Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 22(1): 25-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33154590

ABSTRACT

Clinical manifestations of COVID-19 caused by the new coronavirus SARS-CoV-2 are associated with age1,2. Adults develop respiratory symptoms, which can progress to acute respiratory distress syndrome (ARDS) in the most severe form, while children are largely spared from respiratory illness but can develop a life-threatening multisystem inflammatory syndrome (MIS-C)3-5. Here, we show distinct antibody responses in children and adults after SARS-CoV-2 infection. Adult COVID-19 cohorts had anti-spike (S) IgG, IgM and IgA antibodies, as well as anti-nucleocapsid (N) IgG antibody, while children with and without MIS-C had reduced breadth of anti-SARS-CoV-2-specific antibodies, predominantly generating IgG antibodies specific for the S protein but not the N protein. Moreover, children with and without MIS-C had reduced neutralizing activity as compared to both adult COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children independent of whether they develop MIS-C, with implications for developing age-targeted strategies for testing and protecting the population.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation/immunology , COVID-19/immunology , Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adolescent , Adult , Aged , COVID-19/virology , Child , Child, Preschool , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Middle Aged , SARS-CoV-2/physiology , Young Adult
2.
J Virol ; 98(1): e0179123, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38168672

ABSTRACT

In the United States (US), biosafety and biosecurity oversight of research on viruses is being reappraised. Safety in virology research is paramount and oversight frameworks should be reviewed periodically. Changes should be made with care, however, to avoid impeding science that is essential for rapidly reducing and responding to pandemic threats as well as addressing more common challenges caused by infectious diseases. Decades of research uniquely positioned the US to be able to respond to the COVID-19 crisis with astounding speed, delivering life-saving vaccines within a year of identifying the virus. We should embolden and empower this strength, which is a vital part of protecting the health, economy, and security of US citizens. Herein, we offer our perspectives on priorities for revised rules governing virology research in the US.


Subject(s)
Biomedical Research , Containment of Biohazards , Virology , Humans , COVID-19 , United States , Viruses , Biomedical Research/standards
3.
PLoS Biol ; 18(9): e3000849, 2020 09.
Article in English | MEDLINE | ID: mdl-32898168

ABSTRACT

Despite limited genomic diversity, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown a wide range of clinical manifestations in different patient populations. The mechanisms behind these host differences are still unclear. Here, we examined host response gene expression across infection status, viral load, age, and sex among shotgun RNA sequencing profiles of nasopharyngeal (NP) swabs from 430 individuals with PCR-confirmed SARS-CoV-2 and 54 negative controls. SARS-CoV-2 induced a strong antiviral response with up-regulation of antiviral factors such as OAS1-3 and IFIT1-3 and T helper type 1 (Th1) chemokines CXCL9/10/11, as well as a reduction in transcription of ribosomal proteins. SARS-CoV-2 culture in human airway epithelial (HAE) cultures replicated the in vivo antiviral host response 7 days post infection, with no induction of interferon-stimulated genes after 3 days. Patient-matched longitudinal specimens (mean elapsed time = 6.3 days) demonstrated reduction in interferon-induced transcription, recovery of transcription of ribosomal proteins, and initiation of wound healing and humoral immune responses. Expression of interferon-responsive genes, including ACE2, increased as a function of viral load, while transcripts for B cell-specific proteins and neutrophil chemokines were elevated in patients with lower viral load. Older individuals had reduced expression of the Th1 chemokines CXCL9/10/11 and their cognate receptor CXCR3, as well as CD8A and granzyme B, suggesting deficiencies in trafficking and/or function of cytotoxic T cells and natural killer (NK) cells. Relative to females, males had reduced B cell-specific and NK cell-specific transcripts and an increase in inhibitors of nuclear factor kappa-B (NF-κB) signaling, possibly inappropriately throttling antiviral responses. Collectively, our data demonstrate that host responses to SARS-CoV-2 are dependent on viral load and infection time course, with observed differences due to age and sex that may contribute to disease severity.


Subject(s)
Antiviral Agents/immunology , Betacoronavirus/physiology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , COVID-19 , Child , Child, Preschool , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Female , Gene Expression Regulation , Humans , Immunity/genetics , Kinetics , Male , Middle Aged , Nasopharynx/immunology , Nasopharynx/virology , Pandemics , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Ribosomal Proteins/genetics , SARS-CoV-2 , Sex Factors , Signal Transduction/genetics , Viral Load , Wound Healing/genetics , Young Adult
4.
PLoS Pathog ; 16(9): e1008883, 2020 09.
Article in English | MEDLINE | ID: mdl-32956394

ABSTRACT

Infection by human parainfluenza viruses (HPIVs) causes widespread lower respiratory diseases, including croup, bronchiolitis, and pneumonia, and there are no vaccines or effective treatments for these viruses. HPIV3 is a member of the Respirovirus species of the Paramyxoviridae family. These viruses are pleomorphic, enveloped viruses with genomes composed of single-stranded negative-sense RNA. During viral entry, the first step of infection, the viral fusion complex, comprised of the receptor-binding glycoprotein hemagglutinin-neuraminidase (HN) and the fusion glycoprotein (F), mediates fusion upon receptor binding. The HPIV3 transmembrane protein HN, like the receptor-binding proteins of other related viruses that enter host cells using membrane fusion, binds to a receptor molecule on the host cell plasma membrane, which triggers the F glycoprotein to undergo major conformational rearrangements, promoting viral entry. Subsequent fusion of the viral and host membranes allows delivery of the viral genetic material into the host cell. The intermediate states in viral entry are transient and thermodynamically unstable, making it impossible to understand these transitions using standard methods, yet understanding these transition states is important for expanding our knowledge of the viral entry process. In this study, we use cryo-electron tomography (cryo-ET) to dissect the stepwise process by which the receptor-binding protein triggers F-mediated fusion, when forming a complex with receptor-bearing membranes. Using an on-grid antibody capture method that facilitates examination of fresh, biologically active strains of virus directly from supernatant fluids and a series of biological tools that permit the capture of intermediate states in the fusion process, we visualize the series of events that occur when a pristine, authentic viral particle interacts with target receptors and proceeds from the viral entry steps of receptor engagement to membrane fusion.


Subject(s)
Cell Membrane/metabolism , HN Protein/metabolism , Parainfluenza Virus 3, Human/metabolism , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Cell Membrane/ultrastructure , Chlorocebus aethiops , Humans , Parainfluenza Virus 3, Human/ultrastructure , Vero Cells
5.
J Am Chem Soc ; 143(15): 5958-5966, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33825470

ABSTRACT

The lower respiratory tract infections affecting children worldwide are in large part caused by the parainfluenza viruses (HPIVs), particularly HPIV3, along with human metapneumovirus and respiratory syncytial virus, enveloped negative-strand RNA viruses. There are no vaccines for these important human pathogens, and existing treatments have limited or no efficacy. Infection by HPIV is initiated by viral glycoprotein-mediated fusion between viral and host cell membranes. A viral fusion protein (F), once activated in proximity to a target cell, undergoes a series of conformational changes that first extend the trimer subunits to allow insertion of the hydrophobic domains into the target cell membrane and then refold the trimer into a stable postfusion state, driving the merger of the viral and host cell membranes. Lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F inhibit infection by interfering with the structural transitions of the trimeric F assembly. Clinical application of this strategy, however, requires improving the in vivo stability of antiviral peptides. We show that the HRC peptide backbone can be modified via partial replacement of α-amino acid residues with ß-amino acid residues to generate α/ß-peptides that retain antiviral activity but are poor protease substrates. Relative to a conventional α-lipopeptide, our best α/ß-lipopeptide exhibits improved persistence in vivo and improved anti-HPIV3 antiviral activity in animals.


Subject(s)
Lipopeptides/pharmacology , Parainfluenza Virus 3, Human/drug effects , Respiratory Tract Infections/pathology , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Line , Cholesterol/chemistry , Drug Design , Humans , Lipopeptides/chemistry , Lipopeptides/metabolism , Parainfluenza Virus 3, Human/isolation & purification , Protein Multimerization , Rats , Respiratory Tract Infections/virology , Tissue Distribution , Transition Temperature , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects
6.
J Am Chem Soc ; 142(5): 2140-2144, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31951396

ABSTRACT

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) are leading causes of lower respiratory tract infections. There are currently no vaccines or antiviral therapeutics to treat HPIV3 or RSV infections. We recently reported a peptide (VIQKI), derived from the C-terminal heptad repeat (HRC) domain of the HPIV3 fusion (F) glycoprotein that inhibits infection by both HPIV3 and RSV. The dual inhibitory activity of VIQKI is due to its unique ability to bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F, thereby preventing the native HRN-HRC interactions required for viral entry. Here we describe the structure-guided design of dual inhibitors of HPIV3 and RSV fusion with improved efficacy. We show that VIQKI derivatives possessing one (I456F) or two (I454F/I456F) phenylalanine substitutions near the N-terminus exhibit more stable assemblies with the RSV-HRN domain and enhanced antiviral efficacy against both HPIV3 and RSV infection. Cocrystal structures of the new Phe-substituted inhibitors coassembled with HPIV3 or RSV-HRN domains reveal that the I456F substitution makes intimate hydrophobic contact with the core trimers of both HPIV3 and RSV F.


Subject(s)
Antiviral Agents/pharmacology , Oligopeptides/pharmacology , Parainfluenza Virus 3, Human/drug effects , Respiratory Syncytial Virus, Human/drug effects , Virus Internalization/drug effects , Amino Acid Sequence , Antiviral Agents/chemistry , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Molecular Structure , Oligopeptides/chemistry , Parainfluenza Virus 3, Human/physiology , Protein Conformation , Respiratory Syncytial Virus, Human/physiology
7.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30487282

ABSTRACT

During a measles virus (MeV) epidemic in 2009 in South Africa, measles inclusion body encephalitis (MIBE) was identified in several HIV-infected patients. Years later, children are presenting with subacute sclerosing panencephalitis (SSPE). To investigate the features of established MeV neuronal infections, viral sequences were analyzed from brain tissue samples of a single SSPE case and compared with MIBE sequences previously obtained from patients infected during the same epidemic. Both the SSPE and the MIBE viruses had amino acid substitutions in the ectodomain of the F protein that confer enhanced fusion properties. Functional analysis of the fusion complexes confirmed that both MIBE and SSPE F protein mutations promoted fusion with less dependence on interaction by the viral receptor-binding protein with known MeV receptors. While the SSPE F required the presence of a homotypic attachment protein, MeV H, in order to fuse, MIBE F did not. Both F proteins had decreased thermal stability compared to that of the corresponding wild-type F protein. Finally, recombinant viruses expressing MIBE or SSPE fusion complexes spread in the absence of known MeV receptors, with MIBE F-bearing viruses causing large syncytia in these cells. Our results suggest that alterations to the MeV fusion complex that promote fusion and cell-to-cell spread in the absence of known MeV receptors is a key property for infection of the brain.IMPORTANCE Measles virus can invade the central nervous system (CNS) and cause severe neurological complications, such as MIBE and SSPE. However, mechanisms by which MeV enters the CNS and triggers the disease remain unclear. We analyzed viruses from brain tissue of individuals with MIBE or SSPE, infected during the same epidemic, after the onset of neurological disease. Our findings indicate that the emergence of hyperfusogenic MeV F proteins is associated with infection of the brain. We also demonstrate that hyperfusogenic F proteins permit MeV to enter cells and spread without the need to engage nectin-4 or CD150, known receptors for MeV that are not present on neural cells.


Subject(s)
Measles virus/genetics , Subacute Sclerosing Panencephalitis/genetics , Viral Fusion Proteins/genetics , Amino Acid Substitution , Animals , Brain/virology , Cell Adhesion Molecules/metabolism , Chlorocebus aethiops , Epidemics , Female , Genotype , Giant Cells/virology , HEK293 Cells , Humans , Male , Measles/epidemiology , Measles/metabolism , Measles/virology , Mutation , Neurons/virology , South Africa , Subacute Sclerosing Panencephalitis/virology , Vero Cells , Viral Fusion Proteins/metabolism
8.
J Virol ; 93(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30728259

ABSTRACT

A clinical isolate of measles virus (MeV) bearing a single amino acid alteration in the viral fusion protein (F; L454W) was previously identified in two patients with lethal sequelae of MeV central nervous system (CNS) infection. The mutation dysregulated the viral fusion machinery so that the mutated F protein mediated cell fusion in the absence of known MeV cellular receptors. While this virus could feasibly have arisen via intrahost evolution of the wild-type (wt) virus, it was recently shown that the same mutation emerged under the selective pressure of small-molecule antiviral treatment. Under these conditions, a potentially neuropathogenic variant emerged outside the CNS. While CNS adaptation of MeV was thought to generate viruses that are less fit for interhost spread, we show that two animal models can be readily infected with CNS-adapted MeV via the respiratory route. Despite bearing a fusion protein that is less stable at 37°C than the wt MeV F, this virus infects and replicates in cotton rat lung tissue more efficiently than the wt virus and is lethal in a suckling mouse model of MeV encephalitis even with a lower inoculum. Thus, either during lethal MeV CNS infection or during antiviral treatment in vitro, neuropathogenic MeV can emerge, can infect new hosts via the respiratory route, and is more pathogenic (at least in these animal models) than wt MeV.IMPORTANCE Measles virus (MeV) infection can be severe in immunocompromised individuals and lead to complications, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE) occur even in the face of an intact immune response. While they are relatively rare complications of MeV infection, MIBE and SSPE are lethal. This work addresses the hypothesis that despite a dysregulated viral fusion complex, central nervous system (CNS)-adapted measles virus can spread outside the CNS within an infected host.


Subject(s)
Central Nervous System/virology , Encephalitis, Viral , Inclusion Bodies, Viral , Lung/virology , Measles virus/physiology , Measles , Mutation, Missense , Viral Fusion Proteins , Virus Replication , Amino Acid Substitution , Animals , Central Nervous System/metabolism , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/genetics , Encephalitis, Viral/metabolism , Encephalitis, Viral/transmission , Humans , Inclusion Bodies, Viral/genetics , Inclusion Bodies, Viral/metabolism , Lung/metabolism , Measles/metabolism , Measles/transmission , Mice , Mice, Transgenic , Sigmodontinae , Vero Cells , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
10.
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
11.
J Infect Dis ; 218(2): 218-227, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29566184

ABSTRACT

Background: The emerging zoonotic paramyxovirus Nipah virus (NiV) causes severe respiratory and neurological disease in humans, with high fatality rates. Nipah virus can be transmitted via person-to-person contact, posing a high risk for epidemic outbreaks. However, a broadly applicable approach for human NiV outbreaks in field settings is lacking. Methods: We engineered new antiviral lipopeptides and analyzed in vitro fusion inhibition to identify an optimal candidate for prophylaxis of NiV infection in the lower respiratory tract, and we assessed antiviral efficiency in 2 different animal models. Results: We show that lethal NiV infection can be prevented with lipopeptides delivered via the respiratory route in both hamsters and nonhuman primates. By targeting retention of peptides for NiV prophylaxis in the respiratory tract, we avoid its systemic delivery in individuals who need only prevention, and thus we increase the safety of treatment and enhance utility of the intervention. Conclusions: The experiments provide a proof of concept for the use of antifusion lipopeptides for prophylaxis of lethal NiV. These results advance the goal of rational development of potent lipopeptide inhibitors with desirable pharmacokinetic and biodistribution properties and a safe effective delivery method to target NiV and other pathogenic viruses.


Subject(s)
Chemoprevention/methods , Henipavirus Infections/prevention & control , Lipopeptides/administration & dosage , Nipah Virus/physiology , Primate Diseases/prevention & control , Viral Envelope Proteins/antagonists & inhibitors , Viral Fusion Protein Inhibitors/administration & dosage , Animals , Bronchopneumonia/prevention & control , Bronchopneumonia/veterinary , Chlorocebus aethiops , Disease Models, Animal , Female , Humans , Male , Mesocricetus
12.
Molecules ; 22(7)2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28714870

ABSTRACT

A set of lipopeptides was recently reported for their broad-spectrum antiviral activity against viruses belonging to the Paramyxoviridae family, including human parainfluenza virus type 3 and Nipah virus. Among them, the peptide with a 24-unit PEG linker connecting it to a cholesterol moiety (VG-PEG24-Chol) was found to be the best membrane fusion inhibitory peptide. Here, we evaluated the interaction of the same set of peptides with biomembrane model systems and isolated human peripheral blood mononuclear cells (PBMC). VG-PEG24-Chol showed the highest insertion rate and it was among the peptides that induced a larger change on the surface pressure of cholesterol rich membranes. This peptide also displayed a high affinity towards PBMC membranes. These data provide new information about the dynamics of peptide-membrane interactions of a specific group of antiviral peptides, known for their potential as multipotent paramyxovirus antivirals.


Subject(s)
Antiviral Agents/chemistry , Cell Membrane/chemistry , Lipopeptides/chemistry , Polyethylene Glycols/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol/chemistry , Humans , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Lipopeptides/metabolism , Lipopeptides/pharmacology , Liposomes/chemistry , Paramyxovirinae/chemistry , Structure-Activity Relationship
13.
J Virol ; 88(22): 13495-502, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25210187

ABSTRACT

UNLABELLED: Human parainfluenza viruses (HPIVs) cause widespread respiratory infections, with no vaccines or effective treatments. We show that the molecular determinants for HPIV3 growth in vitro are fundamentally different from those required in vivo and that these differences impact inhibitor susceptibility. HPIV infects its target cells by coordinated action of the hemagglutinin-neuraminidase receptor-binding protein (HN) and the fusion envelope glycoprotein (F), which together comprise the molecular fusion machinery; upon receptor engagement by HN, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. Peptides derived from key regions of F can potently inhibit HPIV infection at the entry stage, by interfering with the structural transition of F. We show that clinically circulating viruses have fusion machinery that is more stable and less readily activated than viruses adapted to growth in culture. Fusion machinery that is advantageous for growth in human airway epithelia and in vivo confers susceptibility to peptide fusion inhibitors in the host lung tissue or animal, but the same fusion inhibitors have no effect on viruses whose fusion glycoproteins are suited for growth in vitro. We propose that for potential clinical efficacy, antivirals should be evaluated using clinical isolates in natural host tissue rather than lab strains of virus in cultured cells. The unique susceptibility of clinical strains in human tissues reflects viral inhibition in vivo. IMPORTANCE: Acute respiratory infection is the leading cause of mortality in young children under 5 years of age, causing nearly 20% of childhood deaths worldwide each year. The paramyxoviruses, including human parainfluenza viruses (HPIVs), cause a large share of these illnesses. There are no vaccines or drugs for the HPIVs. Inhibiting entry of viruses into the human cell is a promising drug strategy that blocks the first step in infection. To develop antivirals that inhibit entry, it is critical to understand the first steps of infection. We found that clinical viruses isolated from patients have very different entry properties from those of the viruses generally studied in laboratories. The viral entry mechanism is less active and more sensitive to fusion inhibitory molecules. We propose that to interfere with viral infection, we test clinically circulating viruses in natural tissues, to develop antivirals against respiratory disease caused by HPIVs.


Subject(s)
Parainfluenza Virus 3, Human/physiology , Virus Internalization , Animals , Cells, Cultured , Female , HN Protein/metabolism , Humans , Sigmodontinae , Viral Fusion Proteins/metabolism
14.
J Virol ; 87(24): 13619-27, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109225

ABSTRACT

Paramyxoviruses, including the human pathogen measles virus (MV) and the avian Newcastle disease virus (NDV), enter host cells through fusion of the viral envelope with the target cell membrane. This fusion is driven by the concerted action of two viral envelope glycoproteins: the receptor binding protein and the fusion protein (F). The MV receptor binding protein (hemagglutinin [H]) attaches to proteinaceous receptors on host cells, while the receptor binding protein of NDV (hemagglutinin-neuraminidase [HN]) interacts with sialic acid-containing receptors. The receptor-bound HN/H triggers F to undergo conformational changes that render it competent to mediate fusion of the viral and cellular membranes. The mechanism of fusion activation has been proposed to be different for sialic acid-binding viruses and proteinaceous receptor-binding viruses. We report that a chimeric protein containing the NDV HN receptor binding region and the MV H stalk domain can activate MV F to fuse, suggesting that the signal to the stalk of a protein-binding receptor binding molecule can be transmitted from a sialic acid binding domain. By engineering the NDV HN globular domain to interact with a proteinaceous receptor, the fusion activation signal was preserved. Our findings are consistent with a unified mechanism of fusion activation, at least for the Paramyxovirinae subfamily, in which the receptor binding domains of the receptor binding proteins are interchangeable and the stalk determines the specificity of F activation.


Subject(s)
HN Protein/metabolism , Hemagglutinins/metabolism , Measles virus/metabolism , Measles/virology , N-Acetylneuraminic Acid/metabolism , Newcastle Disease/virology , Newcastle disease virus/metabolism , Animals , Birds , Cell Line , HN Protein/chemistry , HN Protein/genetics , Hemagglutinins/chemistry , Hemagglutinins/genetics , Humans , Measles/genetics , Measles/metabolism , Measles virus/chemistry , Measles virus/genetics , Newcastle Disease/genetics , Newcastle Disease/metabolism , Newcastle disease virus/chemistry , Newcastle disease virus/genetics , Protein Binding , Protein Structure, Tertiary , Receptors, Virus/genetics , Receptors, Virus/metabolism , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization
15.
J Virol ; 87(24): 13785-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109233

ABSTRACT

Measles virus (MV) infection causes an acute childhood disease that can include infection of the central nervous system and can rarely progress to severe neurological disease for which there is no specific treatment. We generated potent antiviral peptide inhibitors of MV entry and spreading and MV-induced cell fusion. Dimers of MV-specific peptides derived from the C-terminal heptad repeat region of the MV fusion protein, conjugated to cholesterol, efficiently protect SLAM transgenic mice from fatal MV infection. Fusion inhibitors hold promise for the prophylaxis of MV infection in unvaccinated and immunocompromised people, as well as potential for the treatment of grave neurological complications of measles.


Subject(s)
Antiviral Agents/pharmacology , Brain/virology , Measles virus/drug effects , Measles/prevention & control , Viral Fusion Proteins/antagonists & inhibitors , Animals , Brain/drug effects , Cell Line , Humans , Measles/drug therapy , Measles/mortality , Measles/virology , Measles virus/genetics , Measles virus/physiology , Mice , Mice, Transgenic , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects
16.
J Virol ; 87(20): 10980-96, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903846

ABSTRACT

Paramyxoviruses, including the emerging lethal human Nipah virus (NiV) and the avian Newcastle disease virus (NDV), enter host cells through fusion of the viral and target cell membranes. For paramyxoviruses, membrane fusion is the result of the concerted action of two viral envelope glycoproteins: a receptor binding protein and a fusion protein (F). The NiV receptor binding protein (G) attaches to ephrin B2 or B3 on host cells, whereas the corresponding hemagglutinin-neuraminidase (HN) attachment protein of NDV interacts with sialic acid moieties on target cells through two regions of its globular domain. Receptor-bound G or HN via its stalk domain triggers F to undergo the conformational changes that render it competent to mediate fusion of the viral and cellular membranes. We show that chimeric proteins containing the NDV HN receptor binding regions and the NiV G stalk domain require a specific sequence at the connection between the head and the stalk to activate NiV F for fusion. Our findings are consistent with a general mechanism of paramyxovirus fusion activation in which the stalk domain of the receptor binding protein is responsible for F activation and a specific connecting region between the receptor binding globular head and the fusion-activating stalk domain is required for transmitting the fusion signal.


Subject(s)
Nipah Virus/physiology , Viral Envelope Proteins/metabolism , Virus Internalization , Cell Line , DNA Mutational Analysis , Humans , Newcastle disease virus/genetics , Nipah Virus/genetics , Protein Interaction Mapping , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viral Envelope Proteins/genetics
17.
Nat Microbiol ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858594

ABSTRACT

Human parainfluenza virus type 3 (hPIV3) is a respiratory pathogen that can cause severe disease in older people and infants. Currently, vaccines against hPIV3 are in clinical trials but none have been approved yet. The haemagglutinin-neuraminidase (HN) and fusion (F) surface glycoproteins of hPIV3 are major antigenic determinants. Here we describe naturally occurring potently neutralizing human antibodies directed against both surface glycoproteins of hPIV3. We isolated seven neutralizing HN-reactive antibodies and a pre-fusion conformation F-reactive antibody from human memory B cells. One HN-binding monoclonal antibody (mAb), designated PIV3-23, exhibited functional attributes including haemagglutination and neuraminidase inhibition. We also delineated the structural basis of neutralization for two HN and one F mAbs. MAbs that neutralized hPIV3 in vitro protected against infection and disease in vivo in a cotton rat model of hPIV3 infection, suggesting correlates of protection for hPIV3 and the potential clinical utility of these mAbs.

18.
Science ; 384(6703): eadm8693, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38935733

ABSTRACT

Measles virus (MeV) presents a public health threat that is escalating as vaccine coverage in the general population declines and as populations of immunocompromised individuals, who cannot be vaccinated, increase. There are no approved therapeutics for MeV. Neutralizing antibodies targeting viral fusion are one potential therapeutic approach but have not yet been structurally characterized or advanced to clinical use. We present cryo-electron microscopy (cryo-EM) structures of prefusion F alone [2.1-angstrom (Å) resolution], F complexed with a fusion-inhibitory peptide (2.3-Å resolution), F complexed with the neutralizing and protective monoclonal antibody (mAb) 77 (2.6-Å resolution), and an additional structure of postfusion F (2.7-Å resolution). In vitro assays and examination of additional EM classes show that mAb 77 binds prefusion F, arrests F in an intermediate state, and prevents transition to the postfusion conformation. These structures shed light on antibody-mediated neutralization that involves arrest of fusion proteins in an intermediate state.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Measles virus , Viral Fusion Proteins , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/chemistry , Measles virus/immunology , Measles virus/chemistry , Viral Fusion Proteins/immunology , Viral Fusion Proteins/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/chemistry , Humans , Protein Conformation
19.
J Biol Chem ; 287(1): 778-793, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22110138

ABSTRACT

Parainfluenza viruses enter host cells by fusing the viral and target cell membranes via concerted action of their two envelope glycoproteins: the hemagglutinin-neuraminidase (HN) and the fusion protein (F). Receptor-bound HN triggers F to undergo conformational changes that render it fusion-competent. To address the role of receptor engagement and to elucidate how HN and F interact during the fusion process, we used bimolecular fluorescence complementation to follow the dynamics of human parainfluenza virus type 3 (HPIV3) HN/F pairs in living cells. We show that HN and F associate before receptor engagement. HN drives the formation of HN-F clusters at the site of fusion, and alterations in HN-F interaction determine the fusogenicity of the glycoprotein pair. An interactive site, at the HN dimer interface modulates HN fusion activation property, which is critical for infection of the natural host. This first evidence for the sequence of initial events that lead to viral entry may indicate a new paradigm for understanding Paramyxovirus infection.


Subject(s)
Hemagglutinins, Viral/metabolism , Neuraminidase/metabolism , Parainfluenza Virus 3, Human/physiology , Viral Fusion Proteins/metabolism , Virus Internalization , HEK293 Cells , Hemagglutinins, Viral/chemistry , Hemagglutinins, Viral/genetics , Humans , Models, Molecular , Molecular Imaging , Mutation , Neuraminidase/chemistry , Neuraminidase/genetics , Parainfluenza Virus 3, Human/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Receptors, Cell Surface/metabolism , Spectrometry, Fluorescence , Substrate Specificity
20.
J Virol ; 86(10): 5730-41, 2012 May.
Article in English | MEDLINE | ID: mdl-22438532

ABSTRACT

The hemagglutinin-neuraminidase (HN) protein of paramyxoviruses carries out three distinct activities contributing to the ability of HN to promote viral fusion and entry: receptor binding, receptor cleavage (neuraminidase), and activation of the fusion protein. The relationship between receptor binding and fusion triggering functions of HN are not fully understood. For Newcastle disease virus (NDV), one bifunctional site (site I) on HN's globular head can mediate both receptor binding and neuraminidase activities, and a second site (site II) in the globular head is also capable of mediating receptor binding. The receptor analog, zanamivir, blocks receptor binding and cleavage activities of NDV HN's site I while activating receptor binding by site II. Comparison of chimeric proteins in which the globular head of NDV HN is connected to the stalk region of either human parainfluenza virus type 3 (HPIV3) or Nipah virus receptor binding proteins indicates that receptor binding to NDV HN site II not only can activate its own fusion (F) protein but can also activate the heterotypic fusion proteins. We suggest a general model for paramyxovirus fusion activation in which receptor engagement at site II plays an active role in F activation.


Subject(s)
HN Protein/metabolism , Newcastle disease virus/physiology , Paramyxoviridae Infections/metabolism , Paramyxovirinae/metabolism , Receptors, Virus/metabolism , Viral Proteins/metabolism , Virus Internalization , Binding Sites , Carrier Proteins , Cell Line , HN Protein/chemistry , HN Protein/genetics , Humans , Newcastle disease virus/chemistry , Newcastle disease virus/enzymology , Newcastle disease virus/genetics , Paramyxoviridae Infections/virology , Paramyxovirinae/genetics , Protein Structure, Tertiary , Viral Fusion Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL