Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Virol ; 96(2): e0164821, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34730394

ABSTRACT

Human parainfluenza virus 3 (HPIV3) belongs to the Paramyxoviridae, causing annual worldwide epidemics of respiratory diseases, especially in newborns and infants. The core components consist of just three viral proteins: nucleoprotein (N), phosphoprotein (P), and RNA polymerase (L), playing essential roles in replication and transcription of HPIV3 as well as other paramyxoviruses. Viral genome encapsidated by N is as a template and recognized by RNA-dependent RNA polymerase complex composed of L and P. The offspring RNA also needs to assemble with N to form nucleocapsids. The N is one of the most abundant viral proteins in infected cells and chaperoned in the RNA-free form (N0) by P before encapsidation. In this study, we presented the structure of unassembled HPIV3 N0 in complex with the N-terminal portion of the P, revealing the molecular details of the N0 and the conserved N0-P interaction. Combined with biological experiments, we showed that the P binds to the C-terminal domain of N0 mainly by hydrophobic interaction and maintains the unassembled conformation of N by interfering with the formation of N-RNA oligomers, which might be a target for drug development. Based on the complex structure, we developed a method to obtain the monomeric N0. Furthermore, we designed a P-derived fusion peptide with 10-fold higher affinity, which hijacked the N and interfered with the binding of the N to RNA significantly. Finally, we proposed a model of conformational transition of N from the unassembled state to the assembled state, which helped to further understand viral replication. IMPORTANCE Human parainfluenza virus 3 (HPIV3) causes annual epidemics of respiratory diseases, especially in newborns and infants. For the replication of HPIV3 and other paramyxoviruses, only three viral proteins are required: phosphoprotein (P), RNA polymerase (L), and nucleoprotein (N). Here, we report the crystal structure of the complex of N and its chaperone P. We describe in detail how P acts as a chaperone to maintain the unassembled conformation of N. Our analysis indicated that the interaction between P and N is conserved and mediated by hydrophobicity, which can be used as a target for drug development. We obtained a high-affinity P-derived peptide inhibitor, specifically targeted N, and greatly interfered with the binding of the N to RNA, thereby inhibiting viral encapsidation and replication. In summary, our results provide new insights into the paramyxovirus genome replication and nucleocapsid assembly and lay the basis for drug development.


Subject(s)
Molecular Chaperones/chemistry , Nucleocapsid Proteins/chemistry , Parainfluenza Virus 3, Human/chemistry , Phosphoproteins/chemistry , Amino Acid Sequence , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Parainfluenza Virus 3, Human/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Conformation , RNA, Viral/metabolism
2.
Vet Microbiol ; 259: 109129, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34087675

ABSTRACT

Caprine parainfluenza virus type 3 (CPIV3) is one of the most important viral respiratory pathogens of goat. Accumulating evidence demonstrates that apoptosis is a cellular mechanism for the host response to pathogens, and it participates in regulating viral replication. However, there is little study on CPIV3-induced host cells apoptosis. In this study, primary goat tracheal epithelial (GTE) cells were established as a cellular model that is permissive to CPIV3 infection. Then, we showed that CPIV3 infection induced apoptosis in GTE cells, as determined by morphological changes, flow cytometry and TUNEL assay. Moreover, Caspase activity and the expression of pro-apoptotic genes further suggested that CPIV3 induced apoptosis by activating both the intrinsic and extrinsic pathways. Mechanistically, the ability of CPIV3 to induce apoptosis was activated by N protein, and the viral protein increased CPIV3 replication through effecting apoptosis. Overall, our findings showed that GTE cells that will enable further analysis of CPIV3 infection and offers novel insights into the mechanisms of CPIV3-induced apoptosis in host cells.


Subject(s)
Apoptosis/genetics , Nucleocapsid Proteins/genetics , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Respirovirus Infections/genetics , Respirovirus Infections/veterinary , Virus Replication/genetics , Animals , Cells, Cultured , Epithelial Cells/virology , Goat Diseases/virology , Goats/virology , Nucleocapsid Proteins/metabolism , Parainfluenza Virus 3, Human/pathogenicity , Respirovirus Infections/virology , Trachea/cytology
3.
Virus Genes ; 56(1): 37-48, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31768710

ABSTRACT

Human parainfluenza virus type 3 (HPIV3) causes the majority of childhood viral pneumonia around the world. Fusing the viral and target cell membranes is crucial for its entry into target cells, and the fusion process requires the concerted actions of two viral glycoproteins: hemagglutinin-neuraminidase (HN) and fusion (F) protein. After binding to the cell surface receptor, sialic acids, HN triggers F to undergo large conformational rearrangements to execute the fusion process. Although it has been reported that several domains of F had important impacts on regulating the membrane fusion activity, what role the DI-DII linker (residues 369-374, namely L1 linker) of the HPIV3 F protein plays in the fusion process still remains confused. We have obtained three chimeric mutant proteins (Ch-NDV-L1, Ch-MV-L1, Ch-HPIV1-L1) containing the full length of HPIV3 F protein but their corresponding DI-DII linker derived from the F protein of Newcastle disease virus (NDV), Measles virus (MV), and Human parainfluenza virus type 1 (HPIV1), respectively. One deletion mutant protein (De-L1), whose DI-DII linker was deleted, has been established simultaneously. Then vaccinia virus-T7 RNA polymerase transient expression system and standard plasmid system were utilized to express the mutant F proteins in BHK-21 cells. These four mutants were determined for membrane fusogenic activity, cell surface expression level, and total mutant F protein expression. All of them resulted in a significant reduction in fusogenic activity in all steps of cell-cell membrane fusion process. There was no significant difference in cell surface protein expression level for the mutants compared with wild-type F. The mutant proteins with inability in fusogenic activity were all at the form of precursor protein, F0, which were not hydrolyzed by intracellular protease furin. The results above suggest that the involvement of the DI-DII linker region is necessary for the complete fusion of the membranes.


Subject(s)
Parainfluenza Virus 3, Human/metabolism , Respirovirus Infections/virology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Cell Line , Cell Membrane/virology , HN Protein/genetics , HN Protein/metabolism , Humans , Membrane Fusion , Newcastle disease virus/genetics , Newcastle disease virus/metabolism , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Protein Domains , Viral Fusion Proteins/genetics
4.
J Am Chem Soc ; 141(32): 12648-12656, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31268705

ABSTRACT

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) cause lower respiratory infection in infants and young children. There are no vaccines for these pathogens, and existing treatments have limited or questionable efficacy. Infection by HPIV3 or RSV requires fusion of the viral and cell membranes, a process mediated by a trimeric fusion glycoprotein (F) displayed on the viral envelope. Once triggered, the pre-fusion form of F undergoes a series of conformational changes that first extend the molecule to allow for insertion of the hydrophobic fusion peptide into the target cell membrane and then refold the trimeric assembly into an energetically stable post-fusion state, a process that drives the merger of the viral and host cell membranes. Peptides derived from defined regions of HPIV3 F inhibit infection by HPIV3 by interfering with the structural transitions of the trimeric F assembly. Here we describe lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F that potently inhibit infection by both HPIV3 and RSV. The lead peptide inhibits RSV infection as effectively as does a peptide corresponding to the RSV HRC domain itself. We show that the inhibitors bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F with high affinity. Co-crystal structures of inhibitors bound to the HRN domains of HPIV3 or RSV F reveal remarkably different modes of binding in the N-terminal segment of the inhibitor.


Subject(s)
Lipopeptides/pharmacology , Parainfluenza Virus 3, Human/drug effects , Peptide Fragments/pharmacology , Respiratory Syncytial Viruses/drug effects , Viral Fusion Protein Inhibitors/pharmacology , Viral Fusion Proteins/pharmacology , Amino Acid Sequence , Crystallography, X-Ray , Humans , Lipopeptides/metabolism , Microbial Sensitivity Tests , Parainfluenza Virus 3, Human/chemistry , Peptide Fragments/metabolism , Protein Binding , Respiratory Mucosa/virology , Respiratory Syncytial Viruses/chemistry , Viral Fusion Protein Inhibitors/metabolism , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects
5.
Proc Natl Acad Sci U S A ; 115(48): 12265-12270, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30420505

ABSTRACT

Parainfluenza virus types 1-4 (PIV1-4) are highly infectious human pathogens, of which PIV3 is most commonly responsible for severe respiratory illness in newborns, elderly, and immunocompromised individuals. To obtain a vaccine effective against all four PIV types, we engineered mutations in each of the four PIV fusion (F) glycoproteins to stabilize their metastable prefusion states, as such stabilization had previously enabled the elicitation of high-titer neutralizing antibodies against the related respiratory syncytial virus. A cryoelectron microscopy structure of an engineered PIV3 F prefusion-stabilized trimer, bound to the prefusion-specific antibody PIA174, revealed atomic-level details for how introduced mutations improved stability as well as how a single PIA174 antibody recognized the trimeric apex of prefusion PIV3 F. Nine combinations of six newly identified disulfides and two cavity-filling mutations stabilized the prefusion PIV3 F immunogens and induced 200- to 500-fold higher neutralizing titers in mice than were elicited by PIV3 F in the postfusion conformation. For PIV1, PIV2, and PIV4, we also obtained stabilized prefusion Fs, for which prefusion versus postfusion titers were 2- to 20-fold higher. Elicited murine responses were PIV type-specific, with little cross-neutralization of other PIVs. In nonhuman primates (NHPs), quadrivalent immunization with prefusion-stabilized Fs from PIV1-4 consistently induced potent neutralizing responses against all four PIVs. For PIV3, the average elicited NHP titer from the quadrivalent immunization was more than fivefold higher than any titer observed in a cohort of over 100 human adults, highlighting the ability of a prefusion-stabilized immunogen to elicit especially potent neutralization.


Subject(s)
Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 4, Human/immunology , Respirovirus Infections/immunology , Viral Fusion Proteins/chemistry , Viral Vaccines/chemistry , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cryoelectron Microscopy , Female , Humans , Macaca mulatta , Male , Mice , Parainfluenza Virus 1, Human/chemistry , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/chemistry , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 4, Human/chemistry , Parainfluenza Virus 4, Human/genetics , Respiratory Syncytial Virus Infections , Respirovirus Infections/prevention & control , Respirovirus Infections/virology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
6.
J Gen Virol ; 99(6): 763-767, 2018 06.
Article in English | MEDLINE | ID: mdl-29683419

ABSTRACT

HPIV3 is a respiratory virus causing airway diseases, including pneumonia, croup, and bronchiolitis, during infancy and childhood. Currently there is no effective vaccine or anti-viral therapy for this virus. Studies have suggested that poor T cell proliferation following HPIV3 infection is responsible for impaired immunological memory associated with this virus. We have previously demonstrated that NK cells mediate regulation of T cell proliferation during HPIV3 infection. Here we add to these studies by demonstrating that the regulation of T cell proliferation during HPIV3 infection is mediated via NK receptors NKp44 and NKp46 and involves the surface glycoprotein haemagglutinin-neuraminidase but not the fusion protein of the virus. These studies extend our knowledge of the regulatory repertoire of NK cells and provide mechanistic insights which may explain reoccurring failures of vaccines against this virus.


Subject(s)
HN Protein/chemistry , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Natural Cytotoxicity Triggering Receptor 2/metabolism , Parainfluenza Virus 3, Human/chemistry , T-Lymphocytes/cytology , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , HN Protein/genetics , Humans , Lipopolysaccharide Receptors/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 2/genetics , Parainfluenza Virus 3, Human/genetics , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , T-Lymphocytes/immunology
7.
Virus Genes ; 54(3): 333-342, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29516315

ABSTRACT

Human parainfluenza virus type 3 (hPIV3) is an important respiratory pathogen that causes the majority of viral pneumonia of infants and young children. hPIV3 can infect host cells through the synergistic action of hemagglutinin-neuraminidase (HN) protein and the homotypic fusion (F) protein on the viral surface. HN protein plays a variety of roles during the virus invasion process, such as promoting viral particles to bind to receptors, cleaving sialic acid, and activating the F protein. Crystal structure research shows that HN tetramer adopted a "heads-down" conformation, at least two heads dimmer on flank of the four-helix bundle stalk, which forms a symmetrical interaction interface. The stalk region determines interactions and activation of F protein in specificity, and the heads in down position statically shield these residues. In order to make further research on the function of these amino acids at the hPIV3 HN stalk/head interface, fifteen mutations (8 sites from stalk and 7 sites from head) were engineered into this interface by site-directed mutagenesis in this study. Alanine substitution in this region of hPIV3 HN had various effects on cell fusion promotion, receptor binding, and neuraminidase activity. Besides, L151A also affected surface protein expression efficiency. Moreover, I112A, D120A, and R122A mutations of the stalk region that were masked by global head in down position had influence on the interaction between F and HN proteins.


Subject(s)
Amino Acids/physiology , HN Protein/chemistry , HN Protein/physiology , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/physiology , Virus Internalization , Alanine/chemistry , Cell Line , Cell Membrane/metabolism , Giant Cells/virology , HN Protein/genetics , Hemadsorption , Humans , Membrane Fusion/physiology , Mutagenesis, Site-Directed , Neuraminidase/metabolism , Parainfluenza Virus 3, Human/genetics , Protein Conformation , Receptors, Virus/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/physiology
8.
Antiviral Res ; 123: 216-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26364554

ABSTRACT

The human parainfluenza virus type 3 (hPIV3) hemagglutinin-neuraminidase (HN) has opposing functions of binding sialic acid receptors and cleaving them, facilitating virus release. The crystal structure of hPIV3 HN complexed with the substrate analogue difluorosialic acid (DFSA) revealed that catalysis by HN involves the formation of a covalently linked sialosyl-enzyme intermediate which was trapped along with a transition-state analogue resembling an oxocarbenium ion. This mechanism of enzyme catalysis was also confirmed in the crystal structure of the influenza N9 neuraminidase complexed with DFSA. Additionally, novel secondary receptor binding sites were identified in the hPIV3 HN-DFSA complex including one near the catalytic cavity which upon binding DFSA imposes subtle changes and may help the HN balance the opposing functions. Multiple receptor binding sites may increase avidity to facilitate cell binding and fusion promotion. The secondary receptor binding sites in the paramyxoviruses are so far unique to each virus type.


Subject(s)
HN Protein/chemistry , HN Protein/metabolism , Parainfluenza Virus 3, Human/enzymology , Sialic Acids/chemistry , Sialic Acids/metabolism , Binding Sites , Biotransformation , Crystallography, X-Ray , Humans , Parainfluenza Virus 3, Human/chemistry , Protein Binding , Protein Conformation
9.
mBio ; 6(1): e02393-14, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25691596

ABSTRACT

UNLABELLED: In order to deliver their genetic material to host cells during infection, enveloped viruses use specialized proteins on their surfaces that bind cellular receptors and induce fusion of the viral and host membranes. In paramyxoviruses, a diverse family of single-stranded RNA (ssRNA) viruses, including several important respiratory pathogens, such as parainfluenza viruses, the attachment and fusion machinery is composed of two separate proteins: a receptor binding protein (hemagglutinin-neuraminidase [HN]) and a fusion (F) protein that interact to effect membrane fusion. Here we used negative-stain and cryo-electron tomography to image the 3-dimensional ultrastructure of human parainfluenza virus 3 (HPIV3) virions in the absence of receptor engagement. We observed that HN exists in at least two organizations. The first were arrays of tetrameric HN that lacked closely associated F proteins: in these purely HN arrays, HN adopted a "heads-down" configuration. In addition, we observed regions of complex surface density that contained HN in an apparently extended "heads-up" form, colocalized with prefusion F trimers. This colocalization with prefusion F prior to receptor engagement supports a model for fusion in which HN in its heads-up state and F may interact prior to receptor engagement without activating F, and that interaction with HN in this configuration is not sufficient to activate F. Only upon receptor engagement by HN's globular head does HN transmit its activating signal to F. IMPORTANCE: Human parainfluenza virus 3 (HPIV3) is an enveloped, ssRNA virus that can cause serious respiratory illness, especially in children. HPIV3, like most other paramyxoviruses, uses two specialized proteins to mediate cell entry: the fusion protein (F) and the receptor binding protein, hemagglutinin-neuraminidase (HN). F becomes activated to mediate fusion during entry when it is triggered by a signal from HN. Here we used electron tomography to reconstruct the 3-dimensional ultrastructure of HPIV3. From these structures, we could discern the distribution and, in some cases, conformation of HN and F proteins, which provided an understanding of their interrelationship on virions. HN is found in arrays alone in one conformation and interspersed with prefusion F trimers in another. The data support a model of paramyxovirus membrane fusion in which HN associates with F before receptor engagement, and receptor engagement by the globular head of HN switches the HN-F interaction into one of fusion activation.


Subject(s)
Electron Microscope Tomography , HN Protein/metabolism , HN Protein/ultrastructure , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/ultrastructure , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/ultrastructure , Cryoelectron Microscopy , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/ultrastructure , Protein Binding , Receptors, Cell Surface/metabolism , Staining and Labeling
10.
Virus Genes ; 47(1): 10-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23686695

ABSTRACT

The human parainfluenza virus type 3 (HPIV3) phosphoprotein (P) gene is unusual as it contains an editing site where nontemplated ribonucleotide residues can be inserted. This RNA editing can lead to the expression of the viral P, PD, putative W, and theoretical V protein from a single gene. Although the HPIV3 PD protein has been detected, its function and those of the W and V proteins are poorly understood. Therefore, we first used reverse genetics techniques to construct and rescue a recombinant (r)HPIV3 clone with a polyhistidine sequence at the 5' end of the P gene for tagged protein detection. Western blot analysis demonstrated the presence of the P, PD, and W proteins, but no V protein was detected. Then, we functionally studied the D domain of the PD protein by constructing two rHPIV3 knockout clones that are deficient in the expression of the D domain. Results from growth kinetic studies with infected MA-104 and A596 cells showed that viral replication of the two knockout viruses (rHPIV3-ΔES and rHPIV3-ΔD) was comparable to that of the parental virus in both cell lines. However, viral mRNA transcription and genomic replication was significantly reduced. Furthermore, cytokine/chemokine profiles of A549 cells infected with either knockout virus were unchanged or showed lower levels compared to those from cells infected with the parental virus. These data suggest that the D domain of the PD protein may play a luxury role in HPIV3 RNA synthesis and may also be involved in disrupting the expression of beta interferon.


Subject(s)
Interferon-beta/genetics , Parainfluenza Virus 3, Human/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , RNA, Viral/genetics , Respirovirus Infections/genetics , Viral Proteins/chemistry , Viral Proteins/metabolism , Cell Line , Down-Regulation , Humans , Interferon-beta/immunology , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Phosphoproteins/genetics , Protein Structure, Tertiary , RNA, Viral/metabolism , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sequence Deletion , Viral Proteins/genetics
11.
Bing Du Xue Bao ; 29(5): 500-8, 2013 Sep.
Article in Chinese | MEDLINE | ID: mdl-24386838

ABSTRACT

To determine the functions of N-carbohydrate chains in human parainfluenza virus type 3 hemagglutinin-neuraminidase(HN) protein, a PCR-based site-directed mutagenesis method was used to obtain N-glycan mutants. Protein electrophoresis rate, cell surface expression,receptor binding activity, neuraminidase activity and cell fusion promotion activity were determined. The HN proteins of single mutants (G1, G2, and G4) and multiple mutants (G12, G14, G24 and G124) migrated faster than the wild-type (wt) HN protein on polyacrylamide gels, while G3-mutated protein and wt HN protein migrated at the same position. There was no statistic difference in cell surface expression and neuraminidase activity between wt and each mutant HN protein (P>0.05), but receptor binding activity and cell fusion promotion activity of each mutant protein was reduced to significant extent (P<0.05). G1, G2 and G4 mutants exhibited re duced receptor binding activity, which was 83.94%, 76.45% and 55.32% of the wt level, respectively. G1, G2 and G4-mutated proteins also showed reductions in fusion promotion activity, which was 80.84%, 77.83% and 64.16%, respectively. Multiple mutants with G12-, G14-, G24- and G124- substitutions could further reduce receptor binding activities, 33.07%, 20.67%, 19.96% and 15.11% of the wt HN level, respectively. G12, G14, G24 and G124 mutants exhibited levels of fusion promotion activity that were only 46.360, 12.04%, 13.43% and 4.05% of the wt amount, respectively. As N-glycans of hPIV3 HN protein play an important role in receptor binding activity and cell fusion promotion activity of HN protein. We propose that the loss of N-glycans change the conformation or orientation of globular domain that is responsible for receptor binding and lower receptor binding activity and cell fusion promotion activi ty.


Subject(s)
HN Protein/chemistry , HN Protein/metabolism , Parainfluenza Virus 3, Human/enzymology , Respirovirus Infections/virology , Glycosylation , HN Protein/genetics , Humans , Mutation , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/physiology , Protein Binding , Receptors, Virus/metabolism , Respirovirus Infections/metabolism , Virus Internalization
12.
J Am Chem Soc ; 134(44): 18447-52, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23057491

ABSTRACT

Human parainfluenza virus type 3 (hPIV-3) is a clinically significant pathogen and is the causative agent of pneumonia and bronchiolitis in children. In this study the solution dynamics of human parainfluenza type 3 hemagglutinin-neuraminidase (HN) have been investigated. A flexible loop around Asp216 that adopts an open conformation in direct vicinity of the active site of the apo-form of the protein and closes upon inhibitor binding has been identified. To date, no available X-ray crystal structure has shown the molecular dynamics simulation-derived predominant loop-conformation states found in the present study. The outcomes of this study provide additional insight into the dynamical properties of hPIV-3 HN and may have important implications in defining HN glycan recognition events, receptor specificity, and antiparainfluenza virus drug discovery.


Subject(s)
HN Protein/chemistry , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/enzymology , Catalytic Domain , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Protein Conformation , Respirovirus Infections/virology
13.
J Virol ; 84(6): 3094-100, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20053750

ABSTRACT

The hemagglutinin-neuraminidase (HN) glycoprotein plays a critical role in parainfluenza virus replication. We recently found that in addition to the catalytic binding site, HN of human parainfluenza virus type 1 (hPIV-1) may have a second receptor-binding site covered by an N-linked glycan at residue 173, which is near the region of the second receptor-binding site identified in Newcastle disease virus (NDV) HN (I. A. Alymova, G. Taylor, V. P. Mishin, M. Watanabe, K. G. Murti, K. Boyd, P. Chand, Y. S. Babu, and A. Portner, J. Virol. 82:8400-8410, 2008). Sequence analysis and superposition of the NDV and hPIV-3 HN dimer structures revealed that, similar to what was seen in hPIV-1, the N-linked glycan at residue 523 on hPIV-3 HN may cover a second receptor-binding site. Removal of this N-linked glycosylation site by an Asn-to-Asp substitution at residue 523 (N523D) changed the spectrum of the mutant virus's receptor specificity, delayed its elution from both turkey and chicken red blood cells, reduced mutant sensitivity (by about half) to the selective HN inhibitor BCX 2855 in hemagglutination inhibition tests, and slowed its growth in LLC-MK(2) cells. The neuraminidase activity of the mutant and its sensitivity to BCX 2855 in neuraminidase inhibition assays did not change, indicating that the mutation did not affect the virus's catalytic-binding site and that all observed effects were caused by the exposure of the purported second receptor-binding site. Our data are consistent with the idea that, similar to the case for hPIV-1, the N-linked glycan shields a second receptor-binding site on hPIV-3 HN.


Subject(s)
HN Protein/chemistry , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/metabolism , Polysaccharides/chemistry , Animals , Binding Sites , Chickens , Glycosylation , HN Protein/genetics , HN Protein/metabolism , Humans , Models, Molecular , Neuraminidase/metabolism , Parainfluenza Virus 1, Human/metabolism , Parainfluenza Virus 3, Human/genetics , Polysaccharides/genetics , Polysaccharides/metabolism , Protein Multimerization , Protein Structure, Quaternary , Turkeys , Virus Replication
14.
Antiviral Res ; 72(1): 1-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16730076

ABSTRACT

Endoplasmic reticulum (ER) alpha-glucosidase inhibitors block the trimming of N-linked glycosylation and thus prevent the production of several viruses. The present study investigates the antiviral effects of the alpha-glucosidase and alpha-mannosidase inhibitors (castanospermine, 1-deoxynojirimycin, bromoconduritol, deoxymannojirimycin and swainsonine) on human parainfluenza virus type 3 (HPIV3). The alpha-glucosidase inhibitors (castanospermine, 1-deoxynojirimycin) in recombinant expression systems reduced the surface and intracellular expression of both HPIV3 F and HN proteins. On the other hand, alpha-mannosidase inhibitors prevented processing of the oligosaccharides on HPIV3 glycoproteins into the complex form. Consequently, alpha-glycosidase inhibitors (castanospermine and 1-deoxynojirimycin) significantly inhibited viral fusion activity. We demonstrated that the alpha-glucosidase inhibitors (castanospermine and 1-deoxynojirimycin) reduced the infectivity of newly released viral particles. We postulate that alpha-glucosidase inhibitors can prevent the first steps of HPIV3 envelope glycoprotein processing and that the inhibition of glucose trimming has antiviral effects.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucose/metabolism , Glycosylation/drug effects , Parainfluenza Virus 3, Human/drug effects , alpha-Glucosidases/pharmacology , alpha-Mannosidase/pharmacology , Animals , Cell Line , Glycoside Hydrolase Inhibitors , HN Protein/genetics , HN Protein/metabolism , Humans , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/enzymology , Parainfluenza Virus 3, Human/physiology , Recombinant Proteins/biosynthesis , Transfection , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Replication , alpha-Mannosidase/antagonists & inhibitors
15.
Virus Res ; 99(2): 177-85, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14749183

ABSTRACT

The promotion of membrane fusion by the fusion (F) protein of human parainfluenza virus 3 (hPIV3) is dependent on a virus-specific contribution from the hemagglutinin-neuraminidase (HN) protein. By evaluation of chimeric hPIV3-Newcastle disease virus (NDV) HN proteins, we have previously shown that hPIV3-F-specificity is determined by a domain that extends from the middle of the membrane anchor to the 82nd residue in the ectodomain [Virology 209, (1995) 457; Arch. Virol. 13 (1997) 115]. If the corresponding NDV-derived residues replace the two C-terminal residues in this domain, no fusion is detected. However, these substitutions restore a glycosylation site present in NDV HN, but not in hPIV3 HN. Deletion of this site from a nested set of chimeras with hPIV3-derived N-terminal portions of decreasing length partially restores fusion, suggesting that an oligosaccharide near the top of hPIV3 HN stalk modulates fusion. In addition, further mutational analyses show that a chimera with only 125 N-terminal hPIV3-derived residues (72 in the stalk) actually promotes fusion more efficiently than the wt protein. These findings localize the C-terminus of the F-specific domain in hPIV3 HN a full 10 residues closer to the membrane than previously shown.


Subject(s)
HN Protein/chemistry , HN Protein/physiology , Membrane Fusion , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/physiology , Animals , Cell Line , Cricetinae , Glycosylation , HN Protein/genetics , Newcastle disease virus/genetics , Oligosaccharides , Parainfluenza Virus 3, Human/genetics , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/physiology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/physiology
16.
J Mol Biol ; 335(5): 1343-57, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14729348

ABSTRACT

The three-dimensional structure of the haemagglutinin-neuraminidase (HN) from a human parainfluenza virus is described at ca 2.0 A resolution, both in native form and in complex with three substrate analogues. In support of earlier work on the structure of the homologous protein from the avian pathogen Newcastle disease virus (NDV), we observe a dimer of beta-propellers and find no evidence for spatially separated sites performing the receptor-binding and neuraminidase functions of the protein. As with the NDV HN, the active site of the HN of parainfluenza viruses is structurally flexible, suggesting that it may be able to switch between a receptor-binding state and a catalytic state. However, in contrast to the NDV structures, we observe no ligand-induced structural changes that extend beyond the active site and modify the dimer interface.


Subject(s)
HN Protein/chemistry , HN Protein/metabolism , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/metabolism , Receptors, Virus/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Crystallization , Dimerization , HN Protein/genetics , Humans , Hymecromone/analogs & derivatives , Hymecromone/metabolism , Ligands , Molecular Sequence Data , Newcastle disease virus/chemistry , Parainfluenza Virus 3, Human/drug effects , Protein Conformation , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Respirovirus Infections/drug therapy , Respirovirus Infections/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship
17.
Microb Pathog ; 20(3): 179-84, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8965678

ABSTRACT

In order for the fusion protein (F) of the human parainfluenza virus type 3 (HPF3) to promote membrane fusion and viral entry, the haemagglutinin-neuraminidase (HN) glycoprotein must interact with its receptor. Sialoglycoconjugates are known to be the receptors for the HPF3 HN, however specific attachment factors or receptors for HPF3 have not been identified. In this report we describe the analysis of variants of HPF3 with increased fusion-promoting phenotypes that were selected by treatment with viral neuraminidase. The results suggest that for HPF3, the virus is specific in its use of sialic acid receptors; the majority of sialic-acid containing molecules are not targets for HPF3.


Subject(s)
Parainfluenza Virus 3, Human/chemistry , Receptors, Virus/analysis , Genetic Variation , Neuraminidase/metabolism , Viral Fusion Proteins/analysis
18.
Proc Natl Acad Sci U S A ; 93(5): 2186-91, 1996 Mar 05.
Article in English | MEDLINE | ID: mdl-8700906

ABSTRACT

The synthetic peptides DP-107 and DP-178 (T-20), derived from separate domains within the human immunodeficiency virus type 1 (HIV-1) transmembrane (TM) protein, gp4l, are stable and potent inhibitors of HIV-1 infection and fusion. Using a computer searching strategy (computerized antiviral searching technology, C.A.S.T.) based on the predicted secondary structure of DP-107 and DP-178 (T-20), we have identified conserved heptad repeat domains analogous to the DP-107 and DP-178 regions of HIV-1 gp41 within the glycoproteins of other fusogenic viruses. Here we report on antiviral peptides derived from three representative paramyxoviruses, respiratory syncytial virus (RSV), human parainfluenza virus type 3 (HPIV-3), and measles virus (MV). We screened crude preparations of synthetic 35-residue peptides, scanning the DP-178-like domains, in antiviral assays. Peptide preparations demonstrating antiviral activity were purified and tested for their ability to block syncytium formation. Representative DP-178-like peptides from each paramyxovirus blocked homologous virus-mediated syncytium formation and exhibited EC50 values in the range 0.015-0.250 microM. Moreover, these peptides were highly selective for the virus of origin. Identification of biologically active peptides derived from domains within paramyxovirus F1 proteins analogous to the DP-178 domain of HIV-1 gp4l is compelling evidence for equivalent structural and functional features between retroviral and paramyxoviral fusion proteins. These antiviral peptides provide a novel approach to the development of targeted therapies for paramyxovirus infections.


Subject(s)
Antiviral Agents , Measles virus/chemistry , Membrane Fusion , Parainfluenza Virus 3, Human/chemistry , Respiratory Syncytial Viruses/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Circular Dichroism , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Secondary , Sequence Alignment , Structure-Activity Relationship
19.
J Infect Dis ; 152(6): 1219-30, 1985 Dec.
Article in English | MEDLINE | ID: mdl-2999259

ABSTRACT

The envelope glycoproteins of human parainfluenza type 3 virus were characterized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and reactivity with specific monoclonal antibodies. The molecular weight of the hemagglutinin-neuraminidase (HN) glycoprotein was found to be 72,000, and the fusion (F) glycoprotein appeared to consist of 74,000 (F0) or 56,000 (F1) species. Envelope glycoproteins were solubilized with octyl-glucoside and, after removal of the detergent by dialysis, were used for immunization of hamsters. Other animals were immunized with a formalin-inactivated preparation of whole virus. A single subcutaneous immunization with these antigen preparations induced a serum antibody response to the HN and F glycoproteins, as determined by plaque neutralization, hemagglutination inhibition, inhibition of virus-induced cell fusion, and immune precipitation tests. An IgG antibody response to both glycoproteins was also observed in bronchial washings. Animals immunized with the highest dose of envelope glycoproteins showed complete protection from challenge infection, whereas immunization with inactivated virus did not completely protect animals.


Subject(s)
Hemagglutinins, Viral/analysis , Parainfluenza Virus 3, Human/chemistry , Respirovirus/chemistry , Viral Vaccines/analysis , Animals , Antibodies, Monoclonal , Antibodies, Viral/biosynthesis , Bronchi/immunology , Bronchi/microbiology , Cricetinae , Female , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/immunology , Humans , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/physiology , Paramyxoviridae Infections/microbiology , Paramyxoviridae Infections/prevention & control , Peptides/analysis , Peptides/immunology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/analysis , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...