MAb 13H8 to domains I and II, and the ability of this MAb to recognise DENV-4 treated at low pH, suggested that MAb 13H8 may block epitopes exposed at low pH that are required for low pH mediated virus fusion to host cell membranes. Overall, the different methods used in this study identified epitopes involved in the neutralisation of DENV-4. The distribution of epitopes involved in neutralisation throughout the DENV-4 E protein were similar to the distribution of epitopes involved in neutralisation on the DENV-1, 2 and 3 E proteins. This suggested that it might be possible to elicit neutralising antibodies against multiple DENV serotypes using chimeric E-proteins derived from two or more DENV serotypes and therefore, facilitate the design of novel tetravalent DENV vaccines.
Article
The structure of recombinant domain III of the envelope protein (rED3) of yellow fever virus (YFV), containing the major neutralization site, was determined using NMR spectroscopy. The amino acid sequence and structure of the YFV-rED3 shows differences from ED3s of other mosquito-borne flaviviruses; in particular, the partially surface-exposed BC loop where methionine-304 and valine-324 were identified as being critical for the structure of the loop. Variations in the structure and surface chemistry of ED3 between flaviviruses affect neutralization sites and may affect host cell receptor interactions and play a role in the observed variations in viral pathogenesis and tissue tropism.
Article
In 1998, the US Centers for Disease Control and Prevention was notified of three patients who developed severe illnesses days after yellow fever vaccination. A similar case occurred in 1996. All four patients were more than 63 years old. Vaccine strains of yellow fever virus, isolated from the plasma of two patients and the cerebrospinal fluid of one, were characterised by genomic sequencing. Clinical samples were subjected to neutralisation assays, and an immunohistochemical analysis was done on one sample of liver obtained at biopsy. The clinical presentations were characterised by fever, myalgia, headache, and confusion, followed by severe multisystemic illnesses. Three patients died. Vaccine-related variants of yellow fever virus were found in plasma and cerebrospinal fluid of one vaccinee. The convalescent serum samples of two vaccinees showed antibody responses of at least 1:10240. Immunohistochemical assay of liver tissue showed yellow fever antigen in the Kuppfer cells of the liver sample. The clinical features, their temporal association with vaccination, recovery of vaccine-related virus, antibody responses, and immunohistochemical assay collectively suggest a possible causal relation between the illnesses and yellow fever vaccination. Yellow fever remains an important cause of illness and death in South America and Africa; hence, vaccination should be maintained until the frequency of these events is quantified.
Article
Full-text available
A hamster viscerotropic strain of yellow fever (YF) virus has been derived after serial passage of strain Asibi through hamsters. The parental Asibi/hamster p0 virus causes a mild and transient viremia in hamsters with no outward, clinical signs of illness. In contrast, the viscerotropic Asibi/hamster p7 virus causes a robust viremia, severe illness, and death in subadult hamsters. The genome of the hamster viscerotropic Asibi/hamster p7 virus has been sequenced and compared with the parental nonviscerotropic Asibi/hamster p0 virus identifying 14 nucleotide changes encoding only seven amino acid substitutions. The majority of these substitutions (five of seven) fall within the envelope (E) protein at positions Q27H, D28G, D155A, K323R, and K331R. These results support an important role for the E protein in determining YF virus viscerotropism.
Article
Full-text available
Dengue virus (DV) is a flavivirus and infects mammalian cells through mosquito vectors. This study investigates the roles of domain III of DV type 2 envelope protein (EIII) in DV binding to the host cell. Recombinant EIII interferes with DV infection to BHK21 and C6/36 cells by blocking dengue virion adsorption to these cells. Inhibition of EIII on BHK21 cells was broad with no serotype specificity; however, inhibition of EIII on C6/36 cells was relatively serotype specific. Soluble heparin completely blocks binding of EIII to BHK21 cells, suggesting that domain III binds mainly to cell surface heparan sulfates. This suggestion is supported by the observation that EIII binds very weakly to gro2C and sog9 mutant mammalian cell lines that lack heparan sulfate. In contrast, heparin does not block binding of EIII to mosquito cells. Furthermore, a synthetic peptide that includes amino acids (aa) 380 to 389 of EIII, IGVEPGQLKL, inhibits binding of EIII to C6/36 but not BHK21 cells. This peptide corresponds to a lateral loop region on domain III of E protein, indicating a possible role of this loop in binding to mosquito cells. In summary, these results suggest that EIII plays an important role in binding of DV type 2 to host cells. In addition, EIII interacts with heparan sulfates when binding to BHK21 cells, and a loop region containing aa 380 to 389 of EIII may participate in DV type 2 binding to C6/36 cells.
Article
Full-text available
A chimeric flavivirus infectious cDNA was constructed by exchanging the premembrane (prM) and envelope (E) genes of the yellow fever virus vaccine strain 17D (YF17D) with the corresponding genes of Modoc virus (MOD). This latter virus belongs to the cluster of the "not-known vector" flaviviruses and is, unlike YF17D, neuroinvasive in SCID mice. Replication of in vitro-transcribed RNA from this chimeric flavivirus was shown by [(3)H]uridine labeling and RNA analysis. Expression of the MOD prM and E proteins was monitored by radioimmunoprecipitation and revealed that the MOD proteins were correctly and efficiently produced from the chimeric precursor protein. The MOD E protein was shown to be N-linked glycosylated, whereas prM, as predicted from the genome sequence, did not contain N-linked carbohydrates. In Vero cells, the chimeric virus replicated with a similar efficiency as the parental viruses, although it formed smaller plaques than YF17D and MOD. In SCID mice that had been infected intraperitoneally with the chimeric virus, the viral load increased steadily until death. The MOD/YF virus, like MOD from which it had acquired the prM and E structural proteins, but unlike YF, proved neuroinvasive in SCID mice. Animals developed neurological symptoms about 15 days after inoculation and died shortly thereafter. The distribution of MOD/YF RNA in the brain of infected mice was similar to that observed in MOD-infected mice. The observations provide compelling evidence that the determinants of neuroinvasiveness of flaviviruses are entirely located in the envelope proteins prM and E.
Article
Yellow fever, a mosquito-borne viral hemorrhagic fever, is one of the most lethal diseases of humankind. The etiologic agent is the prototype member of the genus Flavivirus, family Flaviviridae, a group of small, enveloped, positive-sense, single-strand RNA viruses. Approximately one in seven people who become infected develop a rapidly progressive illness, with hepatitis, renal failure, hemorrhage and cardiovascular shock, with a case fatality rate of 20-50%. Yellow fever occurs in sub-Saharan Africa and tropical South America, where it remains a continuing public health problem of varying magnitude, depending on the level of vaccination coverage in the human population and cyclical, ecologic and climatic factors that influence virus transmission.
Article
The live attenuated Yellow Fever Vaccine 17D [YF-17D] is one of the most effective vaccines available. During the 70 years since its development, the vaccine has been administered to more than 400 million people worldwide with minimal incident of severe side effects. Despite its efficacy, the immunological mechanisms that mediate its efficacy are poorly understood. Here we review the development of YF-17D in a historical context, and then present some emerging evidence which suggests that YF-17D activates multiple Toll-like receptors (TLRs) on dendritic cells (DCs) to elicit a broad spectrum of innate and adaptive immune responses. Interestingly, the resulting adaptive immune responses are characterized by a mixed T helper cell (Th)1/Th2 cytokine profile and antigenspecific CD8(+) T cells, and distinct TLRs appear to differentially control the Th1/Th2 balance. These data offer some new insights into the molecular mechanism of action of YF-17D, and highlight the potential of vaccination strategies that use combinations of different TLR ligands to stimulate polyvalent immune responses.
Article
Full-text available
We have compared the deduced envelope (E) protein sequences of two biologically well-characterized yellow fever (YF) virus vaccine strains. The 17DD strain has been produced in Brazil for more than 50 years and used to successfully vaccinate millions of people worldwide. The 17D-213 is a candidate vaccine strain produced in tissue culture which has previously passed the monkey neurovirulence assay for testing human YF vaccines. Nucleotide sequence analysis of polymerase chain reaction-amplified cDNA revealed a number of mutations which were strain- and substrain-specific. A major difference of 17DD and 17D-213 as compared to 17D-204 and Asibi was the existence of a potential N-linked glycosylation site located at amino acid residues 153 and 151 of 17DD and 17D-213, respectively. These acceptor sites are apparently utilized for the addition of high-mannose carbohydrate chains as shown by endoglycosidase analyses of immunoprecipitated E proteins. Glycosylated E protein is also used to assemble YF vaccine virions. This work and eventual complete nucleotide sequence analysis of both vaccine strains should help to define possible changes involved in YF virus attenuation and allow their biological importance to be determined using a recently developed system for generating YF virus from cDNA. In addition, these data provide an estimate on the extent of genetic variability among YF 17D seeds and vaccines.
Article
Full-text available
Monoclonal antibodies prepared against vaccine strains of yellow fever (YF) virus were initially characterized by fluorescence microscopy of Vero cells infected with YF virus strain 17D. When similarly tested against representatives of all flavivirus subgroups, the antibodies produced a wide spectrum of reactions ranging from the monospecific to the broadly cross-reactive; at least five antigenic domains in the YF virus envelope glycoprotein were identified. Monoclonal antibodies differentiated between YF virus vaccine strains (17D, 17DD, FNV), wild-type viruses and plaque variants selected from a 17D pool. One isolate from a patient with YF was antigenically similar to the Brazilian vaccine strain 17DD. Several of the antibodies reacting with the YF viral envelope glycoprotein in biological tests identified the 54K envelope glycoprotein; 45K and 26K polypeptides in YF 17D virus-infected cells were also identified by radioimmunoprecipitation and polyacrylamide gel electrophoresis. Neither of these polypeptides was found in uninfected cells. They may represent short-lived precursors of the 54K protein, post-translational cleavage or breakdown products. Other antibodies reacted with a 48K polypeptide in virus-infected cell lysates. This may be the non-structural NV3 protein described for YF virus. Its appearance on the surface of unfixed infected cells, but not on released virions, was demonstrated by fluorescence microscopy.
Article
Full-text available
Monoclonal antibodies (MAbs) against the Asibi wild-type strain of yellow fever (YF) virus were prepared and characterized. One of the MAbs (designated MAb 117) was shown, by cross-immunofluorescence tests with flaviviruses, to be specific for wildtype YF virus. This MAb was used in indirect immunofluorescence tests to identify wild-type antigenic variants in several different YF vaccine pools. Simultaneously, a vaccine-specific MAb prepared previously (MAb 864) was used to identify YF strain 17D vaccine type variants in the wild-type Asibi virus preparation. One variant, isolated by plaque purification from a 17D vaccine pool, possessed the wild-type epitope and was neurovirulent in infant mice whereas other variants, lacking the wild-type epitope but with vaccine-specific epitopes (identified by MAb 411), were avirulent in infant mice. Avirulent variants were able to infect mice and induce antibody. Virus-specific antigen was still detected in the brains of these mice 4 weeks after inoculation, suggesting that persistent infections were developing. These results to the potential risk of selection of wild-type variants in YF vaccine preparations. They also point to the potential risk of selection of wild-type variants in YF vaccine preparations and re-emphasize the need for modernization of techniques and more effective control measures to be taken during the production of YF vaccine.
Article
Full-text available
Monoclonal and polyclonal antibodies with known specificity for either the 54K envelope glycoprotein or the 48K non-structural glycoprotein of yellow fever (YF) virus-infected cells were studied in plaque reduction neutralization tests. Viruses employed in the tests comprised wild-type and vaccine strains of YF and a selection of other flaviviruses. Of 17 monoclonal antibodies examined, six of the 54K-specific antibodies neutralized at least one YF preparation. Both vaccine and wild-type YF viruses varied in their susceptibility to neutralization and there were also differences between individual 17D vaccine strains. The monoclonal antibodies produced a range of titres with the different viruses, the most potent, 864, leaving no non-neutralizable fraction. Addition of anti-globulin, complement or other antibodies did not affect the results. YF-neutralize antibodies which bound to other flaviviruses did not necessarily neutralize them; hence, neutralization could be defined as either homotypic, heterotypic or both homotypic and heterotypic. A polyclonal antiserum and a broadly reacting monoclonal antibody produced almost identical neutralization results in tests with wild-type YF viruses. In contrast, the polyclonal antiserum produced higher titres with vaccine strains of YF. In mouse passive protection experiments on the other hand, the monoclonal antibody did not differentiate between these viruses.
Article
Full-text available
The capacity of monoclonal antibodies to protect mice passively against yellow fever (YF) virus infection was investigated. Both neutralizing (54K-specific) and non-neutralizing (54K- and 48K-specific) antibodies protected mice against challenge with the RMP substrain of YF virus. Average survival times of mice inoculated intracerebrally with a standard lethal dose of YF virus differed according to the strain used: thus mice inoculated with the most neurovirulent viruses, FNV and Asibi, survived for 6 X 50 and 7 X 65 days respectively, and those with RMP virus survived for 15 X 75 days. The capacity of antibodies to protect mice passively against virus challenge was directly related to virus neurovirulence. Possible mechanisms and the significance of protection by antibodies against non-structural proteins that do not mediate neutralization, are discussed.
Article
Full-text available
Antigenic variants of CVS-11 strain of rabies virus were selected after treatment of virus populations with monoclonal antibodies directed against the glycoprotein antigen of the virus. These variants resisted neutralization by the hybridoma antibody used for their selection. Two independently mutating antigenic sites could be distinguished when five variants were tested with nine hybridoma antibodies. The frequency of single epitope variants in a cloned rabies virus seed was approximately 1:10,000. Animals were not or only partially protected when challenged with the parent virus or with another variant, but were fully protected against challenge with the virus used for immunization. Variants were also detected among seven street viruses obtained from fatal cases of human rabies. Animals immunized with standard rabies vaccine were protected against challenge with some but not all street rabies variants. A comparative antigenic analysis between vaccine strain and challenge virus by means of monoclonal antiglycoprotein antibodies showed a slightly closer degree of antigenic relatedness between vaccine and challenge strain in the combinations where vaccination resulted in protection. It remains unknown, however, whether these apparently minor antigenic differences in the glycoproteins account for the varying degrees of protection. The results of this study clearly indicate that the selection of vaccine strains and the methods used to evaluate the potency of rabies vaccines need to be revised.
Article
Full-text available
Two neutralization-resistant variants of dengue virus type 2 were selected using the neutralizing monoclonal antibody G8D11. Virus N-GV4 was derived from the New Guinea C strain and virus P-GV3 from the PUO-218 strain. Both variants had an identical change at nucleotide 919 in the E gene, causing a substitution of glutamic acid for lysine at residue 307 in the E glycoprotein. The substitution abolished the ability of antibody G8D11 to bind to the E glycoprotein in radioimmunoprecipitation experiments. The epitope was sensitive to treatment with SDS and was dependent on the formation of a disulfide bridge. This dependency was determined by mutagenesis of Cys residues 11 and 12 in the E glycoprotein.
Article
Full-text available
Of four wild-type strains (Nakayama-original, SA14, 826309 and Beijing-1) of Japanese encephalitis (JE) virus that were passaged six times in HeLa cells (HeLa p6), two (Nakayama-original and 826309) became attenuated for mice. In the case of strain Nakayama-original, the virulence for mice was markedly reduced and attenuation was retained on passage in primary chicken embryo fibroblast, LLC-MK2 and C6/36 cells. The binding of non-HeLa-passaged Nakayama virus to mouse brain membrane receptor preparations could be differentiated from binding by Nakayama HeLa p6 virus, suggesting that the envelope (E) protein is involved in the attenuated phenotype. Both of the attenuated viruses can be distinguished from the virulent non-HeLa-passaged parental viruses by examination with E protein reactive vaccine and wild-type-specific monoclonal antibodies (MAbs). The vaccine-specific MAb V23, which is only reactive with the SA14 series of live vaccine viruses, recognized the HeLa cell-attenuated Nakayama-original and 826309 viruses, whereas two wild-type-specific MAbs (MAbs K13 and K39) lost reactivity. Comparison of the nucleotide sequences of the structural protein genes of the 826309 and Nakayama-original virulent parent and attenuated HeLa p6 viruses revealed that the viruses differed by 37 and 46 nucleotides coding for eight and nine amino acid mutations, respectively. However, other than one amino acid in the E protein, the membrane and E protein amino acid sequences of the two attenuated HeLa p6 viruses were identical.
Article
Full-text available
To identify the molecular determinants for attenuation of wild-type Japanese encephalitis (JE) virus strain SA14, the RNA genome of wild-type strain SA14 and its attenuated vaccine virus SA14-2-8 were reverse transcribed, amplified by PCR and sequenced. Comparison of the nucleotide sequence of SA14-2-8 vaccine virus with virulent parent SA14 virus and with two other attenuated vaccine viruses derived from SA14 virus (SA14-14-2/PHK and SA14-14-2/PDK) revealed only seven amino acids in the virulent parent SA14 had been substituted in all three attenuated vaccines. Four were in the envelope (E) protein (E-138, E-176, E-315 and E-439), one in non-structural protein 2B (NS2B-63), one in NS3 (NS3-105), and one in NS4B (NS4B-106). The substitutions at E-315 and E-439 arose due to correction of the SA14/CDC sequence published previously by Nitayaphan et al. (Virology 177, 541-552, 1990). The mutations in NS2B and NS3 are in functional domains of the trypsin-like serine protease. Attenuation of SA14 virus may therefore, in part, be due to alterations in viral protease activity, which could affect replication of the virus.
Article
Full-text available
Seven mutant viruses were derived from a Scottish strain of louping ill virus using a virus envelope-specific neutralizing monoclonal antibody. None of the mutants was neutralized and immunofluorescence microscopy confirmed that they did not bind to this antibody. Four mutants showed reduced mouse neurovirulence compared with parent virus and two mutants failed to induce protective immune responses in mice challenged with virulent tick-borne encephalitis virus. The mutants with the lowest virulence showed poor or undetectable haemagglutinating activity. The nucleotide sequence of the envelope glycoprotein gene of each of the seven mutants was determined and the deduced amino acid sequence was compared with parent virus. For each mutant, only a single amino acid codon change was detected and all the amino acid substitutions occurred within amino acid positions 308 to 311. A change from the amino acid aspartate to asparagine at amino acid position 308, which represented a potential glycosylation site, was the most effective substitution in reducing mouse neurovirulence. The results demonstrate the importance of critical sites within the envelope glycoprotein as determinants of virus virulence.
Article
Full-text available
Two monoclonal antibody neutralization resistant (MAbR) variants of the yellow fever (YF) 17D-204 vaccine virus strain were selected using YF type-specific MAb B39. These B39R variants were compared with the variant virus selected by Lobigs et al. (Virology 161, 474-478, 1987) using a second YF-type specific MAb (2E10) which mapped to amino acid position 71/72 in the envelope (E) protein. Neutralization assays with a panel of MAbs suggested that these two YF-type-specific epitopes are located in two discrete regions of the folded E protein. Each of the B39R variants had a single nucleotide mutation which encoded an amino acid substitution at either position E-155 or E-158. Thus, YF type-specific epitopes map to both domain I (B39) and II (2E10) of the YF virus E protein. The B39 defined epitope represents the first flavivirus neutralizing epitope localized to this region of domain I of the E protein.
Article
The Sarawak strain of Japanese encephalitis virus (1E-Sar) is virulent in 3-week-old mice when inoculated intraperitoneally. The nucleotide sequence for the envelope glycoprotein (E) of this virus was determined and compared with the published sequences of four other strains. There were several silent nucleotide differences and five codon changes. Monoclonal antibodies (MAbs) against the E protein of JE-Sar virus were prepared and characterized. MAb-resistant mutants of 1E-Sar were selected to determine if mutations in the E protein gene could affect its virulence for mice. Eight mutants were isolated using five different MAbs that identified virus-specific or group-reactive epitopes on the E protein. The mutants lost either complete or partial reactivity with selecting MAb. Several showed decreased virulence in 3-week-old mice after intraperitoneal inoculation. Two (r27 and r30) also showed reduced virulence in 2-week-old mice. JE-Sar and the derived mutants were comparable in their virulence for mice, when inoculated intracranially. Mutant r30 but not r27 induced protective immunity in adult mice against intracranial challenge with parent virus. However, r27-2 did induce protective immunity against itself. Nucleotide sequencing of the E coding region for the mutants revealed single base changes in both r30 and r27 resulting in a predicted change from isoleucine to serine at position 270 in r30 and from glycine to aspartic acid at position 333 in r27. The altered capacity of the mutants to induce protective immunity is consistent with the immunogenicity changes predicted by computer analysis using the Protean II program.
Article
To study the role of the precursor to the membrane protein (prM) in flavivirus maturation, we inhibited the proteolytic processing of the Murray Valley encephalitis (MVE) virus prM to membrane protein in infected cells by adding the acidotropic agent ammonium chloride late in the virus replication cycle. Viruses purified from supernatants of ammonium chloride-treated cells contained prM protein and were unable to fuse C6/36 mosquito cells from without. When ammonium chloride was removed from the cells, both the processing of prM and the fusion activity of the purified viruses were partially restored. By using monoclonal antibodies (MAbs) specific for the envelope (E) glycoprotein of MVE virus, we found that at least three epitopes were less accessible to their corresponding antibodies in the prM-containing MVE virus particles. Amino-terminal sequencing of proteolytic fragments of the E protein which were reactive with sequence-specific peptide antisera or MAb enabled us to estimate the site of the E protein interacting with the prM to be within amino acids 200 to 327. Since prM-containing viruses were up to 400-fold more resistant to a low pH environment, we conclude that the E-prM interaction might be necessary to protect the E protein from irreversible conformational changes caused by maturation into the acidic vesicles of the exocytic pathway.
Article
The nucleotide sequences of the envelope protein of the Kamiyama 1 strain of Japanese encephalitis (JE) virus and a passaged mutant (Kamiyama 2 strain) were determined. Two amino acid differences, Ser-Phe at residue 364 and Asn-Ile at residue 367, distinguished Kamiyama 2 from Kamiyama 1. Six neutralization-resistant variants were selected from these two strains using a JE species-specific monoclonal antibody with neutralization and hemagglutination-inhibition reactivities. All variants had a single amino acid substitution at residue 52 and significantly reduced reactivity with other JE species-specific monoclonal antibodies. The variants derived from Kamiyama 2 strain showed reduced virulence in 3-week-old mice after peripheral inoculation but were as virulent as the parent virus when inoculated intracranially. These variants also showed altered early virus-cell interaction but not replication and reproduction in Vero cells. These findings indicate that the mutations at residues 52, 364, and 367 affect early virus-cell interaction in Vero cells and virulence in mice.
Article
The polymerase chain reaction (PCR) was used to amplify viral cDNAs from selected regions of dengue genomic RNA by using appropriate 'consensus' primers. DNA amplicons containing the structural genes from all 4 dengue serotypes were prepared and directly sequenced using dengue-virus-specific primers. This method can characterize reliably flavivirus field isolates at the molecular level without extensive virus propagation and molecular cloning, and will be a valuable tool for molecular epidemiological studies.
Article
The 15 amino acids which precede the sequence of the envelope (E) protein in the yellow fever virus (YFV) polyprotein precursor have been proposed to function as a signal peptide for the E protein (P. Desprès A. Cahour, C. Wychowski, M. Girard and M. Bouloy; Ann. Inst. Pasteur/Virol., 139, 59-67, 1988). To confirm this hypothesis, recombinant SV40 genomes were constructed in which the sequence of the E protein, or that of the poliovirus VP0 capsid polypeptide were placed immediately downstream of and in frame with the sequence of the putative signal peptide, under the control of the late SV40 promoter. The E protein expressed by the hybrid virus SV-E was recognized by two neutralizing monoclonal antibodies directed against the YFV envelope protein. In this construct, the E protein was deleted of its C-terminal transmembrane zone. Therefore, as expected, the protein appeared to be efficiently transported along the exocytic pathway and excreted into the cell culture medium. In addition, when the putative signal peptide was fused in frame with poliovirus polypeptide VP0, the expressed chimeric polypeptide was targeted to the endoplasmic reticulum where it underwent glycosylation.
Article
We have determined the virulence characteristics of seven monoclonal antibody escape mutants of tick-borne encephalitis virus in the mouse model. One of the mutants with an amino acid substitution from tyrosine to histidine at residue 384 revealed strongly reduced pathogenicity after peripheral inoculation of adult mice but retained its capacity to replicate in the mice and to induce a high-titered antibody response. Infection with the attenuated mutant resulted in resistance to challenge with virulent virus. Assessment of nonconservative amino acid substitutions in other attenuated flaviviruses suggests that a structural element including residue 384 may represent an important determinant of flavivirus virulence in general.
Article
Monoclonal antibodies (MCA), with defined molecular specificity, were used in a competition binding enzyme-linked immunosorbent assay (ELISA) to locate the relative positions of the epitopes on the envelope glycoprotein of yellow fever 17D vaccine virus and its wild type parent virus, Asibi (AS). Five topographically distinct antigenic domains were defined on the E glycoprotein of the 17D vaccine. Three of these (A, B, and C) were represented by one MCA each, a fourth (D) was represented by two MCA, and a fifth domain (E) comprised a major cluster of at least five overlapping epitopes. Asibi virus also possessed domain E which is proposed to be a conserved antigenic region within the envelope glycoprotein of all flaviviruses. Domains A and C were not represented on Asibi virus and one epitope, situated proximal to the E domain, showed structural alterations in physical overlap. Functional activities were assigned to physically mapped epitopes by haemagglutination inhibition (HAI), virus neutralisation (N), and passive protection in mice. The HAI and N functions were not necessarily linked but only MCA with N activity were able to protect mice passively against lethal infection. All domains demonstrated a heterogeneous range of biological properties dependent upon the virus strain rather than the epitope.
Article
The location of a major antigenic determinant involved in the neutralization of a flavivirus, yellow fever virus (YF), has been defined in terms of its position in the amino acid sequence of the E protein. Neutralization escape variants of the 17D vaccine strain of YF were selected with two neutralizing monoclonal antibodies. Nucleotide sequencing of the envelope protein genes (E and M) of the variants showed that in each variant there was a single nucleotide change in the E gene leading to a nonconservative amino acid substitution in the E protein at position 71 or 72. The changes are in a region of the E protein which is hydrophilic, rich in cysteine residues, and not conserved between flavivirus subgroups. Since the selecting monoclonal antibodies neutralize attenuated 17D and virulent Asibi strains of YF with equal efficiency (J. J. Schlesinger, M. W. Brandriss, and T. P. Monath, 1983, Virology 125, 8-17), it can be concluded that the neutralization determinant defined for 17D YF is also present in Asibi YF.
Article
A model of the tick-borne encephalitis virus envelope protein E is presented that contains information on the structural organization of this flavivirus protein and correlates epitopes and antigenic domains to defined sequence elements. It thus reveals details of the structural and functional characteristics of the corresponding protein domains. The localization of three antigenic domains (composed of 16 distinct epitopes) within the primary structure was performed by (i) amino-terminal sequencing of three immunoreactive fragments of protein E and (ii) sequencing the protein E-coding regions of seven antigenic variants of tick-borne encephalitis virus that had been selected in the presence of neutralizing monoclonal antibodies directed against the E protein. Further information about variable and conserved regions was obtained by a comparative computer analysis of flavivirus E protein amino acid sequences. The search for potential T-cell determinants revealed at least one sequence compatible with an amphipathic alpha-helix which is conserved in all flaviviruses sequenced so far. By combining these data with those on the location of disulfide bridges (T. Nowak and G. Wengler, Virology 156:127-137, 1987) and the structural characteristics of epitopes, such as dependency on conformation or on intact disulfide bridges or both, a model was established that goes beyond the location of epitopes in the primary sequence and reveals features of the folding of the polypeptide chain, including the generation of discontinuous protein domains.
Article
Two panels of envelope glycoprotein reactive monoclonal antibodies (mAbs) were prepared against yellow fever (YF) 17D vaccine viruses. Five mAbs were prepared against the World Health Organization 17D-204 avian leukosis virus-free secondary seed virus and eight mAbs against 17DD vaccine manufactured in Brazil. The majority of these mAbs were type-specific and displayed differing reactions in neutralization tests. One, B14, would only neutralize YF vaccine virus grown in invertebrate cells. Others would differentiate 17D-204 and 17DD vaccines, from different manufacturers, in neutralization tests when the viruses were grown in vertebrate cells. The data indicate that heterogeneity exists between the epitopes that elicit neutralizing antibody on YF vaccine from different manufacturers.
Article
We have sequenced the virulent Asibi strain of yellow fever virus and compared this sequence to that of the 17D vaccine strain, which was derived from it. These two strains of viruses differ by more than 240 passages. We found that the two RNAs, 10,862 nucleotides long, differ at 68 nucleotide positions; these changes result in 32 amino acid differences. Overall, this corresponds to 0.63% nucleotide sequence divergence, and the changes are scattered throughout the genome. The overall divergence at the level of amino acid substitution is 0.94%, but these changes are not randomly distributed among the virus protein. The capsid protein is unchanged, while proteins NS1, NS3, and NS5 contain 0.5% amino acid substitutions, and proteins ns4a and ns4b average 0.8% substitutions. In contrast, proteins ns2a and ns2b have 3.0 and 2.3% amino acid divergence, respectively. The envelope protein also has a relatively high rate of amino acid change of 2.4% (a total of 12 amino acid substitutions). The large number of changes in ns2a and ns2b, which are largely conservative in nature, may result from lowered selective pressure against alteration in this region; among flaviviruses, these polypeptides are much less highly conserved than NS1, NS3, and NS5. However, many of the amino acid substitutions in the E protein are not conservative. It seems likely that at least some of the difference in virulence between the two strains of yellow fever virus results from changes in the envelope protein that affect virus binding to host receptors. Such differences in receptor binding could result in the reduced neurotropism and vicerotropism exhibited by the vaccine strain.
Article
Neutralization escape variants of Murray Valley encephalitis virus were selected using a type-specific, neutralizing, and passively protective anti-envelope protein (E) monoclonal antibody (4B6C-2) which defines epitope E-1c. Nucleotide sequence analysis revealed single nucleotide changes in the E genes of 15 variants resulting in nonconservative amino acid substitutions in all cases. One variant had a three-nucleotide deletion in the E gene which resulted in loss of serine at residue 277. Changes were clustered into two separate regions of the E polypeptide (residues 126-128 and 274-277), indicating that E-1c is a discontinuous epitope. One variant (BHv1), altered at residue 277 (Ser-->Ile), failed to hemagglutinate across the pH range 5.5-7.5, in contrast to parental virus and the other escape variants which hemagglutinated at an optimal pH of 6.6. BHv1 was also of reduced neuroinvasiveness in 21-day-old mice following intraperitoneal inoculation compared to the other viruses. Parental virus and the neutralization escape variants grew equally well in both vertebrate and invertebrate cell cultures, indicating that the reduced neuroinvasiveness of BHv1 was not due to a major abnormality of replication.
Article
The crystallographically determined structure of a soluble fragment from the major envelope protein of a flavivirus reveals an unusual architecture. The flat, elongated dimer extends in a direction that would be parallel to the viral membrane. Residues that influence binding of monoclonal antibodies lie on the outward-facing surface of the protein. The clustering of mutations that affect virulence in various flaviviruses indicates a possible receptor binding site and, together with other mutational and biochemical data, suggests a picture for the fusion-activating, conformational change triggered by low pH.
Article
The nucleic acid and deduced amino acid sequences of the structural proteins of the 17DD vaccine strain of yellow fever (YF) virus originating from Senegal are compared with those published for other vaccine strains of YF virus. Even though the 17D-204 and 17DD substrains of 17D diverged at passage 195 of 17D, they show a very high degree of nucleotide (99.5%) and amino acid (99.5%) homology over this region. The sequences are discussed with respect to monoclonal antibodies that can distinguish between 17DD and 17D-204 substrains of YF vaccines.
Article
The live-attenuated yellow fever (YF) vaccine virus, strain 17D-204, has long been known to consist of a heterologous population of virions. Gould et al. (J. Gen. Virol. 70, 1889-1894 (1989)) previously demonstrated that variant viruses exhibiting a YF wild-type-specific envelope (E) protein epitope are present at low frequency in the vaccine pool and were able to isolate representative virus variants with and without this epitope, designated 17D(+wt) and 17D(-wt), respectively. These variants were employed here in an investigation of YF virus pathogenesis in the mouse model. Both the 17D-204 parent and the 17D(+wt) variant viruses were lethal for adult outbred mice by the intracerebral route of inoculation. However, the 17D(-wt) variant was significantly attenuated (18% mortality rate) and replicated to much lower titer in the brains of infected mice. A single amino acid substitution in the envelope (E) protein at E-240 (Ala-->Val) was identified as responsible for the restricted replication of the 17D(-wt) variant in vivo. The 17D(+wt) variant has an additional second-site mutation, believed to encode a reversion to the neurovirulence phenotype of the 17D-204 parent virus. The amino acid substitution in the E protein at E-173 (Thr-->Ile) of the 17D(+wt) variant which results in the appearance of the wild-type-specific epitope or nucleotide changes in the 5' and 3' noncoding regions of the virus are proposed as a candidates.