Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2016 Feb 11;90(5):2306–2315. doi: 10.1128/JVI.02324-15

Interaction of Human Parainfluenza Virus Type 3 Nucleoprotein with Matrix Protein Mediates Internal Viral Protein Assembly

Guangyuan Zhang 1, Yi Zhong 1, Yali Qin 1, Mingzhou Chen 1,
Editor: S Schultz-Cherry
PMCID: PMC4810691  PMID: 26656716

ABSTRACT

Human parainfluenza virus type 3 (HPIV3) belongs to the Paramyxoviridae family. Its three internal viral proteins, the nucleoprotein (N), the phosphoprotein (P), and the polymerase (L), form the ribonucleoprotein (RNP) complex, which encapsidates the viral genome and associates with the matrix protein (M) for virion assembly. We previously showed that the M protein expressed alone is sufficient to assemble and release virus-like particles (VLPs) and a mutant with the L305A point mutation in the M protein (ML305A) has a VLP formation ability similar to that of wild-type M protein. In addition, recombinant HPIV3 (rHPIV3) containing the ML305A mutation (rHPIV3-ML305A) could be successfully recovered. In the present study, we found that the titer of rHPIV3-ML305A was at least 10-fold lower than the titer of rHPIV3. Using VLP incorporation and coimmunoprecipitation assays, we found that VLPs expressing the M protein (M-VLPs) can efficiently incorporate N and P via an N-M or P-M interaction and ML305A-VLPs had an ability to incorporate P via a P-M interaction similar to that of M-VLPs but were unable to incorporate N and no longer interacted with N. Furthermore, we found that the incorporation of P into ML305A-VLPs but not M-VLPs was inhibited in the presence of N. In addition, we provide evidence that the C-terminal region of P is involved in its interaction with both N and M and N binding to the C-terminal region of P inhibits the incorporation of P into ML305A-VLPs. Our findings provide new molecular details to support the idea that the N-M interaction and not the P-M interaction is critical for packaging N and P into infectious viral particles.

IMPORTANCE Human parainfluenza virus type 3 (HPIV3) is a nonsegmented, negative-sense, single-stranded RNA virus that belongs to the Paramyxoviridae family and can cause lower respiratory tract infections in infants and young children as well as elderly or immunocompromised individuals. However, no effective vaccine has been developed or licensed. We used virus-like particle (VLP) incorporation and coimmunoprecipitation assays to determine how the M protein assembles internal viral proteins. We demonstrate that both nucleoprotein (N) and phosphoprotein (P) can incorporate into M-VLPs and N inhibits the M-P interaction via the binding of N to the C terminus of P. We also provide additional evidence that the N-M interaction but not the P-M interaction is critical for the regulation of HPIV3 assembly. Our studies provide a more complete characterization of HPIV3 virion assembly and substantiation that N interaction with M regulates internal viral organization.

INTRODUCTION

Human parainfluenza virus type 3 (HPIV3) is a negative-strand RNA virus (NSV) that belongs to the Paramyxoviridae family and often causes lower respiratory tract infections in infants and young children. The HPIV3 genome consists of 6 open reading frames that encode 6 structural proteins: the nucleoprotein (N), phosphoprotein (P), RNA-dependent RNA polymerase (L), matrix protein (M), and two glycoproteins, hemagglutinin/neuraminidase (HN) and the fusion protein (F). N, P, and L encapsulate the viral RNA to form a helical assembly termed the ribonucleoprotein (RNP) complex, which is the minimum structure required for viral transcription and replication. HN is involved in viral attachment to the host cell, while F is required for fusion with the host cell plasma membrane. The M protein binds directly to the viral envelope. For most NSVs, the M protein is the primary force driving viral assembly, and the budding and formation of virus-like particles (VLPs) are critically dependent on the presence of viral M proteins (16). In some NSVs, M-protein expression alone is sufficient for the formation and release of VLPs, such as the M proteins of human parainfluenza virus type 1 (7), Sendai virus (8), respiratory syncytial virus (RSV) (9), measles virus (10), Nipah virus (5), Newcastle disease virus (4), vesicular stomatitis virus (11), Ebola virus (12), and influenza A virus (13). In contrast, the M proteins of other NSVs, such as mumps virus (6) and parainfluenza virus type 5 (PIV5) (14), require an accessory protein, e.g., F or N, for maximum VLP release efficiency. Previously, we also demonstrated that the M protein of human parainfluenza virus type 3 (HPIV3) alone in mammalian cells could lead to the formation and release of enveloped VLPs (15), which are morphologically similar to virions.

In general, in the process of virion assembly, the M protein links internal viral proteins through interaction with RNPs and envelope glycoproteins via their cytoplasmic tails. However, the mechanism by which RNPs are recruited to budding sites and incorporated into viral particles seems to be different for different NSVs and is not well understood. A recent study showed that, even within the Paramyxoviridae family, the architecture of the various virions is often different (16). This may be due to the differences in RNP assembly into virions. For RSV, a transcription antiterminator, M2-1, mediates the association of RNPs with the M protein and is required for the incorporation of RNPs into virions (17), and further structural analysis showed that M2-1 is located between the RNP and M in isolated viral particles (18). However, for viruses belonging to Paramyxovirinae subfamilies and some other enveloped viruses, such as retroviruses and filoviruses, N has been described to be a mediator of virion assembly and budding (6, 14, 1921). At least two reports suggested that N of influenza virus plays a critical role in virion assembly, possibly through its interaction with M1. One report showed that a temperature-sensitive mutation at N residue 239 results in the production of abnormally shaped virions without affecting virus RNA synthesis (22). Another report showed that three N residues, 214, 217, and 253, which localize at the potential M1 interaction sites on the viral RNP, play a critical role in virion morphology (23).

In addition, interactions between M and the glycoprotein (HN and F) cytoplasmic tails of parainfluenza viruses have been thought to play a critical role in virion assembly at specific locations on plasma membranes (2427), but the contribution of HN and F in virion formation may differ between viruses. For PIV5, the deletion of the HN cytoplasmic tail resulted in a marked defect in viral budding and release, whereas the F cytoplasmic tail was dispensable for normal viral budding (26). In contrast, for Sendai virus, HN was not necessary for virion budding, and the loss of the cytoplasmic tail of F protein resulted in a reduction in the levels of accumulation of M and RNP at the plasma membrane and overall virion production (2830). It is possible that RNP accumulation at assembly sites requires a stable interaction with the M protein at the plasma membrane, which could be provided by the specific interaction of M with the F cytoplasmic tail. So far, no direct evidence has shown that the F cytoplasmic tail is associated with the RNP.

In this study, to further understand the mechanism of HPIV3 assembly and release, we characterized the requirements for internal viral protein assembly using VLP and coimmunoprecipitation assays. Specifically, we sought to determine how N and P are assembled into viral VLPs, whether the P-M interaction is involved in HPIV3 assembly, and how the N-P interaction inhibits the incorporation of P into VLPs expressing the M protein (M-VLPs). Our findings suggest a direct role of N in linking the viral N-P complex to the M protein, thereby promoting internal viral protein incorporation into virions.

MATERIALS AND METHODS

Cells and virus.

Cells (293T and LLC-MK2 [MK2] cells) were maintained in Dulbecco's modified Eagle's medium (Gibco) supplemented with 10% fetal bovine serum (FBS; HyClone). Wild-type HPIV3 (NIH 47885) and recombinant HPIV3 (rHPIV3) expressing the M protein with the L305A point mutation (ML305A) were propagated in MK2 cells by inoculation at a multiplicity of infection (MOI) of 0.1.

Antibodies.

Mouse monoclonal anti-Myc antibody and anti-green fluorescent protein (anti-GFP) antibody were purchased from Santa Cruz Biotechnology. Mouse antihemagglutinin (anti-HA) and anti-Flag monoclonal antibodies were from Sigma. A mouse monoclonal antibody to the anti-HN protein (anti-HN) of parainfluenza virus type 3 was from Abcam.

Plasmid constructs.

The plasmids carrying HA-M, HA-ML305A, HA-P, Myc-M, Myc-ML305A, Myc-N, and N-Flag have been described previously (15). pOCUS-HPIV3 was used as a template for PCR amplification in the genetic manipulations. cDNA encoding mutant NL478A with a Flag epitope tag at the C terminus was cloned into pCAGGS by using site-specific mutagenesis. PCR products encoding P mutants with deletions of 20, 40, 60, 80, or 100 amino acids at the C terminus (PΔC20, PΔC40, PΔC60, PΔC80, and PΔC100, respectively) and an HA tag at the N terminus were also cloned into pCAGGS. The P mutant with a 500-amino-acid deletion at the N terminus (PΔN500) was fused with glutathione S-transferase (GST) and cloned into pCAGGS. The sequences of all the aforementioned constructs were verified via DNA sequencing.

Determination of virus growth curves.

Monolayers of MK2 cells in 6-well plates were grown to 50 to 60% confluence and infected with wild-type HPIV3 or HPIV3 expressing ML305A (HPIV3-ML305A) at an MOI of 0.001. Then, the infection medium was removed and replaced with fresh medium containing 4% FBS, and the supernatant was sampled every 12 h for 4 days prior to determination of the infectious virus titer, which was performed by standard plaque assay on MK2 cells as described previously (15). After the viral titers were calculated, growth curves were determined by using the software GraphPad Prism (version 5.0).

Ultrathin sectioning and transmission electron microscopy.

In 6-well plates, 293T cells were grown to 40 to 50% confluence and transfected with the plasmids indicated below; then, the culture medium was replaced with fresh medium containing 10% FBS at 24 h posttransfection. At 48 h posttransfection, the cells were fixed with 2.5% glutaraldehyde and 4% paraformaldehyde in 0.1 M sodium phosphate buffer (pH 7.4) for 2 h at room temperature. The cells were harvested and fixed with 2.5% glutaraldehyde on ice for 2 h and then fixed in 2% osmium tetroxide. Then, the cells were dehydrated with sequential washes in 50%, 70%, 90%, 95%, and 100% ethanol. Areas containing cells were block mounted and thinly sliced. Final samples were absorbed onto a carbon-coated copper grid negatively stained with 1% phosphotungstic acid (pH 7.0) and then analyzed on a transmission electron microscope.

VLP budding assays.

For the preparation of VLPs to be examined, 293T cells in 6-well plates were grown to 40 to 50% confluence and transfected with a pCAGGS plasmid carrying M, ML305A, N, or P by using calcium phosphate transfection. The plasmid amount was equalized by the amount of empty pCAGGS plasmid. At 48 h posttransfection, the supernatant was harvested and clarified by centrifugation at 13,000 rpm for 3 min and then pelleted through a 20% sucrose cushion and subsequently ultracentrifuged at 35,000 rpm for 2 h at 4°C on a P55 ST2 rotor (Hitachi). The VLPs pelleted at the bottom of the tubes were resuspended in 40 μl cold sodium chloride-Tris-EDTA buffer. Cells were scraped into cold phosphate-buffered saline (PBS), pelleted by centrifugation at 13,000 rpm for 1 min, lysed in cold TNE buffer (50 mM Tris-HCl [pH 7.4], 150 mM NaCl, 2 mM EDTA [pH 8.0], 0.1% 2-mercaptoethanol, protease inhibitor cocktail) for 30 min on ice, and then centrifuged at 13,000 rpm for 30 min at 4°C. VLP pellets and cell lysates were mixed with 5× sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) protein-loading buffer (250 mM Tris-HCl [pH 6.8], 10% SDS, 50% glycerol, 0.3% bromophenol blue, 2.5% 2-mercaptoethanol), incubated at 100°C for 10 min, and subjected to Western blot analysis.

Western blot analysis.

The prepared samples were separated by 10% or 12% SDS-PAGE and then electroblotted onto a nitrocellulose membrane. The membranes were blocked with skim milk in phosphate-buffered saline with Tween 20 (1/1,000 Tween 20) for 30 min at room temperature and subsequently incubated with primary antibodies for 1 h and secondary antibodies for 1 h. The primary antibodies used were as follows: anti-Myc (1:2,500), anti-HA (1:10,000), anti-GFP (1:2,500), anti-Flag (1:10,000), and anti-HN (1:2,500). The secondary antibodies, used at a 1:5,000 dilution, were goat anti-rabbit IgG and goat anti-mouse IgG.

Protease protection assay.

The aforementioned prepared VLPs were divided into four aliquots and subjected to the following treatments: (i) no treatment, (ii) treatment with Triton X-100 to a final concentration of 1%, (iii) treatment with tosylsulfonyl phenylalanyl chloromethyl ketone-treated trypsin (Sigma-Aldrich) to a final concentration of 1 μg/ml, or (iv) treatment with Triton X-100 plus trypsin. All the samples were incubated at 37°C for 30 min and then mixed with 5× SDS-PAGE protein-loading buffer, incubated at 100°C for 10 min, and subjected to Western blot analysis.

Coimmunoprecipitation assay.

In 6-well plates, 293T cells were grown to 40 to 50% confluence and transfected with the indicated plasmids. At 48 h posttransfection, cell lysates were prepared in 350 μl TNE buffer as described above in “VLP budding assays”; 40 μl lysates was removed for input analysis, and the remaining cell lysates were then incubated with relevant antibodies (anti-c-Myc tag affinity gel [BioLegend] or anti-Flag affinity gel [Sigma]) overnight at 4°C with gentle rotation. The beads were collected by centrifugation at 5,000 rpm for 2 min at 4°C and washed 3 times with TNE buffer. The beads were then mixed with 2× SDS-PAGE loading buffer, incubated at 100°C for 10 min, and subjected to Western blot analysis.

Immunofluorescence analysis.

HeLa cells grown on coverslips in 12-well plates were transfected with pCAGGS-N-Flag or pCAGGS-HA-P alone or together with pCAGGS-Myc-M or pCAGGS-Myc-ML305A using the Lipofectamine 2000 reagent (Invitrogen). At 24 h posttransfection, the cells were washed three times with cold PBS, fixed with 4% paraformaldehyde for 20 min, permeabilized with 0.2% Triton X-100 for 20 min, and blocked by 3% bovine serum albumin (BSA) for 30 min, and then the cells were incubated with rabbit polyclonal anti-Myc antibody (1:200; Santa Cruz), mouse monoclonal anti-HA antibody (1:2,000; Sigma), or mouse monoclonal anti-Flag antibody (1:2,000) for 1 h. After these incubations, the cells were washed three times with 1% BSA and incubated with goat anti-rabbit IgG rhodamine (1:100; Thermo) or goat anti-mouse IgG fluorescein (1:200; Thermo) secondary antibody for 1 h. The cells were then washed three times with cold PBS and stained with DAPI (4′,6-diamidino-2-phenylindole) for 5 min. Confocal images were collected to visualize the location of N-M and N-ML305A or P-M and P-ML305A.

RESULTS

Recombinant HPIV3-ML305A shows lower titers than wild-type HPIV3.

Our previous study showed that expression of the M protein alone is sufficient to form and release VLPs, a mutant with a point mutation of the HPIV3 M protein (ML302A) is deficient in VLP production, and recombinant HPIV3 expressing ML302A could not be rescued; in contrast, a mutant with another point mutation of the HPIV3 M protein (ML305A) could release VLPs as efficiently as wild-type M and recombinant HPIV3 expressing ML305A could be readily rescued (15). However, to our surprise, when we performed a growth curve assay with HPIV3-ML305A and compared its growth to that of wild-type HPIV3, HPIV3-ML305A showed markedly slower replication kinetics in MK2 cells, with virus titers being up to 12-fold lower for HPIV3-ML305A than for wild-type HPIV3 (Fig. 1A), although HN protein expression in the cell lysates at different time points appeared to be comparable between wild-type HPIV3 and HPIV3-ML305A (Fig. 1B).

FIG 1.

FIG 1

Recombinant HPIV3 expressing ML305A showed lower titers than wild-type HPIV3, but ML305A was still able to form authentic VLPs. (A) Growth curve of the recombinant wild-type (WT) HPIV3 and HPIV3-ML305A. MK2 cells were infected as described in Materials and Methods, and the supernatant was harvested every 12 h (as indicated on the x axis) for the plaque assay to determine virus growth curves. Standard errors were calculated from two independent experiments. (B) HN expression from cell lysates infected with recombinant wild-type HPIV3 and HPIV3-ML305A. MK2 cells were infected as described in Materials and Methods, and the cells was pelleted every 12 h (as indicated by the numbers beneath the lanes) for the HN expression assay. (C) Representative transmission electron microscopy images of ultrathin sections of 293T cells. 293T cells were transfected with M, ML305A, or ML302A. After 48 h, the cells were treated as described in Materials and Methods and then visualized by transmission electron microscopy. Arrows, the VLPs outside the sliced cells. (D) Protease protection assay for M- and ML305A-VLPs. The indicated purified VLP samples were treated as described in Materials and Methods and then analyzed via Western blotting using anti-HA antibody. WB, Western blotting; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

To determine whether the nature of the VLPs formed by ML305A is indeed the same as that of the VLPs formed by wild-type M protein, we analyzed the morphology of VLPs by using ultrathin sectioning and transmission electron microscopy and found that the VLPs released by ML305A were morphologically similar to those released by wild-type M protein (Fig. 1C). Furthermore, a protease protection assay showed that VLPs released by ML305A were also enclosed by a lipid bilayer because it could be digested only when treated with trypsin plus Triton X-100 (Fig. 1D, lane 4). These data suggest that ML305A alone is indeed fully functional in producing authentic VLPs, as wild-type M protein and the decrease in the HPIV3-ML305A titer might be caused during the virion assembly and budding process.

The M protein can specifically incorporate N or P into M-VLPs.

Since the M protein of paramyxoviruses is the primary driving force of the virion assembly process and plays a linking role by interacting with both viral RNPs and glycoproteins (31) and earlier studies showed that the incorporation of RNPs into viral particles is determined by the interaction of the M protein with the RNPs (32), we first sought to determine whether N or P could be incorporated into M-VLPs. To this end, we coexpressed N or P with the M protein in 293T cells and performed the VLP budding assay. We also included GFP as a negative control since GFP does not interact with M and exists only in lysates and not in the pellet fraction of the culture medium if lysis of cells does not occur. When expressed alone, neither N nor P was detected in the pellet fraction of the culture medium (Fig. 2A and B, top), but N and P could be detected in the pellet fraction of the culture medium when coexpressed with M, whereas GFP could not be detected (Fig. 2A and B, bottom, lanes 4), in spite of its expression in lysates, suggesting that N and P in the pellet fraction of the culture medium were not released by lysis of cells and the M protein could specifically incorporate the N or P protein into VLPs. To confirm that N or P was indeed incorporated into M-VLPs, we performed a protease protection assay, and the fractions from either N-M-transfected or P-M-transfected cell medium were treated as described in the legend to Fig. 1D. The results showed that N and P together with the M protein were all protected from proteolysis by trypsin treatment alone (Fig. 2C and D, lanes 3), but the addition of Triton X-100 disrupted the lipid envelope of VLPs, resulting in the degradation of the N, P, and M proteins in the presence of trypsin (Fig. 2C and D). These data suggest that N or P is specifically incorporated into the M-VLPs.

FIG 2.

FIG 2

M could specifically incorporate N or P into M-VLPs. (A, B) VLP budding assays for the cotransfection of N and M (A) or P and M (B); GFP was included as a negative control. 293T cells were transfected with the indicated plasmids for 48 h. The cell lysates and VLPs were prepared as described in Materials and Methods, and then the samples were subjected to Western blot analysis by using anti-Myc, anti-Flag, anti-HA, or anti-GFP antibodies. Arrows, the bands of the detected proteins. (C, D) Protease protection assays for the N-M and P-M VLPs. 293T cells were cotransfected with either N plus M (C) or P plus M (D). The VLP samples in the supernatant were divided into four aliquots, treated as described in Materials and Methods, and then analyzed via Western blot analysis with anti-Flag, anti-HA, or anti-Myc antibodies. Arrows, the bands of the detected proteins.

ML305A can incorporate P but is deficient in incorporating N into ML305A-VLPs.

Next, we sought to determine whether ML305A could also incorporate N or P as efficiently as wild-type M protein. To this end, we coexpressed N or P with ML305A in 293T cells and performed the VLP assay as described in the legends to Fig. 2A and B. The results showed that when N was coexpressed with the M protein, N could be detected in the VLPs (Fig. 3A, bottom right, lane 2), which is consistent with the results in Fig. 2A; conversely, when coexpressed with ML305A, N could barely be detected in VLPs (Fig. 3A, bottom right, lane 3), even though ML305A had a VLP formation ability similar to that of wild-type M protein. However, when coexpressed with either M or ML305A, P was readily detected in both M- and ML305A-VLPs at a similar intensity (Fig. 3B, bottom right, lanes 2 and 3). We also assessed the ability of the M protein and ML305A to incorporate HN into VLPs, and the results showed that both the M protein and ML305A can incorporate the HN protein into VLPs at the same level (Fig. 3C). Furthermore, we found that ML305A also has an oligomerization ability similar to that of M in a coimmunoprecipitation assay (Fig. 3D). When these data are taken together, these results suggest that ML305A maintained the ability to effectively incorporate P into VLPs, but its ability to incorporate N into VLPs was severely weakened.

FIG 3.

FIG 3

ML305A is able to incorporate P but not N into VLPs. (A to C) VLP budding assays for the cotransfection of N and M/ML305A (A), P and M/ML305A (B), or HN and M/ML305A (C). 293T cells were transfected with HN and M/ML305A. At 48 h posttransfection, VLP budding assays were performed as described in the legend to Fig. 2. (D) ML305A interacts with both M and itself. 293T cells were transfected with plasmids carrying the indicated proteins. At 48 h posttransfection, immunoprecipitation (IP) assays were performed with anti-Myc antibodies. Western blotting was performed with anti-Myc, anti-HA, and anti-HN antibodies. Arrows, the bands of the detected proteins.

ML305A maintained its interaction with P but not with N.

To determine whether the inability of ML305A to incorporate N into VLPs is caused by the lack of interaction between ML305A and N, we examined the interaction of N with M or ML305A by performing a coimmunoprecipitation assay. A plasmid carrying Myc-tagged M or ML305A was transfected either individually or jointly with a plasmid carrying N-Flag into 293T cells, immunoprecipitation was performed by using an anti-Flag affinity gel, and Western blotting was carried out with anti-Myc and anti-Flag monoclonal antibodies. Wild-type M protein was efficiently coimmunoprecipitated with N-Flag, whereas the coimmunoprecipitation ability of ML305A was severely weakened (Fig. 4A, top, lanes 2 and 4); furthermore, immunofluorescence results also showed that the colocalization ability of ML305A with N was dramatically decreased (Fig. 4B), suggesting that M interacts with N, but the ML305A interaction with N was severely weakened. Next, the interaction of HA-P with Myc-tagged M or ML305A was also examined via coimmunoprecipitation assay using an anti-Myc affinity gel, and the results showed that HA-P was coimmunoprecipitated either by Myc-M or by Myc-ML305A at similar levels (Fig. 4C, top, lanes 2 and 3), and immunofluorescence assay results also showed that both M and ML305A similarly colocalized with P (Fig. 4D), suggesting that both M and ML305A can interact with P. As expected, the ability of M or ML305A to interact with N and P correlated with their ability to incorporate N and P into VLPs. So far, we can conclude that the lower titers of HPIV3-ML305A were caused by the inability of ML305A to interact with N and to incorporate N into viral particles.

FIG 4.

FIG 4

ML305A maintained its interaction with P but not N. (A, C) Coimmunoprecipitation assays for N and M/ML305A (A) or P and M/ML305A (C). 293T cells were transfected with plasmids carrying the indicated proteins. At 48 h posttransfection, the cells were harvested and subjected to a coimmunoprecipitation assay as described in Materials and Methods. (A) Immunoprecipitation was performed using an anti-Flag affinity gel, and lysates were detected via Western blotting using anti-Myc and anti-Flag monoclonal antibodies. (C) Immunoprecipitation was performed using an anti-Myc affinity gel, and lysates were detected via Western blotting using anti-HA and anti-Myc monoclonal antibodies. Arrows, the bands of the detected proteins. (B, D) Colocalization assays for N and M/ML305A (B) or P and M/ML305A (D). HeLa cells were transfected with plasmids carrying the indicated proteins. At 24 h posttransfection, cells were fixed and stained with anti-Flag, anti-Myc, and anti-HA antibodies and visualized via confocal microscopy.

N inhibits the incorporation of P into ML305A-VLPs but not M-VLPs.

Having found that wild-type M-VLPs could incorporate either N or P, whereas ML305A-VLPs incorporated P but not N, we next sought to determine what would happen when the N, P, and M proteins were expressed together. Both N and P were efficiently incorporated into M-VLPs when N, P, and M were expressed together (Fig. 5A, right, lane 4). When N, P, and ML305A were coexpressed, not only was it found that N could not be incorporated into ML305A-VLPs, but also the incorporation of P into ML305A-VLPs was found to be severely inhibited, despite the efficient formation and release of ML305A-VLPs (Fig. 5, right, lane 5), which prompted a key question: how does N inhibit the incorporation of P into ML305A-VLPs but not into M-VLPs? To answer this question, we used a point mutant of N (NL478A), which was unable to interact with P in our previous study (33), to further examine whether NL478A also inhibits the incorporation of P into ML305A-VLPs. N or NL478A was coexpressed with P and ML305A. We found that NL478A was unable to inhibit the incorporation of P into ML305A-VLPs (Fig. 5, middle right, lane 7). Of note, similar to the findings for N, NL478A could still be incorporated into M-VLPs other than those induced by ML305A (Fig. 5, bottom right, lanes 6 and 7). Taken together, these data lead to the following conclusions: (i) N inhibits the incorporation of P into ML305A-VLPs but not M-VLPs; (ii) NL478A is unable to interact with P, and therefore, NL478A is also unable to inhibit the incorporation of P into ML305A-VLPs; and (iii) M assembles the N-P complex into viral particles via an N-M interaction but not an P-M interaction because the N-P interaction inhibits the P-M interaction.

FIG 5.

FIG 5

N but not NL478A inhibits the incorporation of P into ML305A. The results of VLP budding assays for the cotransfection of N or NL478A and P and of N or NL478A and M/ML305A are shown. 293T cells were transfected with plasmids carrying the indicated proteins for 48 h. The VLP budding assays were performed as described in the legend to Fig. 2. Then, samples were analyzed via Western blotting with anti-HA, anti-Myc, and anti-Flag antibodies. Arrows, the bands of the detected proteins.

Both M and N interact with the C terminus of P.

Having established that the N-P interaction inhibits the P-ML305A interaction, we sought to identify the functional domains within P that are critical for the incorporation of P into M-VLPs and to determine whether N interacts with P and inhibits the incorporation of P into ML305A-VLPs via this domain. To this end, we constructed a series of mutants with mutations in the C terminus of P, coexpressed each of the mutants with M in 293T cells, and performed VLP budding assays. The PΔC20, PΔC40, PΔC60, and PΔC80 mutants with deletions at the C terminus of P could be incorporated into M-VLPs as efficiently as wild-type P (Fig. 6A, bottom right, lanes 3 to 6); however, the PΔC100 mutant was barely detected in the M-VLPs (Fig. 6A, bottom right, lane 7), suggesting that the residues between residues 80 and 100 of the C terminus of P are critical for its interaction with M and for the incorporation of P into M-VLPs. To exclude the possibility that the failure to incorporate PΔC100 into M-VLPs was due to a conformational alteration of P, we took advantage of another construct, PΔN500, which expresses only 102 residues of the C terminus of P. Since the PΔN500 mutant showed very poor expression of P and GST-fused PΔN500 dramatically enhanced its expression, we used this construct for VLP assays, and the results showed that GST-PΔN500 could be specifically incorporated into M-VLPs as efficiently as wild-type P (Fig. 6B, bottom right, lanes 2 and 6), whereas GST alone could not be incorporated into M-VLPs (Fig. 6B, bottom right, lane 4). Next, by using coimmunoprecipitation assays, we examined the interaction of GST-PΔN500 with M or ML305A. The results showed that GST-PΔN500 was coimmunoprecipitated by both Myc-M and Myc-ML305A (Fig. 6C, top right, and D, top, lanes 4), whereas GST alone was not coimmunoprecipitated by Myc-M or Myc-ML305A (Fig. 6C, top right, and D, top, lanes 2), suggesting that the C terminus of P is indispensable and sufficient to interact with the M protein and to incorporate P into M-VLPs.

FIG 6.

FIG 6

Both ML305A and N can interact with the 100 residues in the C terminus of P. (A) VLP budding assays for the cotransfection of M and mutant P. 293T cells were transfected with plasmids carrying the indicated proteins. At 48 h posttransfection, VLP budding assays were performed as described in the legend to Fig. 2. Then, samples were analyzed via Western blotting with anti-Myc and anti-HA antibodies. (B) VLP budding assays for the cotransfection of M and PΔN500. Plasmids carrying the indicated proteins were transfected into 293T cells for 48 h. VLP budding assays were performed as described in the legend to Fig. 2. Then, samples were analyzed via Western blotting with anti-Myc and anti-HA antibodies. (C, D) Coimmunoprecipitation assays for M and PΔN500 (C) or ML305A and PΔN500 (D). 293T cells were transfected with the plasmids carrying the indicated proteins. At 48 h posttransfection, the cells were processed as described in the legend to Fig. 4, immunoprecipitation was performed using an anti-Myc affinity gel, and lysates were detected via Western blotting using anti-HA and anti-Myc monoclonal antibodies. (E) Coimmunoprecipitation assays for N and PΔN500. Cells were transfected and processed as described in the legend to panel C, except that immunoprecipitation was performed using an anti-Flag affinity gel and lysates were detected via Western blotting using anti-HA and anti-Flag monoclonal antibodies. Arrows, the bands of the detected proteins.

Next, we sought to determine whether N interacts with P via the C terminus of P; thus, we assessed the interaction between GST-PΔN500 and N. GST-PΔN500 was expressed alone or together with N-Flag in 293T cells, and immunoprecipitation was performed by using an anti-Flag affinity gel. The results revealed that, as a positive control, P interacted with N (Fig. 6E, top, lane 2), as did GST-PΔN500 (Fig. 6E, top, lane 6), whereas GST alone failed to interact with N-Flag (Fig. 6E, top, lane 4). Taken together, our results show that both N and M interact with P via the C terminus of P.

N inhibits incorporation of PΔN500 into ML305A-VLPs, while NL478A does not.

Having confirmed that both N and M interact with P via the C terminus of P and that N inhibits the incorporation of P into ML305A-VLPs, we sought to determine whether N also inhibits the incorporation of GST-PΔN500 into ML305A-VLPs. GST-PΔN500 was coexpressed with ML305A or M with or without N, and VLP budding assays were performed. GST-PΔN500 was readily detected in both ML305A- and M-VLPs (Fig. 7A, top right, lanes 2 and 3) in the absence of N, whereas when N was present, GST-PΔN500 was unable to be incorporated into ML305A-VLPs but could still be incorporated into M-VLPs (Fig. 7A, top right, lanes 4 and 5). As expected, NL478A lost the ability to inhibit the incorporation of GST-PΔN500 into ML305A-VLPs (Fig. 7B, top right, lane 5). Taken together, these data show that N but not NL478A indeed inhibits the incorporation of P into ML305A-VLPs via interaction with the C terminus of P.

FIG 7.

FIG 7

N but not NL478A inhibits the incorporation of ML305A into PΔN500. (A) VLP budding assays for the cotransfection of N, PΔN500, and M/ML305A. (B) VLP budding assays for the cotransfection of NL478A, PΔN500, and M/ML305A. 293T cells were transfected with plasmids carrying the indicated proteins for 48 h. The VLP budding assays were performed as described in the legend to Fig. 2, and then samples were analyzed via Western blotting with anti-HA, anti-Flag, and anti-Myc antibodies. Arrows, the bands of the detected proteins.

DISCUSSION

In this study, we found that recombinant HPIV3-ML305A shows lower titers than wild-type HPIV3 (Fig. 1A). Subsequently, we found that the levels of expression of the M protein and ML305A and release of VLPs are similar (Fig. 1C and D). Therefore, we hypothesized that ML305A has an effect on virion assembly. To prove this, we first demonstrated that the M protein of HPIV3 interacts with both N and P and can incorporate both N and P into M-VLPs (Fig. 2 and 4). Because N also interacts with P to form the N-P complex (33), it is unclear whether N or P mediates the interaction of the N-P complex with M in the virion assembly process. Since ML305A might be assembly defective, we sought to determine whether ML305A is defective in its interaction with N or P, or both. Our subsequent results showed that ML305A still interacted with P and could incorporate P into ML305A-VLPs but was unable to interact with N and incorporate N into ML305A-VLPs (Fig. 3A and B). The finding that ML305A interacts with P but fails to interact with N provides a rationale for assessing how the M protein assembles internal viral proteins into particles. On the basis of the result that ML305A interacts with P and incorporates P but not N into ML305A-VLPs, it is reasonable to speculate that one of two scenarios occurs when N, P, and ML305A are expressed together: (i) if P mediates the N-P complex interaction with the M protein, N is also incorporated into ML305A-VLPs, which suggests that P is a key mediator of the incorporation of the N-P complex into viral particles; (ii) in contrast, if N mediates the N-P complex interaction with the M protein, neither N nor P is incorporated into ML305A-VLPs, which suggests that N is a key mediator of the incorporation of the N-P complex into viral particles. Our results clearly demonstrate that N inhibits the incorporation of P into ML305A-VLPs via the formation of the N-P complex, but NL478, a point mutant of N which lost the ability to interact with P for the formation of the N-P complex (33), has no effect on the incorporation of P into ML305A-VLPs (Fig. 5), suggesting that N is the critical mediator of the incorporation of the N-P complex into viral particles.

The results from the aforementioned experiments prompted an interesting question: how does the N-P interaction inhibit the incorporation of P into ML305A-VLPs? We used mutational analysis to identify regions in P that are relevant for the incorporation of P into M-VLPs and the P-M interaction. Our data first showed that the PΔC100 mutant abrogates the incorporation of P into M-VLPs (Fig. 6A). Then, we demonstrated that the expression of GST-PΔN500 is efficient for both the interaction between GST-PΔN500 and M and the recruitment of PΔN500 into M-VLPs (Fig. 6B and C). Interestingly, PΔN500 is also required and sufficient for the interaction of P with N (Fig. 6E). N inhibited the incorporation of not only P but also GST-GST-PΔN500 into ML305A-VLPs (Fig. 7A). One possibility is that N binds to PΔN500 with a higher infinity and competitively prevents M from binding to PΔN500, but we still cannot exclude the possibility that the binding of N to PΔN500 results in structural changes in P or PΔN500 so that it can no longer be recognized by the M protein. It seems impossible that the formation of the N-P complex would result in structural changes in N since the N-P complex can be incorporated into M-VLPs (Fig. 5, right, lane 4).

In general, the finding that N regulates N-P complex assembly via direct interaction with a single amino acid residue in the M protein is remarkable for the study of the enveloped virion assembly process. Despite advancements in our understanding of viral particle assembly, the factors that drive the incorporation of viral RNPs into virions are still incompletely defined, and N has been shown to be indirectly involved in the virion assembly process for several enveloped viruses. For example, Ebola virus VLP formation is driven by VP40 but is significantly enhanced by the presence of N (21), and the N of Marburg virus was found to increase VP40-induced VLP release by recruiting Tsg101 via its PSAP motif (20), possibly to compensate for the absence of a PT/SAP motif in VP40 itself. In our study, although the expression of N had no effect on the formation and release of M-VLPs (data not shown), it could be incorporated into M-VLPs and mediate virion assembly.

Although we found that the interaction of N and the M protein plays a critical role in N-P complex recruitment to viral particles, resulting in the incorporation of internal viral proteins into the virions, we still cannot exclude the possibility that a cellular protein(s) is also involved in this assembly process by interacting with N or M, or both. Recently, a role for the viral N, in cooperation with the M proteins and the ESCRT pathway, has been suggested for viral particle formation of Marburg virus, HIV-1, and Mopeia virus (20, 34, 35). Therefore, additional cellular factors may associate with N through residue L305 in the M protein for efficient N-P complex incorporation into virions. A more detailed analysis of the association of N and the M protein with the components of the ESCRT pathway and its associated proteins will potentially reveal the role of such cellular determinants in N-P complex assembly. In addition, the M proteins of several NSVs, including influenza virus and human RSV, have been reported to interact directly with either RNA or N (36, 37). Further study is required to determine whether the M protein also packages the N-P complex into viral particles through its interaction with viral RNA.

In addition, the domain within N that mediates the interaction of N with the M protein remains unconfirmed. Previous studies have suggested that the C terminus of N of paramyxoviruses may be involved in its binding to P and the M protein (3840). Whether the HPIV3 C-terminal domain interacts with both P and the M protein is not clear. Since NL478A, which fails to bind to P, can still be incorporated into M-VLPs (Fig. 5B, bottom right, lane 6), it seems that N interacts with both P and M via different regions. Further investigations into essential regions within N that regulate the N-M interaction might provide important insights into the mechanism of the incorporation of HPIV3 internal proteins into virions.

Of note, although the N-P interaction inhibits the P-M interaction and N is a critical regulator of the interaction of the N-P complex with M, the possibility that P plays an indirect role in the correct assembly of virions cannot be excluded. By using cryo-electron microscopy and tomography to visualize Ebola VLPs, which were produced with different combinations of viral proteins, Bharat et al. found that when N was coexpressed with VP40, VP40 could condense a loosely coiled helix formed by the binding of N to RNA. When N was coexpressed with VP40, VP24, and VP35, the helix was further rigidified and the helix inside the VLPs was indistinguishable from that in Ebola virus virions (41). Therefore, for HPIV3, it is also possible that the loose helix formed by the binding of N to RNA is condensed by the binding of M to N, which further requires the rigidification of the condensed coils into a tight helix by the binding of P to N. Therefore, it is reasonable to speculate that VLPs formed by N, P, and M are much more compact than those formed by N and M.

In summary, we have demonstrated that the interaction of N but not P with the M protein of HPIV3 mediates internal viral protein assembly and a critical residue, L305, in the M protein is required for the N-M interaction. Our results will strengthen the understanding of the mechanisms that drive HPIV3 internal viral protein assembly and could lead to the development of novel targeted antiviral drugs for therapeutic use against HPIV3 infection.

ACKNOWLEDGMENTS

This work was supported by grants from the China Natural Science Foundation (grant 81471939 and 81271816) and the Major State Basic Research Development Program (973 Program) (2012CB518906).

REFERENCES

  • 1.Schmitt AP, Lamb RA. 2004. Escaping from the cell: assembly and budding of negative-strand RNA viruses. Curr Top Microbiol Immunol 283:145–196. [DOI] [PubMed] [Google Scholar]
  • 2.Shnyrova AV, Ayllon J, Mikhalyov II, Villar E, Zimmerberg J, Frolov VA. 2007. Vesicle formation by self-assembly of membrane-bound matrix proteins into a fluidlike budding domain. J Cell Biol 179:627–633. doi: 10.1083/jcb.200705062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ciancanelli MJ, Basler CF. 2006. Mutation of YMYL in the Nipah virus matrix protein abrogates budding and alters subcellular localization. J Virol 80:12070–12078. doi: 10.1128/JVI.01743-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pantua HD, McGinnes LW, Peeples ME, Morrison TG. 2006. Requirements for the assembly and release of Newcastle disease virus-like particles. J Virol 80:11062–11073. doi: 10.1128/JVI.00726-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Patch JR, Crameri G, Wang LF, Eaton BT, Broder CC. 2007. Quantitative analysis of Nipah virus proteins released as virus like particles reveals central role for the matrix protein. Virol J 4:1. doi: 10.1186/1743-422X-4-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li M, Schmitt PT, Li Z, McCrory TS, He B, Schmitt AP. 2009. Mumps virus matrix, fusion, and nucleocapsid proteins cooperate for efficient production of virus-like particles. J Virol 83:7261–7272. doi: 10.1128/JVI.00421-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Coronel EC, Murti KG, Takimoto T, Portner A. 1999. Human parainfluenza virus type 1 matrix and nucleoprotein genes transiently expressed in mammalian cells induce the release of virus-like particles containing nucleocapsid-like structures. J Virol 73:7035–7038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sugahara F, Uchiyama T, Watanabe H, Shimazu Y, Kuwayama M, Fujii Y, Kiyotani K, Adachi A, Kohno N, Yoshida T, Sakaguchi T. 2004. Paramyxovirus Sendai virus-like particle formation by expression of multiple viral proteins and acceleration of its release by C protein. Virology 325:1–10. doi: 10.1016/j.virol.2004.04.019. [DOI] [PubMed] [Google Scholar]
  • 9.Baviskar PS, Hotard AL, Moore ML, Oomens AG. 2013. The respiratory syncytial virus fusion protein targets to the perimeter of inclusion bodies and facilitates filament formation by a cytoplasmic tail-dependent mechanism. J Virol 87:10730–10741. doi: 10.1128/JVI.03086-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Runkler N, Pohl C, Schneider-Schaulies S, Klenk HD, Maisner A. 2007. Measles virus nucleocapsid transport to the plasma membrane requires stable expression and surface accumulation of the viral matrix protein. Cell Microbiol 9:1203–1214. doi: 10.1111/j.1462-5822.2006.00860.x. [DOI] [PubMed] [Google Scholar]
  • 11.Harty RN, Paragas J, Sudol M, Palese P. 1999. A proline-rich motif within the matrix protein of vesicular stomatitis virus and rabies virus interacts with WW domains of cellular proteins: implications for viral budding. J Virol 73:2921–2929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Timmins J, Scianimanico S, Schoehn G, Weissenhorn W. 2001. Vesicular release of Ebola virus matrix protein VP40. Virology 283:1–6. doi: 10.1006/viro.2001.0860. [DOI] [PubMed] [Google Scholar]
  • 13.Parent LJ, Bennett RP, Craven RC, Nelle TD, Krishna NK, Bowzard JB, Wilson CB, Puffer BA, Montelaro RC, Wills JW. 1995. Positionally independent and exchangeable late budding functions of the Rous sarcoma virus and human immunodeficiency virus Gag proteins. J Virol 69:5455–5460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schmitt AP, Leser GP, Waning DL, Lamb RA. 2002. Requirements for budding of paramyxovirus simian virus 5 virus-like particles. J Virol 76:3952–3964. doi: 10.1128/JVI.76.8.3952-3964.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhang G, Zhang S, Ding B, Yang X, Chen L, Yan Q, Jiang Y, Zhong Y, Chen M. 2014. A leucine residue in the C terminus of human parainfluenza virus type 3 matrix protein is essential for efficient virus-like particle and virion release. J Virol 88:13173–13188. doi: 10.1128/JVI.01485-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Liljeroos L, Krzyzaniak MA, Helenius A, Butcher SJ. 2013. Architecture of respiratory syncytial virus revealed by electron cryotomography. Proc Natl Acad Sci U S A 110:11133–11138. doi: 10.1073/pnas.1309070110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Li D, Jans DA, Bardin PG, Meanger J, Mills J, Ghildyal R. 2008. Association of respiratory syncytial virus M protein with viral nucleocapsids is mediated by the M2-1 protein. J Virol 82:8863–8870. doi: 10.1128/JVI.00343-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kiss G, Holl JM, Williams GM, Alonas E, Vanover D, Lifland AW, Gudheti M, Guerrero-Ferreira RC, Nair V, Yi H, Graham BS, Santangelo PJ, Wright ER. 2014. Structural analysis of respiratory syncytial virus reveals the position of M2-1 between the matrix protein and the ribonucleoprotein complex. J Virol 88:7602–7617. doi: 10.1128/JVI.00256-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bello NF, Dussupt V, Sette P, Rudd V, Nagashima K, Bibollet-Ruche F, Chen C, Montelaro RC, Hahn BH, Bouamr F. 2012. Budding of retroviruses utilizing divergent l domains requires nucleocapsid. J Virol 86:4182–4193. doi: 10.1128/JVI.07105-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dolnik O, Kolesnikova L, Stevermann L, Becker S. 2010. Tsg101 is recruited by a late domain of the nucleocapsid protein to support budding of Marburg virus-like particles. J Virol 84:7847–7856. doi: 10.1128/JVI.00476-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Licata JM, Johnson RF, Han Z, Harty RN. 2004. Contribution of Ebola virus glycoprotein, nucleoprotein, and VP24 to budding of VP40 virus-like particles. J Virol 78:7344–7351. doi: 10.1128/JVI.78.14.7344-7351.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Noton SL, Simpson-Holley M, Medcalf E, Wise HM, Hutchinson EC, McCauley JW, Digard P. 2009. Studies of an influenza A virus temperature sensitive mutant identify a late role for NP in the formation of infectious virions. J Virol 83:562–571. doi: 10.1128/JVI.01424-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bialas KM, Bussey KA, Stone RL, Takimoto T. 2014. Specific nucleoprotein residues affect influenza virus morphology. J Virol 88:2227–2234. doi: 10.1128/JVI.03354-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schmitt AP, He B, Lamb RA. 1999. Involvement of the cytoplasmic domain of the hemagglutinin-neuraminidase protein in assembly of the paramyxovirus simian virus 5. J Virol 73:8703–8712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Schmitt AP, Leser GP, Morita E, Sundquist WI, Lamb RA. 2005. Evidence for a new viral late-domain core sequence, FPIV, necessary for budding of a paramyxovirus. J Virol 79:2988–2997. doi: 10.1128/JVI.79.5.2988-2997.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Waning DL, Schmitt AP, Leser GP, Lamb RA. 2002. Roles for the cytoplasmic tails of the fusion and hemagglutinin-neuraminidase proteins in budding of the paramyxovirus simian virus 5. J Virol 76:9284–9297. doi: 10.1128/JVI.76.18.9284-9297.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ali A, Nayak DP. 2000. Assembly of Sendai virus: M protein interacts with F and HN proteins and with the cytoplasmic tail and transmembrane domain of F protein. Virology 276:289–303. doi: 10.1006/viro.2000.0556. [DOI] [PubMed] [Google Scholar]
  • 28.Stricker R, Roux L. 1991. The major glycoprotein of Sendai virus is dispensable for efficient virus particle budding. J Gen Virol 72:1703–1707. doi: 10.1099/0022-1317-72-7-1703. [DOI] [PubMed] [Google Scholar]
  • 29.Fouillot-Coriou N, Roux L. 2000. Structure-function analysis of the Sendai virus F and HN cytoplasmic domain: different role for the two proteins in the production of virus particle. Virology 270:464–475. doi: 10.1006/viro.2000.0291. [DOI] [PubMed] [Google Scholar]
  • 30.Stone R, Takimoto T. 2013. Critical role of the fusion protein cytoplasmic tail sequence in parainfluenza virus assembly. PLoS One 8:e61281. doi: 10.1371/journal.pone.0061281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Harrison MS, Sakaguchi T, Schmitt AP. 2010. Paramyxovirus assembly and budding: building particles that transmit infections. Int J Biochem Cell Biol 42:1416–1429. doi: 10.1016/j.biocel.2010.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Coronel EC, Takimoto T, Murti KG, Varich N, Portner A. 2001. Nucleocapsid incorporation into parainfluenza virus is regulated by specific interaction with matrix protein. J Virol 75:1117–1123. doi: 10.1128/JVI.75.3.1117-1123.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhang S, Chen L, Zhang G, Yan Q, Yang X, Ding B, Tang Q, Sun S, Hu Z, Chen M. 2013. An amino acid of human parainfluenza virus type 3 nucleoprotein is critical for template function and cytoplasmic inclusion body formation. J Virol 87:12457–12470. doi: 10.1128/JVI.01565-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dussupt V, Javid MP, Abou-Jaoudé G, Jadwin JA, de La Cruz J, Nagashima K, Bouamr F. 2009. The nucleocapsid region of HIV-1 Gag cooperates with the PTAP and LYPXnL late domains to recruit the cellular machinery necessary for viral budding. PLoS Pathog 5:e1000339. doi: 10.1371/journal.ppat.1000339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Shtanko O, Watanabe S, Jasenosky LD, Watanabe T, Kawaoka Y. 2011. ALIX/AIP1 is required for NP incorporation into Mopeia virus Z-induced virus-like particles. J Virol 85:3631–3641. doi: 10.1128/JVI.01984-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Noton SL, Medcalf E, Fisher D, Mullin AE, Elton D, Digard P. 2007. Identification of the domains of the influenza A virus M1 matrix protein required for NP binding, oligomerization and incorporation into virions. J Gen Virol 88:2280–2290. doi: 10.1099/vir.0.82809-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rodríguez L, Cuesta I, Asenjo A, Villanueva N. 2004. Human respiratory syncytial virus matrix protein is an RNA-binding protein: binding properties, location and identity of the RNA contact residues. J Gen Virol 85:709–719. doi: 10.1099/vir.0.19707-0. [DOI] [PubMed] [Google Scholar]
  • 38.Iwasaki M, Takeda M, Shirogane Y, Nakatsu Y, Nakamura T, Yanagi Y. 2009. The matrix protein of measles virus regulates viral RNA synthesis and assembly by interacting with the nucleocapsid protein. J Virol 83:10374–10383. doi: 10.1128/JVI.01056-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lamb RA, Parks GD. 2007. Paramyxoviridae: the viruses and their replication, p 1449–1496. In Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA, Roizman B, Straus SE (ed), Fields virology, 5th ed Lippincott Williams & Wilkins, Philadelphia, PA. [Google Scholar]
  • 40.Longhi S. 2009. Nucleocapsid structure and function. Curr Top Microbiol Immunol 329:103–128. [DOI] [PubMed] [Google Scholar]
  • 41.Bharat TA, Noda T, Riches JD, Kraehling V, Kolesnikova L, Becker S, Kawaoka Y, Briggs JA. 2012. Structural dissection of Ebola virus and its assembly determinants using cryo-electron tomography. Proc Natl Acad Sci U S A 109:4275–4280. doi: 10.1073/pnas.1120453109. [DOI] [PMC free article] [PubMed] [Google Scholar]

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

RESOURCES