Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.069
Filter
1.
Nat Commun ; 15(1): 5458, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937429

ABSTRACT

Respirovirus 3 is a leading cause of severe acute respiratory infections in vulnerable human populations. Entry into host cells is facilitated by the attachment glycoprotein and the fusion glycoprotein (F). Because of its crucial role, F represents an attractive therapeutic target. Here, we identify 13 F-directed heavy-chain-only antibody fragments that neutralize recombinant respirovirus 3. High-resolution cryo-EM structures of antibody fragments bound to the prefusion conformation of F reveal three distinct, previously uncharacterized epitopes. All three antibody fragments bind quaternary epitopes on F, suggesting mechanisms for neutralization that may include stabilization of the prefusion conformation. Studies in cotton rats demonstrate the prophylactic efficacy of these antibody fragments in reducing viral load in the lungs and nasal passages. These data highlight the potential of heavy-chain-only antibody fragments as effective interventions against respirovirus 3 infection and identify neutralizing epitopes that can be targeted for therapeutic development.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Epitopes , Animals , Antibodies, Neutralizing/immunology , Humans , Antibodies, Viral/immunology , Epitopes/immunology , Sigmodontinae , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Viral Fusion Proteins/immunology , Viral Fusion Proteins/chemistry , Female , Camelus/immunology , Camelus/virology
2.
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
3.
Nat Commun ; 15(1): 4629, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821950

ABSTRACT

The Paramyxoviridae family encompasses medically significant RNA viruses, including human respiroviruses 1 and 3 (RV1, RV3), and zoonotic pathogens like Nipah virus (NiV). RV3, previously known as parainfluenza type 3, for which no vaccines or antivirals have been approved, causes respiratory tract infections in vulnerable populations. The RV3 fusion (F) protein is inherently metastable and will likely require prefusion (preF) stabilization for vaccine effectiveness. Here we used structure-based design to stabilize regions involved in structural transformation to generate a preF protein vaccine antigen with high expression and stability, and which, by stabilizing the coiled-coil stem region, does not require a heterologous trimerization domain. The preF candidate induces strong neutralizing antibody responses in both female naïve and pre-exposed mice and provides protection in a cotton rat challenge model (female). Despite the evolutionary distance of paramyxovirus F proteins, their structural transformation and local regions of instability are conserved, which allows successful transfer of stabilizing substitutions to the distant preF proteins of RV1 and NiV. This work presents a successful vaccine antigen design for RV3 and provides a toolbox for future paramyxovirus vaccine design and pandemic preparedness.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Sigmodontinae , Viral Fusion Proteins , Viral Vaccines , Animals , Female , Viral Fusion Proteins/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/chemistry , Mice , Viral Vaccines/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Humans , Mice, Inbred BALB C , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/genetics
4.
Sci Rep ; 14(1): 11823, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38783052

ABSTRACT

Our previous findings indicated that many respiratory syncytial virus (RSV) isolates are unstable at 4 °C compared to 20 °C. Some of the strains completely lose infectivity after 24 h at 4 °C. This study analyzed the inactivation process at 4 °C using a representative strain, RSV/Sendai/851/13. After 24 h of storage at 4 °C, the virus was completely inactivated but retained its ability to attach to and to be taken into host cells. It suggested a reduced fusion ability between the viral and cellular membranes. During storage at 4 °C, the RSV fusion (F) protein underwent a conformational change and was no longer recognized by pre-fusion form-specific antibodies. When the RSV/Sendai/851/13 strain was passaged at 4 °C, a variant with an amino acid substitution, I148T, in the F protein fusion peptide was selected. Also, an amino acid change in G protein demonstrating stability at low temperatures was obtained. These results show that the inactivation of RSV at 4 °C is due to the loss of membrane fusion activity in the F protein, which cannot maintain its pre-fusion state at 4 °C.


Subject(s)
Cold Temperature , Respiratory Syncytial Virus, Human , Viral Fusion Proteins , Virus Inactivation , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/genetics , Viral Fusion Proteins/chemistry , Humans , Respiratory Syncytial Virus, Human/physiology , Animals , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses
5.
Nat Commun ; 15(1): 4330, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773072

ABSTRACT

The Hendra and Nipah viruses (HNVs) are highly pathogenic pathogens without approved interventions for human use. In addition, the interaction pattern between the attachment (G) and fusion (F) glycoproteins required for virus entry remains unclear. Here, we isolate a panel of Macaca-derived G-specific antibodies that cross-neutralize HNVs via multiple mechanisms. The most potent antibody, 1E5, confers adequate protection against the Nipah virus challenge in female hamsters. Crystallography demonstrates that 1E5 has a highly similar binding pattern to the receptor. In cryo-electron microscopy studies, the tendency of 1E5 to bind to the upper or lower heads results in two distinct quaternary structures of G. Furthermore, we identify the extended outer loop ß1S2-ß1S3 of G and two pockets on the apical region of fusion (F) glycoprotein as the essential sites for G-F interactions. This work highlights promising drug candidates against HNVs and contributes deeper insights into the viruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Henipavirus Infections , Viral Fusion Proteins , Animals , Antibodies, Neutralizing/immunology , Female , Antibodies, Viral/immunology , Henipavirus Infections/virology , Henipavirus Infections/immunology , Viral Fusion Proteins/immunology , Viral Fusion Proteins/chemistry , Humans , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Nipah Virus/immunology , Virus Internalization/drug effects , Henipavirus/immunology , Cricetinae , Cross Reactions/immunology , Hendra Virus/immunology , Macaca , Mesocricetus , Crystallography, X-Ray
6.
Nat Commun ; 15(1): 1173, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38332002

ABSTRACT

Respiratory syncytial virus (RSV) is a common cause of acute lower respiratory tract infection in infants, older adults and the immunocompromised. Effective directly acting antivirals are not yet available for clinical use. To address this, we screen the ReFRAME drug-repurposing library consisting of 12,000 small molecules against RSV. We identify 21 primary candidates including RSV F and N protein inhibitors, five HSP90 and four IMPDH inhibitors. We select lonafarnib, a licensed farnesyltransferase inhibitor, and phase III candidate for hepatitis delta virus (HDV) therapy, for further follow-up. Dose-response analyses and plaque assays confirm the antiviral activity (IC50: 10-118 nM). Passaging of RSV with lonafarnib selects for phenotypic resistance and fixation of mutations in the RSV fusion protein (T335I and T400A). Lentiviral pseudotypes programmed with variant RSV fusion proteins confirm that lonafarnib inhibits RSV cell entry and that these mutations confer lonafarnib resistance. Surface plasmon resonance reveals RSV fusion protein binding of lonafarnib and co-crystallography identifies the lonafarnib binding site within RSV F. Oral administration of lonafarnib dose-dependently reduces RSV virus load in a murine infection model using female mice. Collectively, this work provides an overview of RSV drug repurposing candidates and establishes lonafarnib as a bona fide fusion protein inhibitor.


Subject(s)
Dibenzocycloheptenes , Pyridines , Respiratory Syncytial Virus Infections , Animals , Female , Mice , Drug Repositioning , Piperidines/pharmacology , Piperidines/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Viral Fusion Proteins/genetics , Viral Fusion Proteins/chemistry
7.
J Virol ; 97(12): e0134323, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37975688

ABSTRACT

IMPORTANCE: Vaccinia virus infection requires virus-cell membrane fusion to complete entry during endocytosis; however, it contains a large viral fusion protein complex of 11 viral proteins that share no structure or sequence homology to all the known viral fusion proteins, including type I, II, and III fusion proteins. It is thus very challenging to investigate how the vaccinia fusion complex works to trigger membrane fusion with host cells. In this study, we crystallized the ectodomain of vaccinia H2 protein, one component of the viral fusion complex. Furthermore, we performed a series of mutational, biochemical, and molecular analyses and identified two surface loops containing 170LGYSG174 and 125RRGTGDAW132 as the A28-binding region. We also showed that residues in the N-terminal helical region (amino acids 51-90) are also important for H2 function.


Subject(s)
Membrane Fusion , Vaccinia virus , Viral Fusion Proteins , Virus Internalization , Vaccinia virus/chemistry , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
8.
J Virol ; 97(10): e0092923, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37737588

ABSTRACT

IMPORTANCE: Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in infants, infecting all children by age 5. RSV also causes substantial morbidity and mortality in older adults, and a vaccine for older adults based on a prefusion-stabilized form of the viral F glycoprotein was recently approved by the FDA. Here, we investigate a set of antibodies that belong to the same public clonotype and were isolated from individuals vaccinated with a prefusion-stabilized RSV F protein. Our results reveal that these antibodies are highly potent and recognize a previously uncharacterized antigenic site on the prefusion F protein. Vaccination with prefusion RSV F proteins appears to boost the elicitation of these neutralizing antibodies, which are not commonly elicited by natural infection.


Subject(s)
Antibodies, Viral , Epitopes, B-Lymphocyte , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Vaccination , Viral Fusion Proteins , Humans , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism
9.
Annu Rev Virol ; 10(1): 139-161, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37774128

ABSTRACT

There are at least 21 families of enveloped viruses that infect mammals, and many contain members of high concern for global human health. All enveloped viruses have a dedicated fusion protein or fusion complex that enacts the critical genome-releasing membrane fusion event that is essential before viral replication within the host cell interior can begin. Because all enveloped viruses enter cells by fusion, it behooves us to know how viral fusion proteins function. Viral fusion proteins are also major targets of neutralizing antibodies, and hence they serve as key vaccine immunogens. Here we review current concepts about viral membrane fusion proteins focusing on how they are triggered, structural intermediates between pre- and postfusion forms, and their interplay with the lipid bilayers they engage. We also discuss cellular and therapeutic interventions that thwart virus-cell membrane fusion.


Subject(s)
Virus Internalization , Viruses , Animals , Humans , Viral Fusion Proteins/chemistry , Membrane Fusion , Viruses/genetics , Lipids , Mammals/metabolism
10.
Curr Opin Virol ; 61: 101337, 2023 08.
Article in English | MEDLINE | ID: mdl-37544710

ABSTRACT

Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) continue to be a global burden to infants, the elderly, and immunocompromised individuals. In the past ten years, there has been substantial progress in the development of new vaccine candidates and therapies against these viruses. These advancements were guided by the structural elucidation of the major surface glycoproteins for these viruses, the fusion (F) protein and attachment (G) protein. The identification of immunodominant epitopes on the RSV F and hMPV F proteins has expanded current knowledge on antibody-mediated immune responses, which has led to new approaches for vaccine and therapeutic development through the stabilization of pre-fusion constructs of the F protein and pre-fusion-specific monoclonal antibodies with high potency and efficacy. In this review, we describe structural characteristics of known antigenic sites on the RSV and hMPV proteins, their influence on the immune response, and current progress in vaccine and therapeutic development.


Subject(s)
Metapneumovirus , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Aged , Metapneumovirus/metabolism , Antibodies, Viral , Antibodies, Neutralizing , Viral Fusion Proteins/chemistry , Respiratory Syncytial Virus Infections/prevention & control
11.
Biomolecules ; 13(7)2023 07 14.
Article in English | MEDLINE | ID: mdl-37509166

ABSTRACT

Viral entry and fertilization are distinct biological processes that share a common mechanism: membrane fusion. In viral entry, enveloped viruses attach to the host cell membrane, triggering a series of conformational changes in the viral fusion proteins. This results in the exposure of a hydrophobic fusion peptide, which inserts into the host membrane and brings the viral and host membranes into close proximity. Subsequent structural rearrangements in opposing membranes lead to their fusion. Similarly, membrane fusion occurs when gametes merge during the fertilization process, though the exact mechanism remains unclear. Structural biology has played a pivotal role in elucidating the molecular mechanisms underlying membrane fusion. High-resolution structures of the viral and fertilization fusion-related proteins have provided valuable insights into the conformational changes that occur during this process. Understanding these mechanisms at a molecular level is essential for the development of antiviral therapeutics and tools to influence fertility. In this review, we will highlight the biological importance of membrane fusion and how protein structures have helped visualize both common elements and subtle divergences in the mechanisms behind fusion; in addition, we will examine the new tools that recent advances in structural biology provide researchers interested in a frame-by-frame understanding of membrane fusion.


Subject(s)
Membrane Fusion , Virus Diseases , Humans , Viral Fusion Proteins/chemistry , Antiviral Agents , Fertilization
12.
Sci China Life Sci ; 66(4): 729-742, 2023 04.
Article in English | MEDLINE | ID: mdl-36853487

ABSTRACT

Respiratory syncytial virus (RSV) continues to pose serious threats to pediatric populations due to the lack of a vaccine and effective antiviral drugs. RSV fusion (F) glycoprotein mediates viral-host membrane fusion and is a key target for neutralizing antibodies. We generated 23 full-human monoclonal antibodies (hmAbs) against prefusion F protein (pre-F) from a healthy adult with natural RSV infection by single B cell cloning technique. A highly potent RSV-neutralizing hmAb, named as 25-20, is selected, which targets a new site Ø-specific epitope. Site-directed mutagenesis and structural modelling analysis demonstrated that 25-20 mainly targets a highly conserved hydrophobic region located at the a4 helix and a1 helix of pre-F, indicating a site of vulnerability for drug and vaccine design. It is worth noting that 25-20 uses an unreported inferred germline (iGL) that binds very poorly to pre-F, thus high levels of somatic mutations are needed to gain high binding affinity with pre-F. Our observation helps to understand the evolution of RSV antibody during natural infection. Furthermore, by in silico prediction and experimental verification, we optimized 25-20 with KD values as low as picomolar range. Therefore, the optimized 25-20 represents an excellent candidate for passive protection against RSV infection.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Child , Humans , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/chemistry , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Neutralizing , Antibodies, Viral
14.
Sci Adv ; 9(6): eade2727, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36763666

ABSTRACT

Paramyxoviruses-including important pathogens like parainfluenza, measles, and Nipah viruses-use a receptor binding protein [hemagglutinin-neuraminidase (HN) for parainfluenza] and a fusion protein (F), acting in a complex, to enter cells. We use cryo-electron tomography to visualize the fusion complex of human parainfluenza virus 3 (HN/F) on the surface of authentic clinical viruses at a subnanometer resolution sufficient to answer mechanistic questions. An HN loop inserts in a pocket on F, showing how the fusion complex remains in a ready but quiescent state until activation. The globular HN heads are rotated with respect to each other: one downward to contact F, and the other upward to grapple cellular receptors, demonstrating how HN/F performs distinct steps before F activation. This depiction of viral fusion illuminates potentially druggable targets for paramyxoviruses and sheds light on fusion processes that underpin wide-ranging biological processes but have not been visualized in situ or at the present resolution.


Subject(s)
Paramyxoviridae Infections , Viral Fusion Proteins , Humans , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , HN Protein/chemistry , HN Protein/metabolism , Receptors, Cell Surface , Virus Internalization
15.
PLoS Comput Biol ; 19(1): e1010360, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36626370

ABSTRACT

Human respiratory syncytial virus (RSV) is a major cause of lower respiratory infection. Despite more than 60 years of research, there is no licensed vaccine. While B cell response is a major focus for vaccine design, the T cell epitope profile of RSV is also important for vaccine development. Here, we computationally predicted putative T cell epitopes in the Fusion protein (F) and Glycoprotein (G) of RSV wild circulating strains by predicting Major Histocompatibility Complex (MHC) class I and class II binding affinity. We limited our inferences to conserved epitopes in both F and G proteins that have been experimentally validated. We applied multidimensional scaling (MDS) to construct T cell epitope landscapes to investigate the diversity and evolution of T cell profiles across different RSV strains. We find the RSV strains are clustered into three RSV-A groups and two RSV-B groups on this T epitope landscape. These clusters represent divergent RSV strains with potentially different immunogenic profiles. In addition, our results show a greater proportion of F protein T cell epitope content conservation among recent epidemic strains, whereas the G protein T cell epitope content was decreased. Importantly, our results suggest that RSV-A and RSV-B have different patterns of epitope drift and replacement and that RSV-B vaccines may need more frequent updates. Our study provides a novel framework to study RSV T cell epitope evolution. Understanding the patterns of T cell epitope conservation and change may be valuable for vaccine design and assessment.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Epitopes, T-Lymphocyte , Viral Fusion Proteins/chemistry , Antibodies, Viral
16.
Sci Transl Med ; 14(676): eade0424, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36542692

ABSTRACT

There is currently no licensed vaccine for respiratory syncytial virus (RSV). Here, we assess the effect of RSV fusion protein (F) conformation on B cell responses in a post hoc comparison of samples from the DS-Cav1 [prefusion (pre-F)] and MEDI7510 [postfusion (post-F)] vaccine clinical trials. We compared the magnitude and quality of the serological and B cell responses across time points and vaccines. We measured RSV A and B neutralization, F-binding immunoglobulin G titers, and competition assays at week 0 (before vaccination) and week 4 (after vaccination) to evaluate antibody specificity and potency. To compare B cell specificity and activation, we used pre-F and post-F probes in tandem with a 17-color immunophenotyping flow cytometry panel at week 0 (before vaccination) and week 1 (after vaccination). Our data demonstrate that both DS-Cav1 and MEDI7510 vaccination robustly elicit F-specific antibodies and B cells, but DS-Cav1 elicited antibodies that more potently neutralized both RSV A and B. The superior potency was mediated by antibodies that bind antigenic sites on the apex of pre-F that are not present on post-F. In the memory (CD27+) B cell compartment, vaccination with DS-Cav1 or MEDI7510 elicited B cells with different epitope specificities. B cells preferentially binding the pre-F probe were activated in DS-Cav1-vaccinated participants but not in MEDI7510-vaccinated participants. Our findings emphasize the importance of using pre-F as an immunogen in humans because of its deterministic role in eliciting highly potent neutralizing antibodies and memory B cells.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Humans , Antibodies, Viral , Viral Fusion Proteins/chemistry , Antibodies, Neutralizing , Antigens , Vaccines, Subunit , Respiratory Syncytial Virus Infections/prevention & control
17.
Nat Commun ; 13(1): 4772, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35970990

ABSTRACT

Chikungunya virus (CHIKV) is a human pathogen that delivers its genome to the host cell cytoplasm through endocytic low pH-activated membrane fusion mediated by class-II fusion proteins. Though structures of prefusion, icosahedral CHIKV are available, structural characterization of virion interaction with membranes has been limited. Here, we have used cryo-electron tomography to visualize CHIKV's complete membrane fusion pathway, identifying key intermediary glycoprotein conformations coupled to membrane remodeling events. Using sub-tomogram averaging, we elucidate features of the low pH-exposed virion, nucleocapsid and full-length E1-glycoprotein's post-fusion structure. Contrary to class-I fusion systems, CHIKV achieves membrane apposition by protrusion of extended E1-glycoprotein homotrimers into the target membrane. The fusion process also features a large hemifusion diaphragm that transitions to a wide pore for intact nucleocapsid delivery. Our analyses provide comprehensive ultrastructural insights into the class-II virus fusion system function and direct mechanistic characterization of the fundamental process of protein-mediated membrane fusion.


Subject(s)
Chikungunya virus , Virus Internalization , Chikungunya virus/genetics , Glycoproteins/analysis , Humans , Membrane Fusion , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Virion/metabolism
18.
Viral Immunol ; 35(8): 559-565, 2022 10.
Article in English | MEDLINE | ID: mdl-35944261

ABSTRACT

Antimicrobial peptides are proteins that have been found to be an important factor in the natural immune response to a variety of pathogens. Respiratory syncytial virus (RSV) is a respiratory pathogen with the capability to cause serious upper and lower respiratory infections in infants and children and is a major viral cause of infant mortality. There is currently no functional vaccine for the virus, as recent efforts have been hindered by the virus's low immunogenicity, its ability to effectively mutate, and underlying instabilities of potential vaccines. Previous studies have shown that antimicrobial peptides may affect viral replication and spread of RSV. Our study evaluates the susceptibility of chimeric strains of RSV that express different fusion (F) and attachment (G) proteins to susceptibilities to inactivation by LL-37 and human beta-defensins (hBDs) hBD-1, hBD-3, and hBD-4. We show that LL-37 and hBD-3 result in dose-dependent, strain-independent inactivation of RSV, whereas treatment with either hBD-1 or hBD-4 appears more variable between strains. This suggests a potential role of the viral structural proteins in mitigating the inhibitory effects of the peptides. This study provides the first evidence of the sensitivity of RSV to several hBDs and indicates a role of LL-37 and beta-defensins in both limiting establishment of natural RSV infections and in the therapeutic treatment of severe RSV disease.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , beta-Defensins , Antibodies, Viral , Antimicrobial Peptides , Child , Glycoproteins , Humans , Viral Fusion Proteins/chemistry , beta-Defensins/pharmacology
19.
Proc Natl Acad Sci U S A ; 119(25): e2203326119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696580

ABSTRACT

Human metapneumovirus (hMPV) is a leading cause of morbidity and hospitalization among children worldwide, however, no vaccines or therapeutics are currently available for hMPV disease prevention and treatment. The hMPV fusion (F) protein is the sole target of neutralizing antibodies. To map the immunodominant epitopes on the hMPV F protein, we isolated a panel of human monoclonal antibodies (mAbs), and the mAbs were assessed for binding avidity, neutralization potency, and epitope specificity. We found the majority of the mAbs target diverse epitopes on the hMPV F protein, and we discovered multiple mAb binding approaches for antigenic site III. The most potent mAb, MPV467, which had picomolar potency, was examined in prophylactic and therapeutic mouse challenge studies, and MPV467 limited virus replication in mouse lungs when administered 24 h before or 72 h after viral infection. We determined the structure of MPV467 in complex with the hMPV F protein using cryo-electron microscopy to a resolution of 3.3 Å, which revealed a complex novel prefusion-specific epitope overlapping antigenic sites II and V on a single protomer. Overall, our data reveal insights into the immunodominant antigenic epitopes on the hMPV F protein, identify a mAb therapy for hMPV F disease prevention and treatment, and provide the discovery of a prefusion-specific epitope on the hMPV F protein.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Antigens, Viral , Metapneumovirus , Paramyxoviridae Infections , Viral Fusion Proteins , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/chemistry , Antibodies, Viral/isolation & purification , Antibodies, Viral/therapeutic use , Antigens, Viral/chemistry , Antigens, Viral/immunology , Cryoelectron Microscopy , Epitopes/immunology , Humans , Metapneumovirus/immunology , Mice , Paramyxoviridae Infections/prevention & control , Primary Prevention , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology
20.
Sci Adv ; 8(19): eabj6894, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35544562

ABSTRACT

Endogenous viral elements (EVEs), accounting for 15% of our genome, serve as a genetic reservoir from which new genes can emerge. Nematode EVEs are particularly diverse and informative of virus evolution. We identify Atlas virus-an intact retrovirus-like EVE in the human hookworm Ancylostoma ceylanicum, with an envelope protein genetically related to GN-GC glycoproteins from the family Phenuiviridae. A cryo-EM structure of Atlas GC reveals a class II viral membrane fusion protein fold not previously seen in retroviruses. Atlas GC has the structural hallmarks of an active fusogen. Atlas GC trimers insert into membranes with endosomal lipid compositions and low pH. When expressed on the plasma membrane, Atlas GC has cell-cell fusion activity. With its preserved biological activities, Atlas GC has the potential to acquire a cellular function. Our work reveals structural plasticity in reverse-transcribing RNA viruses.


Subject(s)
Phlebovirus , RNA Viruses , Ancylostomatoidea/metabolism , Animals , Humans , Phlebovirus/chemistry , Phlebovirus/genetics , Phlebovirus/metabolism , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL