Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2014 Nov 26;89(3):1838–1850. doi: 10.1128/JVI.02277-14

Nipah Virus Attachment Glycoprotein Stalk C-Terminal Region Links Receptor Binding to Fusion Triggering

Qian Liu a, Birgit Bradel-Tretheway a, Abrrey I Monreal c, Jonel P Saludes c, Xiaonan Lu d, Anthony V Nicola a,b, Hector C Aguilar a,b,
Editor: D S Lyles
PMCID: PMC4300768  PMID: 25428863

ABSTRACT

Membrane fusion is essential for paramyxovirus entry into target cells and for the cell-cell fusion (syncytia) that results from many paramyxoviral infections. The concerted efforts of two membrane-integral viral proteins, the attachment (HN, H, or G) and fusion (F) glycoproteins, mediate membrane fusion. The emergent Nipah virus (NiV) is a highly pathogenic and deadly zoonotic paramyxovirus. We recently reported that upon cell receptor ephrinB2 or ephrinB3 binding, at least two conformational changes occur in the NiV-G head, followed by one in the NiV-G stalk, that subsequently result in F triggering and F execution of membrane fusion. However, the domains and residues in NiV-G that trigger F and the specific events that link receptor binding to F triggering are unknown. In the present study, we identified a NiV-G stalk C-terminal region (amino acids 159 to 163) that is important for multiple G functions, including G tetramerization, conformational integrity, G-F interactions, receptor-induced conformational changes in G, and F triggering. On the basis of these results, we propose that this NiV-G region serves as an important structural and functional linker between the NiV-G head and the rest of the stalk and is critical in propagating the F-triggering signal via specific conformational changes that open a concealed F-triggering domain(s) in the G stalk. These findings broaden our understanding of the mechanism(s) of receptor-induced paramyxovirus F triggering during viral entry and cell-cell fusion.

IMPORTANCE The emergent deadly viruses Nipah virus (NiV) and Hendra virus belong to the Henipavirus genus in the Paramyxoviridae family. NiV infections target endothelial cells and neurons and, in humans, result in 40 to 75% mortality rates. The broad tropism of the henipaviruses and the unavailability of therapeutics threaten the health of humans and livestock. Viral entry into host cells is the first step of henipavirus infections, which ultimately cause syncytium formation. After attaching to the host cell receptor, henipaviruses enter the target cell via direct viral-cell membrane fusion mediated by two membrane glycoproteins: the attachment protein (G) and the fusion protein (F). In this study, we identified and characterized a region in the NiV-G stalk C-terminal domain that links receptor binding to fusion triggering via several important glycoprotein functions. These findings advance our understanding of the membrane fusion-triggering mechanism(s) of the henipaviruses and the paramyxoviruses.

INTRODUCTION

Nipah virus (NiV) and Hendra virus (HeV) are two important emergent viruses in the genus Henipavirus (HNV) within the Paramyxoviridae family, which includes other important pathogens such as measles virus (MeV), mumps virus, Newcastle disease virus (NDV), human parainfluenza virus (PIV), and respiratory syncytial virus. NiV is an emergent zoonotic pathogen present at high frequencies in its natural reservoir, fruit bats, and can be transmitted from animal to animal, animal to human, and human to human (1). NiV exhibits a broad species tropism (including dogs, cats, pigs, horses, and humans). This is at least in part due to the widely distributed and highly conserved NiV cellular receptors ephrinB2 (B2) and ephrinB3 (B3) (24). NiV targets endothelial and neuronal cells in humans, causing acute encephalitis and pulmonary syndrome with high mortality rates and respiratory and neurological sequelae (5). NiV is classified as a biosafety level 4 (BSL4) pathogen because of its high pathogenicity and potential for bioterrorism (6).

Paramyxovirus attachment proteins are designated HN, H, or G, depending on their hemagglutinin (H) and/or neuraminidase (N) activities (7). The attachment proteins are type II transmembrane glycoproteins consisting of an N-terminal cytoplasmic tail, a transmembrane domain, an extracellular stalk, and a globular head. X-ray crystallography has revealed a conserved paramyxovirus HN-H-G receptor-binding globular head domain with a six-bladed β propeller structure (812). The NiV-G head domain has been crystallized in the presence or absence of cellular receptors, and the common six-bladed β propeller structure is shown with the center of the top face binding the ephrinB3 receptor (8, 10). Despite this commonality, differences among paramyxovirus attachment proteins exist. For example, the HN proteins of NDV and PIV5 bind and cleave sialic acid, while the H and G proteins of MeV and NiV bind protein receptors (7). Furthermore, sialic acid receptor binding appears to induce HN-F association, while protein receptor binding appears to induce G/H-F dissociation of a previously associated G/H-F complex (13, 14).

NiV-F is a class I fusion protein that has structural and functional features in common with fusion proteins of many families (e.g., HIV-1 gp41 or influenza virus hemagglutinin [HA]), such as an ectodomain with a hydrophobic fusion peptide and two heptad repeat regions (15). Upon triggering, F (prefusion conformation) undergoes conformational changes such that it inserts its fusion peptide into the target membrane to form a metastable prehairpin intermediate (PHI) (13, 16). Subsequently, two heptad repeat regions in the PHI fold together to form a postfusion six-helix bundle (16). The energy released by these conformational changes allows F to execute membrane fusion.

NiV enters host cells by fusion of the viral and host cell membranes at physiological pH without requiring viral endocytosis. Membrane fusion enables entry of the viral ribonucleoprotein complex into the host cell, followed by virus replication. Syncytium formation (cell-cell fusion) is a pathological hallmark of NiV infections, resulting in cell-to-cell spread, inflammation, and destruction of endothelial cells and neurons (17). Both NiV entry and syncytium formation require the concerted efforts of the attachment (G) and fusion (F) glycoproteins. Upon receptor binding, NiV-G triggers a conformational cascade in NiV-F that executes viral and/or cell membrane fusion.

Numerous studies suggest that paramyxovirus attachment protein stalk domains play a significant role in interacting with and triggering their cognate fusion proteins (1823). The recently obtained crystal structure of the PIV5- and NDV-HN stalk domain revealed a four-helix bundle (4HB) (11, 19, 24). Interestingly, amino acid residues in the NDV-HN stalk domain that interact with F appear to be largely obscured by the head, as suggested by mutagenesis analysis (11, 2123). Furthermore, it has been recently shown for several paramyxoviruses, including our report for NiV, that headless HN-H-G mutant proteins that carry an intact 4HB tetramerization domain are still able to trigger cell-cell fusion with their cognate F, suggesting that the stalk domain of the attachment protein has F-triggering features in common within the paramyxovirus family (2528). Although the crystal structures of the NiV- and HeV-G stalk domains are unsolved, several studies including site-directed mutagenesis, construction of NDV/NiV chimeras, and removal or addition of N-glycan moieties have suggested that NiV- and HeV-G stalk domains are important in modulating fusion and G oligomerization (18, 2831). We recently reported a three-step spatiotemporal mechanism of receptor-induced NiV membrane fusion. Receptor binding induces at least two sequential conformational changes in the G head, followed by the exposure of a G stalk domain, which interacts with and triggers F (28).

In the present study, we attempted to precisely identify residues in the NiV-G stalk responsible for F triggering and the events that occur in NiV-G that link receptor binding to F triggering. We identified a NiV-G stalk C-terminal region (amino acids [aa] 159 to 163) that is important for NiV-G oligomerization, conformational integrity, G-F interactions, receptor-induced conformational changes in G, and F triggering. This region was mapped to the membrane-distal portion of the stalk, close to the NiV-G head. Multidisciplinary approaches, including conformation-dependent antibody analysis, coimmunoprecipitation studies, and Raman spectroscopy, suggest that, upon receptor binding, this stalk region propagates the “F-triggering signal” from the head to the stalk.

MATERIALS AND METHODS

Cell culture.

PK13 and 293T cells were cultured in Dulbecco's modified Eagle medium (DMEM), and Vero cells were cultured in minimal essential medium, both supplemented with 10% fetal bovine serum (FBS), 50 IU of penicillin ml–1, 50 μg of streptomycin ml–1, and 2 mM glutamine.

Expression plasmids, antisera, and antibodies.

Expression plasmids for codon-optimized wild-type (WT) NiV-G and -F, untagged or tagged at the C terminus with the HA and AU1 tags, respectively, were previously described (32). The hyperfusogenic NiV-F N-glycan mutant FF3F5 and cytoplasmic tail mutant FK1A proteins were also previously described (32, 33). The NiV-G headless mutant proteins were constructed by inserting two stop codons (TAA TAA) at the corresponding sites. The NiV-F hyperfusogenic (FK1A F3F5) and hypofusogenic (FP221A) mutant proteins and NiV-G full-length point mutant proteins were constructed by site-directed mutagenesis (Agilent Technologies Inc.). All mutations were verified by DNA sequencing (Eurofins).

The production of rabbit polyclonal anti-G antisera (806 and Ab167) and conformational monoclonal antibodies Mab45 and Mab213 has been described previously (28, 33, 34). The soluble recombinant mouse ephrinB2 human Fc chimera (B2), containing ectodomain residues 27 to 227 of mouse ephrinB2, was purchased from R&D Systems. The rabbit anti-HA monoclonal antibody (Bethyl Laboratories Inc.) and mouse anti-AU1 monoclonal antibody (Covance) were used at 1:2,000 and 1:500 dilutions in immunoblot assays, respectively. Fluorescently labeled secondary antibody Alexa Fluor 647 goat anti-rabbit IgG (Life Technologies, NY) was used at a 1:200 dilution for flow cytometry. Alexa Fluor 647 goat anti-mouse IgG antibody and Alexa Fluor 488 goat ant-rabbit IgG antibodies were used at a 1:2,000 dilution for immunoblot assays.

Quantification of NiV-F and -G cell surface expression (CSE) and antibody and ephrinB2 binding by flow cytometry.

Two micrograms of WT or mutant NiV-G expression plasmid was transfected into 293T or PK13 cells (per well of a six-well plate, 70% confluence) to achieve a linear correlation between the CSE levels and the amount of DNA transfected (32). Cells were collected at 20 to 24 h posttransfection. To measure CSE and ephrinB2 (B2) binding levels, 293T cells expressing WT or mutant NiV-G protein were incubated with anti-NiV-G polyclonal antisera or soluble B2 (see above) at 4°C for 1 h, respectively. The cells were washed twice with phosphate-buffered saline (PBS) supplemented with 1% FBS and then incubated with a fluorescently labeled secondary antibody for 30 min at 4°C. To measure the binding levels of conformation-dependent antibodies, PK13 cells expressing WT or mutant NiV-G were incubated with soluble B2 for 15 min at 4°C and then subjected to a procedure identical to that described above. Cells were fixed with 0.5% paraformaldehyde, and the mean fluorescence intensity was measured by flow cytometry (Guava easyCyte 8HT; EMD Millipore). Cells transfected with the vector alone (pcDNA3.1) served as a negative control.

Quantification of syncytia and EphB3 inhibition.

To achieve a linear correlation between the amount of DNA transfected and the levels of syncytia, WT NiV-F and WT or mutant NiV-G expression plasmids were transfected into 293T or Vero cells (six-well plates, 70% confluence) at a 1:1 ratio (2 μg total). At 12 to 28 h posttransfection, syncytium formation was quantified by counting the nuclei inside syncytia per 200× field. A syncytium is defined as four or more nuclei within the same cell membrane. EphB3 is a natural ligand of ephrinB2 or ephrinB3 (35). For the EphB3 inhibition experiment, mouse EphB3-human Fc (R&D Systems) was added to the cells at 6 to 8 h posttransfection to achieve a final concentration of 40 nM, whereas PBS was added as a negative control (0 nM EphB3).

Quantification of viral genome copies and viral entry.

NiV-F and NiV-G were pseudotyped onto a vesicular stomatitis virus (VSV) reporter expressing the Renilla Luc reporter gene (NiV/VSV-rLuc) as previously described (32, 33). Briefly, 293T cells were transfected with the respective WT and mutant NiV-G and NiV-F expression plasmids and subsequently infected with recombinant VSV-ΔG-rLuc. NiV/VSV-rLuc pseudotyped virions were collected at ∼30 h postinfection and purified over a 20% sucrose cushion. Virions were resuspended in NaCl-Tris-EDTA (NTE) buffer supplemented with 5% sucrose and stored at −80°C. Viral RNA was extracted with the QIAamp viral RNA minikit (Qiagen), and the isolated RNA was reverse transcribed with the SuperScriptIII first-strand synthesis system for reverse transcription-PCR (Invitrogen). The VSV genome copy number was quantified by using a quantitative TaqMan PCR protocol as previously described (32).

To quantify viral entry, 293T cells were seeded into a 96-well plate and infected with 10-fold serial dilutions of NiV/VSV-rLuc pseudotyped virions in infection buffer (PBS plus 1% FBS) at 37°C. Complete growth medium (DMEM) was added to the cells at 2 h postinfection. 293T cells were lysed at 20 to 24 h postinfection, and luciferase activity was measured in relative light units (RLU) with a Renilla Luciferase Flash Assay kit (Pierce) and an Infinite M1000 microplate reader (Tecan Group Ltd.). RLU were plotted against numbers of genome copies per milliliter and regressed with GraphPad Prism 5 (GraphPad Software Inc.).

Detection of glycoprotein expression and viral incorporation by Western blotting.

293T cells transfected with WT or mutant NiV-G expression plasmids were lysed in 1× radioimmunoprecipitation assay (RIPA) buffer (EMD Millipore) supplemented with protease inhibitors (cOmplete, Mini; Roche) at 24 to 48 h posttransfection. Cell lysates or 109 VSV/NiV-rLuc pseudotyped virions (genome copies) were subjected to SDS-PAGE and immunoblotting. NiV-G proteins were detected with anti-G polyclonal antisera (described above). Fluorescently labeled secondary antibodies (described above) were used at a dilution of 1:2,000 and detected by a Chemidoc gel imager system (Bio-Rad).

Coimmunoprecipitation.

293T cells transfected with WT NiV-F and WT or mutant NiV-G (HA-tagged) expression plasmids were lysed in RIPA buffer at 24 to 36 h posttransfection. Cell lysates were incubated with rabbit anti-HA μMAC Microbeads (Miltenyi Biotec) for 1 h at 4°C with rotation and then purified and eluted over μMAC columns (Miltenyi Biotec). Total cell lysates and purified elutions were subjected to SDS-PAGE and immunoblotting as described above. Protein band intensities were measured by densitometry with a Chemidoc imaging system (Bio-Rad).

PNGase F treatment.

293T cells transfected with headless NiV-G expression plasmids were lysed in RIPA buffer at 20 to 24 h posttransfection. Cell lysates were denatured for 10 min at 60°C and treated with or without peptide-N-glycosidase F (PNGase F; New England BioLabs Inc.) for 1 h at 37°C according to the manufacturer's protocol. The PNGase F-treated cell lysates were subjected to 15% SDS-PAGE and immunoblotting with anti-AU1 monoclonal antibody against F and polyclonal antiserum Ab167 against G.

Raman spectroscopy.

We reported that the ephrinB2-containing VSV pseudotyped virions (VSV/B2) were able to induce membrane fusion on receptor-negative cells expressing NiV-G and -F (33). In this study, VSV/B2 virions were used in a cell-free system to initiate the conformational changes in NiV-G and -F on virions as previously described (36). Briefly, pseudotyped VSV containing WT or mutant NiV-G was incubated with VSV/B2 virions for 30 min on ice to allow receptor binding. The mixture was then transferred from the ice bath directly onto a glass microarray slide coated with gold (Thermo Scientific Inc.). The viral samples on the gold-coated microarray slide were immediately subjected to Raman spectral collection by using a confocal micro-Raman spectrometer above 25°C (Renishaw). The spectra of the virions containing only WT or mutant NiV-G were collected for spectral subtraction. The Raman spectroscopic settings and spectral processing were identical to those described in our recent publication (36).

Statistics.

P values were calculated by unpaired Student t test and corrected by using the respective Bonferroni correction factors. Average values ± the standard errors of the means (SEM) from at least three independent experiments are presented.

RESULTS

The C-terminal region of the NiV-G stalk (aa 159 to 167) is important for fusion triggering.

Our previous studies revealed that NiV-G headless mutant protein 167 (aa 1 to 167) can trigger NiV-F-mediated 293T cell-cell fusion to approximately WT (full-length) levels, as measured by counting relative amounts of syncytial nuclei (28). These results indicate that headless mutant G protein 167 contains the necessary and sufficient F-triggering domain(s). To pinpoint the region(s) in G important for F triggering, we made two more G headless truncation versions, mutant proteins 158 (aa 1 to 158) and 164 (aa 1 to 164), by inserting stop codons after their most C-terminal residues (Fig. 1A). Compared to mutant 167, which triggered 293T cell-cell fusion at levels similar to those of the WT, mutant 164 induced fusion at only ∼20% of WT G levels (Fig. 1B and C) and mutant protein 158 did not induce any 293T cell-cell fusion, although both were expressed to at least 50% of the WT NiV-G level at the cell surface (Fig. 1B and C). We used flow cytometry to determine the 293T CSE of WT and headless mutant NiV-G by using anti-G polyclonal antiserum Ab167, raised against headless mutant protein 167 (28). All of the mutant proteins were expressed on the cell surface to various degrees (Fig. 1B), resembling results obtained by Western blot analysis of total cell lysates with Ab167 (Fig. 1D).

FIG 1.

FIG 1

The C-terminal region of the NiV-G stalk (aa 159 to 167) is important for fusion triggering. (A) A schematic of the full-length WT NiV-G protein (aa 1 to 602) with its cytoplasmic tail (CT), transmembrane (TM), and extracellular (aa 71 to 602) domains is shown. The amino acid sequence of the stalk C-terminal region (Stalk C-term; residues 146 to 187) is shown together with the NiV-G truncation mutant proteins, which are named after their most C-terminal residues (158, 164, and 167). Cysteine residues important for NiV-G tetramerization are marked with asterisks. (B) WT and mutant NiV-G CSE, ephrinB2 (B2) binding, and cell-cell fusion levels were determined in 293T cells. CSE and B2 levels were measured by flow cytometry with Ab167 or soluble ephrinB2-Fc, respectively. Cell-cell fusion was determined by counting syncytial nuclei (see Materials and Methods) induced by WT NiV-F and WT NiV-G (G) or headless mutant proteins (158, 164, and 167). All levels were normalized to that of WT G. Average values ± the standard error of the mean from at least three independent experiments are shown. (C) Representative cell-cell fusion images from panel B (16 to 20 h posttransfection) induced by WT NiV-F and full-length G (G), truncated G (158, 164, or 167), or the vector. (D) Western blot analysis of 293T cell lysates transfected with WT (G) or headless (158, 164, or 167) NiV-G. The upper segment of the left panel shows the higher-molecular-weight bands (full-length G) and the lower segment shows the lower-molecular-weight bands resulting from the truncated mutant proteins. The right panel shows the banding pattern of the headless mutant proteins in the absence (−) or presence (+) of PNGase F. The values to the left and right are molecular weights in thousands.

Interestingly, we observed a double banding pattern for mutant protein 158 and a triple banding pattern for mutant proteins 164 and 167. We confirmed that the loss of the upper band for mutant protein 158 was due to the removal of an N-glycosylation site from this mutant protein (an N-glycan is normally added to N159 in the NiV-G stalk, as previously shown by Biering et al. [18]). Treatment with PNGase F eliminated the upper band from mutant proteins 164 and 167 (Fig. 1D). We do not know what caused the remaining duplet band, but O-glycosylation and/or other posttranslational modifications of these truncated mutant proteins may be the reason (37). We also confirmed by flow cytometry that the fusion-triggering ability of the three NiV-G headless mutant proteins was independent of their ephrinB2 receptor-binding capabilities (Fig. 1B). As expected, all three headless mutant proteins (158, 164, and 167) were unable to bind soluble ephrinB2, since the known ephrinB2-binding site lies within the G head (10). Overall, these results suggest that the NiV-G stalk C-terminal region from aa 158 to aa 167 is important for F triggering and fusion promotion.

Residues 159 to 163 within the NiV-G stalk modulate NiV-induced cell-cell fusion.

To corroborate the importance of the region from aa 158 to aa 167 in modulating fusion and to map potential specific residues within the region from aa 158 to aa 167 that modulate fusion, we performed alanine scanning mutagenesis of this region in the context of the full-length NiV-G glycoprotein (Fig. 2A). Cysteine 162 was left unchanged, since it has previously been shown, together with two other cysteine residues (C146 and C158), to be important for the tetrameric stability of G (30).

FIG 2.

FIG 2

Residues 159 to 163 within the NiV-G stalk modulate NiV-induced cell-cell fusion. (A) Schematic representation of the C-terminal stalk domain (Stalk C-term) is shown together with the alanine scanning mutant proteins. Alanine scanning mutant proteins were made in the context of full-length NiV-G. (B) Expression levels of full-length WT NiV-G (G) and alanine mutant proteins were measured by Western blotting of transfected 293T cell lysates with polyclonal anti-G antiserum Ab167. (C) The relative levels of CSE (806), ephrinB2 (B2) binding, and fusion of WT NiV-G (G) and alanine mutant proteins were determined in 293T cells. CSE and B2 receptor binding were measured by flow cytometry with polyclonal antibody 806 and soluble ephrinB2-Fc (B2), respectively, and cell-cell fusion levels were determined in the presence of WT NiV-F by counting syncytial nuclei (see Materials and Methods). All levels were normalized to those of WT NiV-G. Average values ± the standard error of the mean from at least three independent experiments are shown. P values were calculated by using Student's unpaired t test. (D) Lysates from 293T cells transfected as shown in Fig. 2B were separated by nondenaturing 8% SDS-PAGE, and NiV-G was detected by immunoblotting with anti-G polyclonal antibody 806. The monomer (1×), dimer (2×), and tetramer (4×) forms of NiV-G are shown. The values to the right of panels B and D are molecular weights in thousands.

293T cells expressing the WT or mutant NiV-G protein were lysed and subjected to SDS-PAGE and Western blot analysis with Ab167. As expected, mutations in positions 159 and 161 (N159A, S161A) resulted in a faster migration pattern of these proteins because of the disruption of the N-glycosylation site (NIS) we previously reported (Fig. 2B) (18). Interestingly, mutation of the isoleucine centering the glycosylation site resulted in additional bands of slightly higher molecular weight, indicating that this mutation might also have an effect on G folding, glycosylation, and/or other posttranslational modifications. All of the alanine mutant proteins were expressed at levels roughly similar to that of WT G, except the N159A and S161A mutant proteins, which showed slightly lower total cell expression levels (Fig. 2B). Next, we determined by flow cytometry with anti-G polyclonal antiserum 806 whether expression levels at the cell surface corresponded to that of the whole-cell lysates (34). These data indicate that the alanine point mutations had no drastic effects on the CSE levels of G (Fig. 2C).

To measure the ability of the NiV-G alanine mutant proteins to promote cell-cell fusion, we quantified the relative amounts of 293T cell-cell fusion induced by WT or mutant G in combination with WT NiV-F. We previously showed that cell-cell fusion and CSE levels are linearly correlated during transfection with the amount of DNA used in this study (2 μg per well of a six-well plate) (32). Table 1 shows the calculated fusion index (fusion/CSE) of each mutant protein, which is defined as the ratio of the cell-cell fusion level to the CSE level (each normalized to WT levels) (18, 32). Thus, the fusion index of WT G and F was set to 1, meaning that fusion indices higher than 1 describe a hyperfusogenic phenotype and fusion indices below 1 describe a hypofusogenic phenotype. Interestingly, the N159A and I160A mutant proteins did not induce any 293T cell-cell fusion (fusion indices of 0), although they were expressed on the cell surface (Fig. 2C; Table 1). In contrast, the S161A mutation induced a higher level of cell-cell fusion, despite the slightly lower CSE level than that of WT G (fusion index of 2.63) (Fig. 2C; Table 1), and the P163A and L166A mutant proteins displayed severe and mild hypofusogenic phenotypes (fusion indices of 0.05 and 0.42), respectively (Fig. 2C; Table 1). Overall, alanine mutations at N159, I160, S161, and P163 had pronounced effects on the fusion promotion ability of NiV-G (P < 0.001 for each).

TABLE 1.

Fusion indices (fusion/CSE ratios) of WT NiV-G and alanine mutant glycoproteinsa

G env % CSE % Fusion Fusion index
NiV-G 100 100 1.00
N159A mutant 68 0 0.00
I160A mutant 105 0 0.00
S161A mutant 65 171 2.63
P163A mutant 81 4 0.05
N164A mutant 81 90 1.11
P165A mutant 76 71 0.93
L166A mutant 97 41 0.42
P167A mutant 84 81 0.96
a

Both fusion (nuclei in syncytia) and CSE levels were normalized to those of WT NiV-G, which were set at 100%. The ratio of normalized fusion to CSE was calculated for each mutant protein.

Generally, the avidity of cell receptor (ephrinB2) and NiV-G interactions affects the fusion promotion capability of the viral attachment protein (2, 3, 7). Thus, we quantified the ability of WT and mutant NiV-G proteins to bind ephrinB2 via flow cytometry. The normalized receptor-binding level of each mutant was roughly similar to its normalized CSE level (Fig. 2C). These data indicate that the hyper- or hypofusogenic phenotypes of the alanine mutant proteins are not due to aberrant receptor-binding abilities, and aa 159 to 163 are indeed important in modulating cell-cell fusion.

The stalk domain is known to be important for tetramerization of the paramyxovirus attachment proteins, including NiV-G (27, 30, 38). Since cysteines at positions 146, 158, and 162 (C146, C158, and C162) have been implicated as important for G tetramerization (30), we investigated whether the hyper- and/or hypofusogenic phenotypes of the 159 to 163 alanine mutant proteins resulted from alterations in the tetrameric stability of G. We expressed WT or mutant NiV-G in 293T cells and subjected the cell lysates to nonreducing SDS-PAGE and immunoblotting with polyclonal antiserum 806. While for the WT and most of the mutant NiV-G proteins, the most prominent structures were tetrameric and dimeric forms of G, the I160A mutant protein showed less tetrameric (5.8-fold decrease) and more prominent dimeric and monomeric species (2.0- and 4.7-fold increases, respectively) than WT G (Fig. 2D). The relatively weaker tetrameric structure of the I160A mutant protein may be attributed to reduced hydrophobic interactions (lack of a hydrophobic isoleucine). This result, in combination with results for NiV-G stalk cysteine mutant proteins (30), suggests that severe weakening of the tetrameric structure has profound effects on the fusion promotion ability of G. Interestingly, the I160A mutant protein monomers and dimers migrated more slowly than those of WT NiV-G, possibly because of altered posttranslational modifications of this mutant protein.

Additionally, lower levels of dimers were detected for the N159A (18%) and S161A (23%) mutant proteins than for WT NiV-G (44%) (Fig. 2D). This suggests that these two mutant G proteins have increased oligomeric stability and that the glycosylation motif NIS is important for this oligomeric stability. However, neither the absence of an N-glycan nor the stability of the tetramer explains why the S161A mutant protein results in a hyperfusogenic phenotype while the N159A mutant protein results in a severely hypofusogenic phenotype, as neither protein is N-glycosylated at this site and both have similar overall distributions of tetrameric, dimeric, and monomeric forms.

Membrane fusion proteins with mutations in aa 159 to 163 within the NiV-G stalk modulate receptor induced conformational changes differently than WT NiV-G.

Since alanine mutations in aa 159 to 163 induced both hyper- and hypofusogenic phenotypes, we used a panel of previously reported conformation-dependent antibodies to investigate whether mutations in this region modulate receptor-induced conformational changes in NiV-G (28, 34). We recently described three receptor-induced conformational steps in NiV-G important for F triggering, detectable by Mab213 (step 1), Mab45 (step 2), and Ab167 (step 3) (28). We showed that ephrinB2 binding to NiV-G decreases the binding of conformational antibody Mab213 (step 1) to NiV-G by approximately 70%. We previously mapped the Mab213-binding epitope to region β3H1/β3H2 (aa 371 to 392) at the membrane-distal portion of the NiV-G head, near the receptor-binding site, and showed that receptor binding does not compete with Mab213 for binding to NiV-G (28, 34). Here, we found that the addition of ephrinB2 to cells expressing the N159A, I160A, S161A, and P163A mutant proteins also resulted in an ∼70% decrease in Mab213 binding (Fig. 3A), indicating that these mutations had no significant effect on the first conformational change at the membrane-distal portion of the NiV-G head.

FIG 3.

FIG 3

Proteins with alanine mutations in aa 159 to 163 within the NiV-G stalk modulate receptor induced conformational changes differently than WT NiV-G. WT or mutant NiV-G proteins were expressed in PK13 cells (ephrinB2 deficient), and cell surface binding levels were determined by flow cytometry with conformational antibodies Mab213 (213), Mab45 (45), and Ab167. (A) In the absence (0 nM) or presence (100 nM) of ephrinB2, antibody binding levels at 100 nM B2 were normalized to the binding levels at 0 nM B2 (dashed line) for each mutant protein. (B) In the absence of the receptor, antibody binding levels were normalized to the level of polyclonal anti-G antiserum 806 binding (CSE). The ratios of conformational antibody binding levels to CSE levels were determined and normalized to the WT NiV-G ratios (dotted line). Data shown are average values ± SEM from at least three independent experiments.

EphrinB2 binding to NiV-G also induces a roughly 2-fold increase in Mab45 binding to NiV-G (step 2), a step important for F triggering (28, 30, 31, 34). We previously mapped the Mab45 epitope to region β6S4/β1H1 (aa 177 to 194) at the base of the NiV-G head (28). Here, we found no significant enhancement of Mab45 binding to any of the aa 159 to 163 mutant proteins, with the possible exception of a slight, although not statistically significant (P = 0.08), enhancement for the P163A mutant protein. In contrast, ephrinB2 binding to WT NiV-G resulted in a roughly 2-fold increase in Mab45 binding, as expected (Fig. 3A). These results suggest that none of these mutant proteins undergo conformational step 2, which occurs at the base of the NiV-G head, in the same way that WT NiV-G does.

Conformational Ab167 is a polyclonal antiserum raised against the NiV-G stalk (aa 1 to 167) (28). EphrinB2 binding to NiV-G enhances binding of Ab167 to NiV-G by about 1.5- to 2-fold (step 3) (28). As expected, WT NiV-G showed enhanced receptor-induced binding to Ab167 (1.7-fold), in contrast to any of the aa 159 to 163 mutant proteins, with the exception of the P163A mutant protein, in which a very slight enhancement of Ab167 binding was observed (∼20% increase; P < 0.001) (Fig. 3A).

We then asked whether the lack of receptor-induced conformational changes in the mutant G proteins was due to differences from the pre-receptor-binding conformation of the WT NiV-G protein. To account for CSE differences, we calculated the ratio of the levels of normalized conformational antibody binding to CSE for each mutant protein in the absence of the ephrinB2 receptor (Fig. 3B). As somewhat expected, all of the mutant proteins bound Mab213 at levels similar to that of WT G, with the exception of the I160A mutant (Fig. 3B), which showed only 70% of the Mab213 binding of WT NiV-G, possibly because of the aberrant oligomerization observed for I160A (P < 0.001; Fig. 2D). In contrast, all of the mutant proteins had greater Mab45 binding (1.5- to 2-fold) and, with the exception of the P163A mutant protein, greater Ab167 binding (1.2- to 1.7-fold) than WT NiV-G (P < 0.001; Fig. 3B). Interestingly, the overall trends of Mab45 and Ab167 binding to the WT and mutant NiV-G proteins were similar, suggesting that the receptor-induced conformational changes detected by Mab45 and Ab167 are intricately linked. Moreover, the P163A mutant protein bound Ab167 at levels similar to that of WT G (Fig. 3B). Overall, these results suggest that the pre-receptor-binding conformation of G is altered by mutations in aa 159 to 163, affecting the Mab45 and Ab167 but not the Mab213 epitope binding sites. Therefore, premature exposure of the epitopes recognized by Mab45 and Ab167 prior to receptor binding likely prevents receptor-induced conformational changes in these two epitopes, at least for some of these mutant proteins.

An optimal NiV-G conformation is important for efficient F triggering.

Next, we studied the temporal dynamics of cell-cell fusion induced by the N159A, I160A, S161A, and P163A mutant proteins by measuring cell-cell fusion changes over time (12, 16, 22, and 28 h posttransfection). No cell-cell fusion was observed with the N159A or I160A mutant protein at any time point, in contrast to the P163A mutant protein, which started causing cell-cell fusion at 16 h posttransfection (5% compared to WT G) (Fig. 4A). The cell-cell fusion levels induced by the S161A mutant protein were nearly 2-fold higher than those induced by WT NiV-G at all of the time points studied (Fig. 4A). Note that at 28 h posttransfection, cell-cell fusion had progressed so far for WT G and the S161A mutant protein that cells started lifting off the plate and fusion levels were uncountable. These data suggest that while the N159A and I160A mutant proteins are severely hypofusogenic, the P163A mutant protein is a “slower” fusion mutant.

FIG 4.

FIG 4

An optimal conformation of NiV-G is important for efficient F triggering. (A) Representative images showing 293T cell-cell fusion induced by NiV-F and WT (G) or mutant NiV-G at 12, 16, 22, and 28 h posttransfection. Arrows point to syncytia. (B) 293T cell-cell fusion determined for WT (G) or mutant (S161A) NiV-G transfected together with WT NiV-F. Fusion levels determined in the presence of EphB3 (40 nM) were normalized to those determined in the absence of EphB3 (0 nM). (C) 293T cell-cell fusion determined for WT (G) or mutant (S161A) NiV-G transfected together with WT F or hypofusogenic mutant NiV-F (FP221A). Fusion levels were normalized to those obtained for WT NiV-G and -F. (D) Cell-cell fusion was determined in 293T cells. WT (G) or hypofusogenic NiV-G was cotransfected with WT (F) or hyperfusogenic NiV-F. Fusion levels were normalized to those obtained for WT G and F. (E) Cell-cell fusion levels were quantified in Vero cells coexpressing WT F and WT or mutant NiV-G at 8 to 10 h posttransfection. All fusion levels were normalized to that of WT NiV-G (G).

Interestingly, the S161A mutant protein showed a hyperfusogenic phenotype even though it did not exhibit enhanced Mab45 and Ab167 binding (Fig. 2C and 3A). From these unexpected results, we hypothesized that the S161A mutant protein may have acquired a “prone-to-trigger” conformation, which undergoes the receptor-induced conformational cascade more readily than WT G.

We then tested whether such a prone-to-trigger conformation may occur independently of receptor binding. Thus, we added soluble EphB3 (40 nM), a natural ligand of ephrinB2/B3, to cells transfected with S161A mutant and WT NiV-F (28). Cell-cell fusion was determined at 16 to 24 h posttransfection, and we observed that, at this concentration, EphB3 inhibited S161A mutant protein-induced fusion at levels at least similar to those of WT NiV-G-induced fusion (about 60 to 70% inhibition) (Fig. 4B). We also tested cell-cell fusion of this mutant protein in PK13 cells (receptor deficient) and observed a lack of cell-cell fusion induced by S161A or WT NiV-G (data not shown). Combined, these results indicate that receptor binding is required for S161A mutant G to induce cell-cell fusion.

Next, we asked whether a lower degree of receptor-induced conformational changes may be required for S161A mutant than for WT NiV-G to overcome a presumed energy barrier needed to trigger F (15). Therefore, we tested if S161A mutant G was able to rescue cell-cell fusion induced by our available hypofusogenic F mutant proteins. We observed that S161A mutant G was able to rescue the level of cell-cell fusion of NiV-F hypofusogenic mutant FP221A, increasing the FP221A fusion index from 0.15 to 1.0 (WT fusion index) (Fig. 4C). Although we do not fully understand the mechanism of the hypofusogenicity of FP221A, which is beyond the scope of this study (unpublished data), the fact that the S161A mutant protein can rescue FP221A suggests that the S161A mutant protein is in a prone-to-trigger F conformation. Such a conformation presumably requires a lower degree of conformational changes to trigger F than WT G does (Fig. 3 and 4C). Notably, the S161A mutant protein rescued FP221A without increasing the CSE levels of FP221A (data not shown). Our results also suggest that the S161A conformation may be at least partially downstream of conformational steps 2 and 3, as for S161A receptor-induced conformational steps 2 and 3 were not detected by Mab45 or Ab167, but these antibodies bind S161A at higher-than-WT levels (P < 0.05; Fig. 3).

We then asked if previously described hyperfusogenic NiV-F mutant proteins (FF3F5, FK1A, and FK1A F3F5) could rescue cell-cell fusion induced by the hypofusogenic N159A, I160A, and P163A mutant NiV-G proteins. FF3F5 lacks two N-glycosylation sites in the ectodomain of F, while FK1A is a cytoplasmic tail mutant (32, 33). Interestingly, hyperfusogenic FK1A F3F5, when coexpressed with the N159A mutant protein, reached only 10% of the cell-cell fusion level obtained with WT G in 293T cells (Fig. 4D). Next, as NiV glycoproteins induce cell-cell fusion more readily in Vero cells than in 293T cells, we investigated the fusion promotion of some mutant G proteins in Vero cells (32, 33; data not shown). Interestingly, the N159A mutant protein induced Vero cell-cell fusion at about 40% of the WT G levels in combination with WT NiV-F (Fig. 4E). These results suggest that in highly fusogenic environments, the N159A mutant protein retains limited membrane fusion promotion ability. Interestingly, none of the hyperfusogenic F mutant proteins rescued I160A-induced 293T cell-cell fusion (Fig. 4D). In contrast, all of the hyperfusogenic F mutant proteins rescued P163A mutant protein-induced 293T cell-cell fusion, reaching nearly WT F and G levels when coexpressed with the FK1A F3F5 mutant protein (Fig. 4D). These results indicate that an intact oligomeric structure is required for the fusion promotion ability of NiV-G, as shown by all of the results obtained with the I160A mutant protein. Additionally, these results, together with results in Fig. 3, suggest that mutations at positions N159 and P163 resulted in a suboptimal G conformation for F triggering, causing low levels of fusion and/or slow fusion kinetics (see Fig. 7).

FIG 7.

FIG 7

Mechanistic model of NiV membrane fusion triggering. (A) The NiV-G stalk C-terminal region (C-term) that triggers F is covered by the NiV-G heads prior to ephrinB2 receptor binding. Upon receptor binding, G undergoes conformational step 1 at the top of the G head (B) and conformational step 2 at the base of the G head (C), resulting in the exposure of the C-terminal region of the stalk (conformational step 3). (D) Consequently, receptor-induced conformational changes (conformational step 4) in the C-terminal region of the G stalk trigger F and F undergoes a conformational cascade resulting in dissociation of F from G and membrane fusion. The S161A mutant protein acquires a prone-to-trigger conformation. Upon receptor binding, the S161A mutant protein somewhat bypasses conformational steps 2 and 3 and thus triggers F more readily than the WT does. The P163A mutant protein may have a conformation similar to that of the S161A mutant protein, although the P163A mutant protein is unable to dissociate from F efficiently, thus making fusion inefficient. The N159A mutant protein may be trapped in a low-energy conformation, as energy provided by receptor binding triggers only a very limited degree of conformational changes in F, dissociation of F from G, and membrane fusion. The I160A mutant protein may be in a postfusion conformation, which cannot trigger membrane fusion.

Aa 159 to 163 in the NiV-G stalk C-terminal portion modulate viral entry.

To test whether proteins with mutations in aa 159 to 163 trigger NiV-F equivalently on cell or viral surfaces, we produced NiV/VSV pseudotyped virions expressing a Renilla luciferase reporter gene by using our previously established quantitative BSL2 viral entry assay (18, 28). When compared at several logs of viral input (as determined by VSV genome copy numbers), WT NiV/VSV virions showed up to 3 orders of magnitude increased viral entry compared to the negative-control virus that lacks NiV-F (G alone). In general, the cell-cell fusion phenotypes of the alanine mutant proteins correlated with their viral entry phenotypes (Fig. 2C and 5A). For example, the fusion-dead I160A mutant protein was unable to promote viral entry (similarly to the negative-control “G-alone” virions [Fig. 2C and 5A]), and the hypofusogenic P163A mutant protein yielded about 1-log lower levels than WT “G” virions (Fig. 2C and 5A). The hyperfusogenic S161A mutant virions exhibited entry similar to that of WT virions, as expected, since previous studies showed that generally 3- to 6-fold increases in cell-cell fusion levels are required to detect an increase in viral entry levels (Fig. 5A) (18, 32, 33). Not so surprisingly, the 293T fusion-dead N159A mutant protein induced viral entry at greater-than-background G alone levels, as the N159A mutant protein retained limited F-triggering ability (Fig. 4D and E).

FIG 5.

FIG 5

Aa 159 to 163 of the NiV-G stalk C-terminal portion modulate viral entry. (A) To measure viral entry, serial dilutions of NiV/VSV-rLuc pseudotyped virions containing either WT (G) or mutant NiV-G in combination with or without NiV-F (G alone) were used to infect 293T cells. Cells were lysed at 20 to 24 h postinfection, and relative light units (RLU) were quantified and plotted against the number of viral genomes/ml. Viral entry data are not shown for pseudotyped virions made with F only (F alone) or the empty vector (Vector) since they yielded viral entry levels similar to those of G-alone virions. Data shown are average values ± SEM from at least three independent experiments. P values were calculated by using Student's unpaired t test. Asterisks represent statistically significant differences (*, < 0.001) between the WT and mutant levels of viral entry at the genome copy numbers indicated. (B) NiV/VSV-rLuc pseudotyped virions used in panel A were denatured, and proteins were separated by 10% SDS-PAGE and immunoblotted against F with a mouse anti-AU1 monoclonal antibody and against G with polyclonal antiserum Ab167. The lower panel is an overexposed image showing the higher-molecular-weight species of the I160A mutant protein.

We then monitored the levels of F and G incorporation into virions by Western blotting. Point mutations in the NiV-G region from aa 159 to aa 167 mostly did not significantly affect the incorporation levels of NiV-G, except for the N159, I160, and P163 mutant proteins, for which the incorporation levels were relatively lower than those of WT G (Fig. 5B). The low incorporation level of the I160A mutant protein into virions is likely due to its defective oligomerization stability. Interestingly, the levels of incorporation of NiV-F appeared to be somewhat lower for those mutant G proteins with lower G incorporation levels (Fig. 5B). An overexposed version of these images shows that I160A mutant G is incorporated at very low levels.

Overall, the viral entry and cell-cell fusion results were roughly consistent. In addition, the viral entry levels induced by the hypofusogenic and less-incorporated N159A mutant protein suggest that viral entry might be a more efficient process than cell-cell fusion, at least for this mutant protein (Fig. 2C and 5).

NiV-G stalk aa 159 to 163 modulate F triggering and G-F interactions.

To determine directly whether the alanine mutant proteins are able to trigger NiV-F in NiV/VSV pseudotyped virions, we took advantage of a confocal micro-Raman spectroscopic technique we recently developed (36). This technique allows the detection of receptor-induced conformational changes in NiV-F embedded in the surfaces of the NiV/VSV pseudotyped virions (36). In our recent study, we showed that ephrinB2-induced conformational changes in NiV-F, observed as a downward shift of a Raman signal at a wave number of 1,409 cm−1, corresponded to the extent of conformational changes in F during F triggering. As shown in Fig. 6A, no downward shift was observed in negative-control virions containing NiV-F only (No G, red line), whereas a significant downward shift was observed in WT NiV/VSV virions that harbor both WT F and G (G). As expected, I160A mutant virions yielded a Raman spectral pattern similar to that of negative-control virions. In addition, S161A and P163A mutant virions yielded levels of F triggering similar to those of WT virions, and an intermediate F triggering phenotype was observed for the N159A mutant protein, corroborating its intermediate viral entry phenotype (Fig. 5A and 6A). Although the Raman spectroscopic analysis was not sensitive enough to show differences in receptor-induced conformational changes in F between the P163A and S161A mutant and WT virions, our overall results still suggest important roles for residues in aa 159 to 163 within the NiV-G stalk in F triggering.

FIG 6.

FIG 6

NiV-G stalk aa 159 to 163 modulate F triggering and G-F interactions (A) Raman spectral features were measured for WT NiV-F triggered by ephrinB2-bound WT (dark blue, G) or mutant NiV-G pseudovirions (cyan, N159A mutant protein; green, I160A mutant protein; black, S161A mutant protein; magenta, P163A mutant protein) and negative-control virions (red, No G). The lower the 1,409-cm−1 peak is, the greater is the extent of F triggering. (B) The G-F interactions were determined for WT NiV-F and WT or mutant NiV-G by immunoprecipitating NiV-G from transfected 293T cells with μMACS anti-HA Microbeads. Total cell lysates (lysate) and coimmunoprecipitated proteins (IP: αHA) were separated by 10% SDS-PAGE and immunoblotted with NiV-F specific antibodies (IB: αAU1) and NiV-G-specific polyclonal rabbit anti-HA antiserum (IB: αHA). Lysate from cells transfected with F only or G only served as a negative or positive control, respectively.

For paramyxoviruses, the cell receptor type appears to determine the nature of the interactions between the attachment and fusion glycoproteins. For MeV and NiV, protein receptor binding appears to induce the dissociation of previously associated H-F or G-F complexes, and such a dissociation is a critical step in membrane fusion modulation (14, 32, 33). Therefore, it is plausible that the hypofusogenic phenotypes of the N159A, I160A, and P163A mutant proteins are partly due to their decreased ability to release the cognate F upon receptor binding. We used HA-tagged WT and mutant NiV-G proteins to determine the avidities of G-F interactions, since HA-tagged WT and mutant NiV-G had fusion phenotypes similar to those of their untagged counterparts (data not shown). We immunoprecipitated NiV-G from 293T cells expressing NiV-F and WT or mutant NiV-G, with μMACS anti-HA Microbeads. The cell lysates, as well as immunoprecipitated proteins, were separated by SDS-PAGE and analyzed by immunoblotting with antibodies specific for F and G. Whereas immunoprecipitated WT and mutant NiV-G proteins were detected at roughly equal levels, the levels of coimmunoprecipitated NiV-F (F0, F1) varied among the mutant NiV-G proteins (Fig. 6B, right panel). In general, hypofusogenic the N159A and P163A mutant proteins exhibited somewhat greater G-F avidities than WT G, whereas the hyperfusogenic S161A mutant protein did not. We speculate that the hypofusogenic I160A mutant protein did not show increased G-F avidities likely because of its anomalous tetrameric structure. As expected, no F was pulled down from the F-only-containing cell lysate. These results also indicated that all of the mutant G proteins retain the ability to interact with F.

We measured G-F interaction avidities by calculating the ratios of coimmunoprecipitated F to the corresponding amount of NiV-F in the total cell lysates for WT G and each mutant protein. The ratios were then normalized to that of WT G, which was set as 1. For those mutant NiV-G proteins that retained an intact tetrameric structure, we observed an inverse correlation between the fusion phenotype of our mutant NiV-G proteins and their avidities of G-F interactions, similarly to previous experiments with NiV-F mutant proteins (Fig. 6B; Table 2) (32, 33). The hyperfusogenic S161A mutant protein was able to pull down F at a level similar to that of WT G (G-F interaction avidity of 1.3; P > 0.05), whereas the hypofusogenic N159A and P163A mutant proteins pulled down F at greater levels than WT G (G-F interaction avidities of 5.0 and 2.7, respectively; P < 0.05) (Fig. 6B, right panel; Table 2). Therefore, the hypofusogenic phenotypes of at least the N159A and P163A mutant proteins may be related to their decreased ability to release NiV-F. These data suggest that mutations in aa 159 to 163 within the NiV-G stalk affect the avidities of G-F interactions and likely G-F dissociation upon fusion triggering.

TABLE 2.

Fusion indices and relative avidities of G-F interactions of WT NiV-G and alanine mutant proteinsa

Env protein Fusion index G-F interaction avidity
NiV-G 1.00 1
N159A mutant 0.00 5.0 ± 1.2b
I160A mutant 0.00 1.6 ± 0.2
S161A mutant 2.63 1.3 ± 0.1
P163A mutant 0.05 2.7 ± 0.5b
a

Fusion indices are from Table 1. The amount of F in total cell lysates (Fig. 6B, left panel) and coimmunoprecipitated F (Fig. 6B, right panel) were quantified by densitometry with a Chemidoc gel imaging system as indicated in Materials and Methods. The avidities of G-F interactions are represented by the ratio of the amount of coimmunoprecipitated F to the amount of the corresponding F in the total cell lysates. The G-F interaction avidity of each mutant protein was normalized to that of WT NiV-G. The data presented are average values ± SEM from five independent experiments. P values were calculated with Student's unpaired t test.

b

Statistically significant difference from the WT value (P < 0.05).

DISCUSSION

The stalk domains of the HeV and NiV attachment proteins have been reported as critical for the proper expression of G and stabilization of the G oligomeric structure (29, 30). Recently, we reported for NiV a three-step spatiotemporal mechanism of receptor-induced F triggering by G. Upon ephrinB2/B3 receptor binding, the NiV-G head undergoes at least two conformational changes that result in the exposure of a region likely at the C terminus of the NiV-G stalk that triggers F (28). However, the precise domains or residues in the NiV-G stalk that trigger F, as well as the precise events occurring in G that link receptor binding to F triggering are largely unknown. Here, we identified a short NiV-G stalk C-terminal region of aa 159 to 163 as important for NiV-G oligomerization, conformational integrity, receptor-induced conformational changes in G, G-F interactions, and F triggering. The HNV-G head, but not the HNV-G stalk, has been crystallized (10, 39). Our findings suggest that aa 159 to 163 at the NiV-G stalk C terminus function as an important structural/functional link between the receptor-binding head and the F-triggering stalk.

The stalk ectodomain of NiV-G (residues 71 to 188) can be divided into four subdomains, I (residues 71 to 100), II (residues 101 to 145), III (residues 146 to 173), and IV (residues 174 to 188), on the basis of functional criteria, sequence conservation, and structural homology (30). Aa 159 to 163 lie in domain III, which is 100% conserved between HeV-G and NiV-G. The crystal structure of the PIV5-HN ectodomain revealed a flexible linker between the stalk 4HB and head domains (24). Since NiV-G stalk domain III connects to domain II at its N terminus and domain II aligns well with the rigid 4HB structure of the PIV5 HN stalk, we speculate that for the HNVs, domain III serves as a linker between the HNV-G head and the stalk helical domain.

On the basis of several paramyxoviral studies, Bose et al. proposed an “induced-fit” mechanism to explain the G/H/HN-F interactions upon F triggering (25). This mechanism emphasizes the “flexibility” of the stalk domain and proposes that the F-triggering “signal” is propagated from the head to a flexible stalk region without causing extensive conformational changes in the stalk. Previous studies suggested that the linker region is so flexible that the addition of N-glycan moieties to it in PIV5 HN or the introduction of disulfide bonds to the C-terminal region of the MeV H stalk did not result in a major disruption of their function (19, 40, 41). These results imply that receptor binding to the heads of PIV5-HN and MeV-H may not lead to extensive conformational changes in this highly flexible stalk domain. However, NiV studies suggest that the flexibility of the C-terminal portion of the NiV-G stalk is not as high as that of PIV5-HN or MeV-H (30). NiV-G contains a distinct cluster of three cysteine residues (C146, C158, and C162) in the membrane-distal portion of the stalk domain (30). C158 and C162 likely mediate covalent subunit dimerization, while C146 is involved in interdimeric disulfide bond formation (tetramerization) (30). In contrast, dimers of PIV5 and NDV-HN are noncovalently linked into tetramers through the stalk domain (11, 24). This contrast supports the notion that the NiV-G stalk is not as flexible as the stalk domains of PIV5 and NDV-HN.

Our data suggest that an alanine substitution at I160 strongly weakens the tetrameric structure of NiV-G, likely because of the loss of a hydrophobic side chain (Fig. 2D). Moreover, the I160A mutant protein failed to promote cell-cell fusion or viral entry, although it was expressed on the cell surface and showed ephrinB2 receptor binding levels similar to WT levels (Fig. 2C, 4A, and 5A). In agreement with our results, the stalk cysteine mutant NiV-G proteins, which have aberrant tetrameric structures, were completely deficient in fusion triggering, despite their ephrinB2-binding abilities (30). Interestingly, the I160A mutant protein exhibited an additional electrophoretic species that ran at an apparently slightly higher molecular weight than WT G (Fig. 2B and 5B). Bishop et al. mutated the isoleucines to alanines in the HeV-G stalk region (aa 80 to 177) and observed similar banding patterns for all of the stalk I-to-A mutant HeV-G proteins that were fusion defective (29). This aberrant banding pattern of I160A mutant NiV-G suggests an altered conformation and/or posttranslational modification, likely responsible for the fusion-dead phenotype of the I160A and I-to-A mutant NiV-G proteins in the HeV-G stalk region. Abolishment of the N-glycosylation site NIS in the N159A and S161A mutant NiV-G proteins reduced their levels of the dimeric G and caused prominent differences from WT G in their fusion phenotypes (hypo- and hyperfusogenic, respectively) (Fig. 2C and D). These data suggest that not only the disruption of the N-glycosylation site in the stalk region but also the removal of the hydrophobic isoleucine that is centering the N-glycosylation motif severely affects the function of NiV-G. Therefore, the NiV-G stalk C terminus is likely more rigid than the highly flexible PIV5-HN and MeV-H stalk C terminus and the former does not as readily allow amino acid changes to occur without affecting HNV-G function (19, 40, 41). Thus, it is likely that some conformational changes in this HNV stalk region are required to trigger F upon receptor binding.

In our recent report of at least three receptor-induced spatiotemporal conformational changes in NiV-G (28), key phenotypes associated with the F-triggering ability of NiV-G are the enhancements of Mab45 and Ab167 binding to NiV-G upon receptor engagement (28, 34). Consistently, the strongly hypofusogenic N159A and I160A mutant proteins did not enhance Mab45 or Ab167 binding upon ephrinB2 receptor engagement, in contrast to their enhanced Mab45 and Ab167 binding in the absence of ephrinB2 (Fig. 3). These data suggest that this G region is important for allowing NiV-G to maintain its prefusion conformation, permissible of receptor-induced conformational changes that trigger F (Fig. 7). Additionally, the level of Mab45 binding to the N159A mutant protein being less than that of the I160A mutant protein in the absence of receptor engagement is consistent with N159A mutant protein retaining some fusion-triggering ability, as observed in relatively highly fusion-susceptible Vero cells (Fig. 3B and 4E).

Interestingly, the hyperfusogenic S161A mutant protein did not exhibit significant enhancement of either Mab45 or Ab167 binding upon receptor engagement (Fig. 3A). Compared to WT G, the increased avidities of the S161A mutant protein for Mab45 and Ab167 suggest that the pre-receptor-binding conformation of the S161A mutant protein is distinct from that of WT G. Nonetheless, the S161A mutant protein required receptor binding to trigger F and rescued the hypofusogenic phenotype of FP221A in 293T cells (Fig. 4B and C). Furthermore, the F-triggering ability of the S161A mutant protein was similar to that of WT G, as shown by Raman spectroscopic analysis (Fig. 6A). Considering all of these results, we speculate that this mutation provides NiV-G a prone-to-trigger conformation partially downstream of conformational steps 2 and 3 and upstream of putative conformational step 4 (Fig. 7). Conformational step 4 may be a rearrangement of the dimer interface in the NiV-G stalk domain or some other conformational change that directly triggers F. Using circular dichroism, we previously found receptor-induced changes in the secondary structure of a mutant NiV-G protein that contained the entire ectodomain but not in a mutant G protein that contained the head domain only. This indicates that the G stalk may undergo receptor-induced secondary structural changes (34). A partial rearrangement possibly occurring at the dimer interface caused by the S161A mutation may increase the accessibility of the Mab45 and Ab167 epitopes independently of receptor engagement. Thus, the S161A mutant protein may trigger F, bypassing fusion steps such as those detected by Mab45 or Ab167 (Fig. 7).

Although the P163A mutant protein exhibited a conformational antibody binding profile similar to that of the S161A mutant protein (Fig. 3), it induced a slower-fusion phenotype but in combination with a hyperfusogenic mutant F protein (FK1A F3F5) reached fusion levels similar to those of WT F and G (Fig. 2C and 4A and D). These results suggest that the P163A mutant protein may have a compromised F-triggering conformation yet different from that of the other point mutant proteins analyzed in this region or from that of WT G. Additionally, the P163A mutant protein showed increased G-F avidity, suggesting a decreased ability to dissociate from F upon receptor engagement (Fig. 6B; Table 2). Overall, the slower-fusion phenotype of the P163A mutant protein may be due partly to its compromised prefusion conformation and its inefficient dissociation from F upon receptor binding (Fig. 7C).

Some of our point mutations, as well as some hypofusogenic isoleucine mutations in the HeV-G stalk, did not yield receptor-induced conformational changes but also did not affect the tetrameric structure of NiV-G (29). It is thus noteworthy that receptor-induced conformational changes in NiV-G recognized by conformation-dependent antibodies may be linked to secondary, tertiary, and/or quaternary structural changes, as detectable by biophysical techniques such as circular dichroism or confocal micro-Raman spectroscopy (Fig. 6A) (34, 36).

Our findings indicate that NiV-G stalk aa 159 to 163 are a novel fusion-modulatory region in paramyxoviral attachment proteins that is critical to maintain NiV-G in a metastable pre-receptor-binding conformation. Upon receptor binding, this region likely helps the transmission of a “triggering signal” to F via specific conformational changes. Notably, similar trends in Mab45 and Ab167 binding were observed (Fig. 3), suggesting that the conformational changes detected by Mab45 and Ab167 are intricately linked, although Mab45 binds to the base of the G head and Ab167 to the stalk (28). In contrast, the trend for Mab213 binding was very different (Fig. 3). Since the Mab213 binding epitope is located at the top of the G head, our findings suggest that aa 159 to 163 serve as a structural and functional linker between the G head and stalk. The linked conformational changes observed in aa 159 to 163 (detected by Mab45 and Ab167) are consistent with the C-terminal membrane-distal portion of the NiV-G stalk allowing receptor-induced conformational changes in the head to be propagated to the stalk.

In conclusion, our data indicate that NiV-G C-terminal stalk aa 159 to 163 are important for NiV-G conformational integrity, proper tetramerization, avidity of G-F interactions, receptor-induced conformational changes, and F triggering. This region likely serves as a structural and functional linker between the NiV-G head and stalk. The flexibility of this region appears to be tightly regulated for the F-triggering signal to be properly propagated from head to stalk through specific conformational changes. Since this region is 100% conserved between NiV and HeV, these findings should apply to HeV if not also to other paramyxoviruses. To our knowledge, this is the most detailed mechanistic picture of NiV or HeV attachment protein linking receptor binding to F triggering to date. Solving the crystal structure of the full-length HNV G ectodomain will likely reveal structural relationships between the G head and stalk region (aa 159 to 163) and possibly further clarify the factors that govern the regulation of the conformational changes in the G stalk during the F-triggering cascade of events. Further similarities and differences between these events and those occurring in other paramyxoviruses remain to be elucidated.

ACKNOWLEDGMENTS

This study was supported by NIH/NIAID grant AI109022 to H.C.A. In addition, A.V.N. is supported by NIH/NIAID grant AI096103 and X.L. is supported by the Natural Sciences and Engineering Research Council of Canada.

REFERENCES

  • 1.Luby SP, Gurley ES. 2012. Epidemiology of henipavirus disease in humans. Curr Top Microbiol Immunol 359:25–40. doi: 10.1007/82_2012_207. [DOI] [PubMed] [Google Scholar]
  • 2.Negrete OA, Levroney EL, Aguilar HC, Bertolotti-Ciarlet A, Nazarian R, Tajyar S, Lee B. 2005. EphrinB2 is the entry receptor for Nipah virus, an emergent deadly paramyxovirus. Nature 436:401–405. doi: 10.1038/nrmicro1257. [DOI] [PubMed] [Google Scholar]
  • 3.Negrete OA, Wolf MC, Aguilar HC, Enterlein S, Wang W, Mühlberger E, Su SV, Bertolotti-Ciarlet A, Flick R, Lee B. 2006. Two key residues in ephrinB3 are critical for its use as an alternative receptor for Nipah virus. PLoS Pathog 2:e7. doi: 10.1371/journal.ppat.0020007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pernet O, Wang YE, Lee B. 2012. Henipavirus receptor usage and tropism. Curr Top Microbiol Immunol 359:59–78. doi: 10.1007/82_2012_222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wong K, Tan C. 2012. Clinical and pathological manifestations of human henipavirus infection. Curr Top Microbiol Immunol 359:95–104. doi: 10.1007/82_2012_205. [DOI] [PubMed] [Google Scholar]
  • 6.Aguilar HC, Lee B. 2011. Emerging paramyxoviruses: molecular mechanisms and antiviral strategies. Expert Rev Mol Med 13:e6. doi: 10.1017/S1462399410001754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Plattet P, Plemper RK. 2013. Envelope protein dynamics in paramyxovirus entry. mBio 4(4):e00413-13. doi: 10.1128/mBio.00413-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bowden TA, Crispin M, Harvey DJ, Aricescu AR, Grimes JM, Jones EY, Stuart DI. 2008. Crystal structure and carbohydrate analysis of Nipah virus attachment glycoprotein: a template for antiviral and vaccine design. J Virol 82:11628–11636. doi: 10.1128/JVI.01344-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Santiago C, Celma ML, Stehle T, Casasnovas JM. 2010. Structure of the measles virus hemagglutinin bound to the CD46 receptor. Nat Struct Mol Biol 17:124–129. doi: 10.1038/nsmb.1726. [DOI] [PubMed] [Google Scholar]
  • 10.Xu K, Rajashankar KR, Chan Y-P, Himanen JP, Broder CC, Nikolov DB. 2008. Host cell recognition by the henipaviruses: crystal structures of the Nipah G attachment glycoprotein and its complex with ephrin-B3. Proc Natl Acad Sci U S A 105:9953–9958. doi: 10.1073/pnas.0804797105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yuan P, Swanson KA, Leser GP, Paterson RG, Lamb RA, Jardetzky TS. 2011. Structure of the Newcastle disease virus hemagglutinin-neuraminidase (HN) ectodomain reveals a four-helix bundle stalk. Proc Natl Acad Sci U S A 108:14920–14925. doi: 10.1073/pnas.1111691108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yuan P, Thompson TB, Wurzburg BA, Paterson RG, Lamb RA, Jardetzky TS. 2005. Structural studies of the parainfluenza virus 5 hemagglutinin-neuraminidase tetramer in complex with its receptor, sialyllactose. Structure 13:803–815. doi: 10.1016/j.str.2005.02.019. [DOI] [PubMed] [Google Scholar]
  • 13.Aguilar HC, Iorio RM. 2012. Henipavirus membrane fusion and viral entry. Curr Top Microbiol Immunol 359:79–94. doi: 10.1007/82_2012_200. [DOI] [PubMed] [Google Scholar]
  • 14.Iorio RM, Melanson VR, Mahon PJ. 2009. Glycoprotein interactions in paramyxovirus fusion. Future Virol 4:335–351. doi: 10.2217/fvl.09.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.White JM, Delos SE, Brecher M, Schornberg K. 2008. Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit Rev Biochem Mol Biol 43:189–219. doi: 10.1080/10409230802058320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Aguilar HC, Aspericueta V, Robinson LR, Aanensen KE, Lee B. 2010. A quantitative and kinetic fusion protein-triggering assay can discern distinct steps in the Nipah virus membrane fusion cascade. J Virol 84:8033–8041. doi: 10.1128/JVI.00469-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wong KT, Shieh W-J, Kumar S, Norain K, Abdullah W, Guarner J, Goldsmith CS, Chua KB, Lam SK, Tan CT. 2002. Nipah virus infection: pathology and pathogenesis of an emerging paramyxoviral zoonosis. Am J Pathol 161:2153–2167. doi: 10.1016/S0002-9440(10)64493-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Biering SB, Huang A, Vu AT, Robinson LR, Bradel-Tretheway B, Choi E, Lee B, Aguilar HC. 2012. N-glycans on the Nipah virus attachment glycoprotein modulate fusion and viral entry as they protect against antibody neutralization. J Virol 86:11991–12002. doi: 10.1128/JVI.01304-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bose S, Welch BD, Kors CA, Yuan P, Jardetzky TS, Lamb RA. 2011. Structure and mutagenesis of the parainfluenza virus 5 hemagglutinin-neuraminidase stalk domain reveals a four-helix bundle and the role of the stalk in fusion promotion. J Virol 85:12855–12866. doi: 10.1128/JVI.06350-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gravel KA, Morrison TG. 2003. Interacting domains of the HN and F proteins of Newcastle disease virus. J Virol 77:11040–11049. doi: 10.1128/JVI.77.20.11040-11049.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Melanson VR, Iorio RM. 2006. Addition of N-glycans in the stalk of the Newcastle disease virus HN protein blocks its interaction with the F protein and prevents fusion. J Virol 80:623–633. doi: 10.1128/JVI.80.2.623-633.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Melanson VR, Iorio RM. 2004. Amino acid substitutions in the F-specific domain in the stalk of the Newcastle disease virus HN protein modulate fusion and interfere with its interaction with the F protein. J Virol 78:13053–13061. doi: 10.1128/JVI.78.23.13053-13061.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Stone-Hulslander J, Morrison TG. 1999. Mutational analysis of heptad repeats in the membrane-proximal region of Newcastle disease virus HN protein. J Virol 73:3630–3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Welch BD, Liu Y, Kors CA, Leser GP, Jardetzky TS, Lamb RA. 2012. Structure of the cleavage-activated prefusion form of the parainfluenza virus 5 fusion protein. Proc Natl Acad Sci U S A 109:16672–16677. doi: 10.1073/pnas.1213802109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bose S, Song AS, Jardetzky TS, Lamb RA. 2014. Fusion activation through attachment protein stalk domains indicates a conserved core mechanism of paramyxovirus entry into cells. J Virol 88:3925–3941. doi: 10.1128/JVI.03741-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bose S, Zokarkar A, Welch BD, Leser GP, Jardetzky TS, Lamb RA. 2012. Fusion activation by a headless parainfluenza virus 5 hemagglutinin-neuraminidase stalk suggests a modular mechanism for triggering. Proc Natl Acad Sci U S A 109:E2625–E2634. doi: 10.1073/pnas.1213813109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Brindley MA, Suter R, Schestak I, Kiss G, Wright ER, Plemper RK. 2013. A stabilized headless measles virus attachment protein stalk efficiently triggers membrane fusion. J Virol 87:11693–11703. doi: 10.1128/JVI.01945-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Liu Q, Stone JA, Bradel-Tretheway B, Dabundo J, Benavides Montano JA, Santos-Montanez J, Biering SB, Nicola AV, Iorio RM, Lu X. 2013. Unraveling a three-step spatiotemporal mechanism of triggering of receptor-induced Nipah virus fusion and cell entry. PLoS Pathog 9:e1003770. doi: 10.1371/journal.ppat.1003770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bishop KA, Hickey AC, Khetawat D, Patch JR, Bossart KN, Zhu Z, Wang L-F, Dimitrov DS, Broder CC. 2008. Residues in the stalk domain of the Hendra virus G glycoprotein modulate conformational changes associated with receptor binding. J Virol 82:11398–11409. doi: 10.1128/JVI.02654-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Maar D, Harmon B, Chu D, Schulz B, Aguilar HC, Lee B, Negrete OA. 2012. Cysteines in the stalk of the Nipah virus G glycoprotein are located in a distinct subdomain G for fusion activation. J Virol 86:6632–6642. doi: 10.1128/JVI.00076-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhu Q, Biering SB, Mirza AM, Grasseschi BA, Mahon PJ, Lee B, Aguilar HC, Iorio RM. 2013. Individual N-glycans added at intervals along the stalk of the Nipah virus G protein prevent fusion but do not block the interaction with the homologous F protein. J Virol 87:3119–3129. doi: 10.1128/JVI.03084-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Aguilar HC, Matreyek KA, Filone CM, Hashimi ST, Levroney EL, Negrete OA, Bertolotti-Ciarlet A, Choi DY, McHardy I, Fulcher JA. 2006. N-glycans on Nipah virus fusion protein protect against neutralization but reduce membrane fusion and viral entry. J Virol 80:4878–4889. doi: 10.1128/JVI.80.10.4878-4889.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Aguilar HC, Matreyek KA, Choi DY, Filone CM, Young S, Lee B. 2007. Polybasic KKR motif in the cytoplasmic tail of Nipah virus fusion protein modulates membrane fusion by inside-out signaling. J Virol 81:4520–4532. doi: 10.1128/JVI.02205-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Aguilar HC, Ataman ZA, Aspericueta V, Fang AQ, Stroud M, Negrete OA, Kammerer RA, Lee B. 2009. A novel receptor-induced activation site in the Nipah virus attachment glycoprotein (G) involved in triggering the fusion glycoprotein (F). J Biol Chem 284:1628–1635. doi: 10.1074/jbc.M807469200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kullander K, Klein R. 2002. Mechanisms and functions of Eph and ephrin signalling. Nat Rev Mol Cell Biol 3:475–486. doi: 10.1038/nrm856. [DOI] [PubMed] [Google Scholar]
  • 36.Lu X, Liu Q, Benavides-Montano JA, Nicola AV, Aston DE, Rasco BA, Aguilar HC. 2013. Detection of receptor-induced glycoprotein conformational changes on enveloped virions by using confocal micro-Raman spectroscopy. J Virol 87:3130–3142. doi: 10.1128/JVI.03220-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Colgrave ML, Snelling HJ, Shiell BJ, Feng Y-R, Chan Y-P, Bossart KN, Xu K, Nikolov DB, Broder CC, Michalski WP. 2012. Site occupancy and glycan compositional analysis of two soluble recombinant forms of the attachment glycoprotein of Hendra virus. Glycobiology 22:572–584. doi: 10.1093/glycob/cwr180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yuan P, Leser GP, Demeler B, Lamb RA, Jardetzky TS. 2008. Domain architecture and oligomerization properties of the paramyxovirus PIV 5 hemagglutinin-neuraminidase (HN) protein. Virology 378:282–291. doi: 10.1016/j.virol.2008.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Xu K, Chan Y-P, Rajashankar KR, Khetawat D, Yan L, Kolev MV, Broder CC, Nikolov DB. 2012. New insights into the Hendra virus attachment and entry process from structures of the virus G glycoprotein and its complex with ephrin-B2. PLoS One 7:e48742. doi: 10.1371/journal.pone.0048742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ader N, Brindley M, Avila M, Örvell C, Horvat B, Hiltensperger G, Schneider-Schaulies J, Vandevelde M, Zurbriggen A, Plemper RK, Plattet P. 2013. Mechanism for active membrane fusion triggering by morbillivirus attachment protein. J Virol 87:314–326. doi: 10.1128/JVI.01826-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ader N, Brindley MA, Avila M, Origgi FC, Langedijk JP, C Örvell, Vandevelde M, Zurbriggen A, Plemper RK, Plattet P. 2012. Structural rearrangements of the central region of the morbillivirus attachment protein stalk domain trigger F protein refolding for membrane fusion. J Biol Chem 287:16324–16334. doi: 10.1074/jbc.M112.342493. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES