Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Nat Commun ; 15(1): 6892, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134522

ABSTRACT

Nipah virus infection, one of the top priority diseases recognized by the World Health Organization, underscores the urgent need to develop effective countermeasures against potential epidemics and pandemics. Here, we identify a fully human single-domain antibody that targets a highly conserved cryptic epitope situated at the dimeric interface of the Nipah virus G protein (receptor binding protein, RBP), as elucidated through structures by high-resolution cryo-electron microscopy (cryo-EM). This unique binding mode disrupts the tetramerization of the G protein, consequently obstructing the activation of the F protein and inhibiting viral membrane fusion. Furthermore, our investigations reveal that this compact antibody displays enhanced permeability across the blood-brain barrier (BBB) and demonstrates superior efficacy in eliminating pseudovirus within the brain in a murine model of Nipah virus infection, particularly compared to the well-characterized antibody m102.4 in an IgG1 format. Consequently, this single-domain antibody holds promise as a therapeutic candidate to prevent Nipah virus infections and has potential implications for vaccine development.


Subject(s)
Antibodies, Viral , Cryoelectron Microscopy , Epitopes , Henipavirus Infections , Nipah Virus , Single-Domain Antibodies , Nipah Virus/immunology , Humans , Animals , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Henipavirus Infections/virology , Epitopes/immunology , Mice , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Antibodies, Viral/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Female , HEK293 Cells
2.
Biomed Res Int ; 2024: 4066641, 2024.
Article in English | MEDLINE | ID: mdl-38962403

ABSTRACT

The zoonotic viruses pose significant threats to public health. Nipah virus (NiV) is an emerging virus transmitted from bats to humans. The NiV causes severe encephalitis and acute respiratory distress syndrome, leading to high mortality rates, with fatality rates ranging from 40% to 75%. The first emergence of the disease was found in Malaysia in 1998-1999 and later in Bangladesh, Cambodia, Timor-Leste, Indonesia, Singapore, Papua New Guinea, Vietnam, Thailand, India, and other South and Southeast Asian nations. Currently, no specific vaccines or antiviral drugs are available. The potential advantages of epitope-based vaccines include their ability to elicit specific immune responses while minimizing potential side effects. The epitopes have been identified from the conserved region of viral proteins obtained from the UniProt database. The selection of conserved epitopes involves analyzing the genetic sequences of various viral strains. The present study identified two B cell epitopes, seven cytotoxic T lymphocyte (CTL) epitopes, and seven helper T lymphocyte (HTL) epitope interactions from the NiV proteomic inventory. The antigenic and physiological properties of retrieved protein were analyzed using online servers ToxinPred, VaxiJen v2.0, and AllerTOP. The final vaccine candidate has a total combined coverage range of 80.53%. The tertiary structure of the constructed vaccine was optimized, and its stability was confirmed with the help of molecular simulation. Molecular docking was performed to check the binding affinity and binding energy of the constructed vaccine with TLR-3 and TLR-5. Codon optimization was performed in the constructed vaccine within the Escherichia coli K12 strain, to eliminate the danger of codon bias. However, these findings must require further validation to assess their effectiveness and safety. The development of vaccines and therapeutic approaches for virus infection is an ongoing area of research, and it may take time before effective interventions are available for clinical use.


Subject(s)
Computer Simulation , Henipavirus Infections , Nipah Virus , Nipah Virus/immunology , Humans , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Viral Vaccines/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Computational Biology/methods , Epitopes, T-Lymphocyte/immunology , Vaccination , Molecular Docking Simulation , Viral Proteins/immunology , Viral Proteins/chemistry , Viral Proteins/genetics , Animals
3.
Vet Microbiol ; 295: 110167, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38954881

ABSTRACT

Hendra virus (HeV) is lethal to horses and a zoonotic threat to humans in Australia, causing severe neurological and/or respiratory disease with high mortality. An equine vaccine has been available since 2012. Foals acquire antibodies from their dams by ingesting colostrum after parturition, therefore it is assumed that foals of mares vaccinated against HeV will have passive HeV antibodies circulating during the first several months of life until they are actively vaccinated. However, no studies have yet examined passive or active immunity against HeV in foals. Here, we investigated anti-HeV antibody levels in vaccinated mares and their foals. Testing for HeV neutralising antibodies is cumbersome due to the requirement for Biosafety level 4 (BSL-4) containment to conduct virus neutralisation tests (VNT). For this study, a subset of samples was tested for HeV G-specific antibodies by both an authentic VNT with infectious HeV and a microsphere-based immunoassay (MIA), revealing a strong correlation. An indicative neutralising level was then applied to the results of a larger sample set tested using the MIA. Mares had high levels of HeV-specific neutralising antibodies at the time of parturition. Foals acquired high levels of maternal antibodies which then waned to below predictive protective levels in most foals by 6 months old when vaccination commenced. Foals showed a suboptimal response to vaccination, suggesting maternal antibodies may interfere with active vaccination. The correlation analysis between the authentic HeV VNT and HeV MIA will enable further high throughput serological studies to inform optimal vaccination protocols for both broodmares and foals.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Hendra Virus , Henipavirus Infections , Horse Diseases , Vaccination , Viral Vaccines , Animals , Horses , Hendra Virus/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Horse Diseases/immunology , Antibodies, Viral/blood , Henipavirus Infections/prevention & control , Henipavirus Infections/veterinary , Henipavirus Infections/immunology , Henipavirus Infections/virology , Female , Vaccination/veterinary , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Neutralizing/blood , Immunity, Maternally-Acquired , Animals, Newborn/immunology , Pregnancy , Neutralization Tests/veterinary , Australia , Colostrum/immunology
4.
Front Immunol ; 15: 1387811, 2024.
Article in English | MEDLINE | ID: mdl-38911870

ABSTRACT

The Nipah virus (NiV), a highly deadly bat-borne paramyxovirus, poses a substantial threat due to recurrent outbreaks in specific regions, causing severe respiratory and neurological diseases with high morbidity. Two distinct strains, NiV-Malaysia (NiV-M) and NiV-Bangladesh (NiV-B), contribute to outbreaks in different geographical areas. Currently, there are no commercially licensed vaccines or drugs available for prevention or treatment. In response to this urgent need for protection against NiV and related henipaviruses infections, we developed a novel homotypic virus-like nanoparticle (VLP) vaccine co-displaying NiV attachment glycoproteins (G) from both strains, utilizing the self-assembling properties of ferritin protein. In comparison to the NiV G subunit vaccine, our nanoparticle vaccine elicited significantly higher levels of neutralizing antibodies and provided complete protection against a lethal challenge with NiV infection in Syrian hamsters. Remarkably, the nanoparticle vaccine stimulated the production of antibodies that exhibited superior cross-reactivity to homologous or heterologous henipavirus. These findings underscore the potential utility of ferritin-based nanoparticle vaccines in providing both broad-spectrum and long-term protection against NiV and emerging zoonotic henipaviruses challenges.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Ferritins , Henipavirus Infections , Mesocricetus , Nanoparticles , Nipah Virus , Viral Vaccines , Animals , Nipah Virus/immunology , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Ferritins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Cricetinae , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Female , Humans , Nanovaccines
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.
Front Immunol ; 15: 1384417, 2024.
Article in English | MEDLINE | ID: mdl-38726013

ABSTRACT

Nipah virus (NiV) poses a significant threat to human and livestock populations across South and Southeast Asia. Vaccines are required to reduce the risk and impact of spillover infection events. Pigs can act as an intermediate amplifying host for NiV and, separately, provide a preclinical model for evaluating human vaccine candidate immunogenicity. The aim of this study was therefore to evaluate the immunogenicity of an mRNA vectored NiV vaccine candidate in pigs. Pigs were immunized twice with 100 µg nucleoside-modified mRNA vaccine encoding soluble G glycoprotein from the Malaysia strain of NiV, formulated in lipid nanoparticles. Potent antigen-binding and virus neutralizing antibodies were detected in serum following the booster immunization. Antibody responses effectively neutralized both the Malaysia and Bangladesh strains of NiV but showed limited neutralization of the related (about 80% amino acid sequence identity for G) Hendra virus. Antibodies were also capable of neutralizing NiV glycoprotein mediated cell-cell fusion. NiV G-specific T cell cytokine responses were also measurable following the booster immunization with evidence for induction of both CD4 and CD8 T cell responses. These data support the further evaluation of mRNA vectored NiV G as a vaccine for both pigs and humans.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Henipavirus Infections , Nipah Virus , Viral Vaccines , Animals , Nipah Virus/immunology , Nipah Virus/genetics , Swine , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Viral/blood , Antibodies, Viral/immunology , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , RNA, Messenger/genetics , RNA, Messenger/immunology , Immunogenicity, Vaccine , Immunization, Secondary , Cytokines/immunology , Vaccines, Synthetic/immunology , Liposomes , Nanoparticles
7.
PLoS One ; 19(5): e0300507, 2024.
Article in English | MEDLINE | ID: mdl-38728300

ABSTRACT

According to the 2018 WHO R&D Blueprint, Nipah virus (NiV) is a priority disease, and the development of a vaccine against NiV is strongly encouraged. According to criteria used to categorize zoonotic diseases, NiV is a stage III disease that can spread to people and cause unpredictable outbreaks. Since 2001, the NiV virus has caused annual outbreaks in Bangladesh, while in India it has caused occasional outbreaks. According to estimates, the mortality rate for infected individuals ranges from 70 to 91%. Using immunoinformatic approaches to anticipate the epitopes of the MHC-I, MHC-II, and B-cells, they were predicted using the NiV glycoprotein and nucleocapsid protein. The selected epitopes were used to develop a multi-epitope vaccine construct connected with linkers and adjuvants in order to improve immune responses to the vaccine construct. The 3D structure of the engineered vaccine was anticipated, optimized, and confirmed using a variety of computer simulation techniques so that its stability could be assessed. According to the immunological simulation tests, it was found that the vaccination elicits a targeted immune response against the NiV. Docking with TLR-3, 7, and 8 revealed that vaccine candidates had high binding affinities and low binding energies. Finally, molecular dynamic analysis confirms the stability of the new vaccine. Codon optimization and in silico cloning showed that the proposed vaccine was expressed to a high degree in Escherichia coli. The study will help in identifying a potential epitope for a vaccine candidate against NiV. The developed multi-epitope vaccine construct has a lot of potential, but they still need to be verified by in vitro & in vivo studies.


Subject(s)
Glycoproteins , Nipah Virus , Viral Vaccines , Nipah Virus/immunology , Viral Vaccines/immunology , Glycoproteins/immunology , Glycoproteins/chemistry , Humans , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Computer Simulation , Epitopes/immunology , Epitopes/chemistry , Molecular Dynamics Simulation , Nucleocapsid/immunology , Molecular Docking Simulation
8.
Viruses ; 16(5)2024 05 16.
Article in English | MEDLINE | ID: mdl-38793674

ABSTRACT

The Nipah virus (NiV) and the Hendra virus (HeV) are highly pathogenic zoonotic diseases that can cause fatal infections in humans and animals. Early detection is critical for the control of NiV and HeV infections. We present the development of two antigen-detection ELISAs (AgELISAs) using the henipavirus-receptor EphrinB2 and monoclonal antibodies (mAbs) to detect NiV and HeV. The NiV AgELISA detected only NiV, whereas the NiV/HeV AgELISA detected both NiV and HeV. The diagnostic specificities of the NiV AgELISA and the NiV/HeV AgELISA were 100% and 97.8%, respectively. Both assays were specific for henipaviruses and showed no cross-reactivity with other viruses. The AgELISAs detected NiV antigen in experimental pig nasal wash samples taken at 4 days post-infection. With the combination of both AgELISAs, NiV can be differentiated from HeV. Complementing other henipavirus detection methods, these two newly developed AgELISAs can rapidly detect NiV and HeV in a large number of samples and are suitable for use in remote areas where other tests are not available.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Enzyme-Linked Immunosorbent Assay , Ephrin-B2 , Hendra Virus , Henipavirus Infections , Nipah Virus , Hendra Virus/immunology , Animals , Nipah Virus/immunology , Antibodies, Monoclonal/immunology , Enzyme-Linked Immunosorbent Assay/methods , Ephrin-B2/metabolism , Ephrin-B2/immunology , Henipavirus Infections/diagnosis , Henipavirus Infections/virology , Henipavirus Infections/immunology , Antibodies, Viral/immunology , Swine , Humans , Sensitivity and Specificity , Receptors, Virus/metabolism , Antigens, Viral/immunology
9.
Trop Med Int Health ; 29(5): 354-364, 2024 May.
Article in English | MEDLINE | ID: mdl-38415314

ABSTRACT

OBJECTIVES: Nipah and Hendra are deadly zoonotic diseases with pandemic potential. To date, no human vaccine or monoclonal antibody (mAb) has been licensed to prevent disease caused by these pathogens. The aim of this scoping review was to identify and describe all Phase I, II, and III clinical trials of vaccine candidates or mAbs candidates designed to prevent Nipah and Hendra in humans and to compare the characteristics of the vaccine candidates to characteristics outlined in the Target Product Profile drafted by the World Health Organisation as part of the WHO Research & Development Blueprint for Action to Prevent Epidemics. METHODS: We searched 23 clinical trial registries, the Cochrane Central Register of Clinical Trials, and grey literature up to June 2023 to identify vaccine and mAb candidates being evaluated in registered clinical trials. Vaccine candidate and trial characteristics were double-extracted for evaluation and the vaccine candidate characteristics were compared with the preferred and critical criteria of the World Health Organisation's Target Product Profile for Nipah virus vaccine. RESULTS: Three vaccine candidates (Hendra Virus Soluble Glycoprotein Vaccine [HeV-sG-V], PHV02, and mRNA-1215) and one mAb (m102.4) had a registered human clinical trial by June 2023. All trials were phase 1, dose-ranging trials taking place in the United States of America or Australia and enrolling healthy adults. Although all vaccine candidates meet the dose regimen and route of administration criteria of the Target Product Profile, other criteria such as measures of efficacy and reactogenicity will need to be evaluated in the future as evidence becomes available. CONCLUSION: Multiple vaccine candidates and one mAb candidate have reached the stage of human clinical trials and are reviewed here. Monitoring progress during evaluation of these candidates and candidates entering clinical trials in the future can help highlight many of the challenges that remain.


Subject(s)
Antibodies, Monoclonal , Hendra Virus , Henipavirus Infections , Nipah Virus , Viral Vaccines , Humans , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Antibodies, Monoclonal/therapeutic use , Hendra Virus/immunology , Nipah Virus/immunology , Viral Vaccines/immunology , Viral Vaccines/therapeutic use , Clinical Trials as Topic , Animals
10.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502239

ABSTRACT

Nipah virus is one of the most harmful emerging viruses with deadly effects on both humans and animals. Because of the severe outbreaks, in 2018, the World Health Organization focused on the urgent need for the development of effective solutions against the virus. However, up to date, there is no effective vaccine against the Nipah virus in the market. In the current study, the complete proteome of the Nipah virus (nine proteins) was analyzed for the antigenicity score and the virulence role of each protein, where we came up with fusion glycoprotein (F), glycoprotein (G), protein (V), and protein (W) as the candidates for epitope prediction. Following that, the multitope vaccine was designed based on top-ranking CTL, HTL, and BCL epitopes from the selected proteins. We used suitable linkers, adjuvant, and PADRE peptides to finalize the constructed vaccine, which was analyzed for its physicochemical features, antigenicity, toxicity, allergenicity, and solubility. The designed vaccine passed these assessments through computational analysis and, as a final step, we ran a docking analysis between the designed vaccine and TLR-3 and validated the docked complex through molecular dynamics simulation, which estimated a strong binding and supported the nomination of the designed vaccine as a putative solution for Nipah virus. Here, we describe the computational approach for design and analysis of this vaccine.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Henipavirus Infections/prevention & control , Nipah Virus/immunology , Proteome/immunology , Vaccines, Subunit/administration & dosage , Computational Biology , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Molecular Docking Simulation , Protein Conformation , Proteome/analysis , Proteome/metabolism , Vaccines, Subunit/immunology
11.
Cell Rep ; 36(9): 109628, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469726

ABSTRACT

Hendra virus and Nipah virus (NiV), members of the Henipavirus (HNV) genus, are zoonotic paramyxoviruses known to cause severe disease across six mammalian orders, including humans. We isolated a panel of human monoclonal antibodies (mAbs) from the B cells of an individual with prior exposure to equine Hendra virus (HeV) vaccine, targeting distinct antigenic sites. The most potent class of cross-reactive antibodies achieves neutralization by blocking viral attachment to the host cell receptors ephrin-B2 and ephrin-B3, with a second class being enhanced by receptor binding. mAbs from both classes display synergistic activity in vitro. In a stringent hamster model of NiV Bangladesh (NiVB) infection, antibodies from both classes reduce morbidity and mortality and achieve synergistic protection in combination. These candidate mAbs might be suitable for use in a cocktail therapeutic approach to achieve synergistic potency and reduce the risk of virus escape.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antiviral Agents/pharmacology , Ephrin-B2/antagonists & inhibitors , Ephrin-B3/antagonists & inhibitors , Henipavirus Infections/prevention & control , Henipavirus/pathogenicity , Receptors, Virus/antagonists & inhibitors , Animals , Antibody Specificity , Chlorocebus aethiops , Cross Reactions , Disease Models, Animal , Drug Therapy, Combination , Ephrin-B2/immunology , Ephrin-B2/metabolism , Ephrin-B3/immunology , Ephrin-B3/metabolism , Female , Henipavirus Infections/immunology , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Host-Pathogen Interactions , Humans , Mesocricetus , Receptors, Virus/immunology , Receptors, Virus/metabolism , Vero Cells
12.
Viruses ; 13(7)2021 07 02.
Article in English | MEDLINE | ID: mdl-34372504

ABSTRACT

Pandemics are a consequence of a series of processes that span scales from viral biology at 10-9 m to global transmission at 106 m. The pathogen passes from one host species to another through a sequence of events that starts with an infected reservoir host and entails interspecific contact, innate immune responses, receptor protein structure within the potential host, and the global spread of the novel pathogen through the naive host population. Each event presents a potential barrier to the onward passage of the virus and should be characterized with an integrated transdisciplinary approach. Epidemic control is based on the prevention of exposure, infection, and disease. However, the ultimate pandemic prevention is prevention of the spillover event itself. Here, we focus on the potential for preventing the spillover of henipaviruses, a group of viruses derived from bats that frequently cross species barriers, incur high human mortality, and are transmitted among humans via stuttering chains. We outline the transdisciplinary approach needed to prevent the spillover process and, therefore, future pandemics.


Subject(s)
Chiroptera/virology , Global Health , Henipavirus Infections/prevention & control , Henipavirus/pathogenicity , Pandemics/prevention & control , Virus Diseases/prevention & control , Zoonoses/virology , Animals , Henipavirus Infections/epidemiology , Henipavirus Infections/immunology , Henipavirus Infections/transmission , Host Specificity , Humans , Immunity, Innate , Nipah Virus/pathogenicity , Virus Diseases/immunology , Virus Diseases/transmission , Zoonoses/prevention & control , Zoonoses/transmission
13.
Immunol Res ; 69(5): 457-460, 2021 10.
Article in English | MEDLINE | ID: mdl-34357535

ABSTRACT

In this manuscript, COVID-19, Ebola virus disease, Nipah virus infection, SARS, and MERS are suggested to be considered for a novel immunological reclassification as acute onset immune dysrhythmia syndrome (n-AIDS) due to altered monocytic, Th1/Th2, as well as cytokines and chemokines balances. n-AIDs is postulated to be the cause of the acute respiratory distress and multi-inflammatory syndromes which are described with fatal COVID-19, and immunomodulators are suggested to effectively manage the mentioned diseases as well as for other disorders caused by Th1/Th2 imbalance. Meanwhile, para COVID syndrome is suggested to describe various immune-related complications, whether before or after recovery, and to embrace a potential of a latent infection, that might be discovered later, as occurred with Ebola virus disease. Finally, our hypothesis has evolved out of our real-life practice that uses immunomodulatory drugs to manage COVID-19 safely and effectively.


Subject(s)
COVID-19/immunology , Cytokines/immunology , Hemorrhagic Fever, Ebola/immunology , Henipavirus Infections/immunology , Acquired Immunodeficiency Syndrome/immunology , Chemokines/immunology , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Hemorrhagic Fever, Ebola/drug therapy , Henipavirus Infections/drug therapy , Humans , Immunologic Factors/therapeutic use , Lymphocytes/immunology , SARS-CoV-2/physiology , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/immunology , COVID-19 Drug Treatment
14.
Cells ; 10(6)2021 05 25.
Article in English | MEDLINE | ID: mdl-34070626

ABSTRACT

Nipah virus (NiV) is a highly pathogenic zoonotic virus with a broad species tropism, originating in pteropid bats. Human outbreaks of NiV disease occur almost annually, often with high case-fatality rates. The specific events that lead to pathogenesis are not well defined, but the disease has both respiratory and encephalitic components, with relapsing encephalitis occurring in some cases more than a year after initial infection. Several cell types are targets of NiV, dictated by the expression of the ephrin-B2/3 ligand on the cell's outer membrane, which interact with the NiV surface proteins. Vascular endothelial cells (ECs) are major targets of infection. Cytopathic effects (CPE), characterized by syncytia formation and cell death, and an ensuing vasculitis, are a major feature of the disease. Smooth muscle cells (SMCs) of the tunica media that line small blood vessels are infected in humans and animal models of NiV disease, although pathology or histologic changes associated with antigen-positive SMCs have not been reported. To gain an understanding of the possible contributions that SMCs might have in the development of NiV disease, we investigated the susceptibility and potential cytopathogenic changes of human SMCs to NiV infection in vitro. SMCs were permissive for NiV infection and resulted in high titers and prolonged NiV production, despite a lack of cytopathogenicity, and in the absence of detectable ephrin-B2/3. These results indicate that SMC might be important contributors to disease by producing progeny NiV during an infection, without suffering cytopathogenic consequences.


Subject(s)
Endothelial Cells , Henipavirus Infections , Myocytes, Smooth Muscle , Animals , Chlorocebus aethiops , Disease Susceptibility , Endothelial Cells/immunology , Endothelial Cells/virology , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/virology , Nipah Virus , Vero Cells , Virus Replication
15.
Nat Struct Mol Biol ; 28(5): 426-434, 2021 05.
Article in English | MEDLINE | ID: mdl-33927387

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are henipaviruses (HNVs) causing respiratory illness and severe encephalitis in humans, with fatality rates of 50-100%. There are no licensed therapeutics or vaccines to protect humans. HeV and NiV use a receptor-binding glycoprotein (G) and a fusion glycoprotein (F) to enter host cells. HNV F and G are the main targets of the humoral immune response, and the presence of neutralizing antibodies is a correlate of protection against NiV and HeV in experimentally infected animals. We describe here two cross-reactive F-specific antibodies, 1F5 and 12B2, that neutralize NiV and HeV through inhibition of membrane fusion. Cryo-electron microscopy structures reveal that 1F5 and 12B2 recognize distinct prefusion-specific, conserved quaternary epitopes and lock F in its prefusion conformation. We provide proof-of-concept for using antibody cocktails for neutralizing NiV and HeV and define a roadmap for developing effective countermeasures against these highly pathogenic viruses.


Subject(s)
Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , Hendra Virus/immunology , Nipah Virus/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , CHO Cells , Cricetulus , Cross Reactions , HEK293 Cells , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Humans , Mice , Virus Internalization
16.
J Gen Virol ; 102(1)2021 01.
Article in English | MEDLINE | ID: mdl-33054904

ABSTRACT

Although enveloped viruses canonically mediate particle entry through virus-cell fusion, certain viruses can spread by cell-cell fusion, brought about by receptor engagement and triggering of membrane-bound, viral-encoded fusion proteins on the surface of cells. The formation of pathogenic syncytia or multinucleated cells is seen in vivo, but their contribution to viral pathogenesis is poorly understood. For the negative-strand paramyxoviruses respiratory syncytial virus (RSV) and Nipah virus (NiV), cell-cell spread is highly efficient because their oligomeric fusion protein complexes are active at neutral pH. The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has also been reported to induce syncytia formation in infected cells, with the spike protein initiating cell-cell fusion. Whilst it is well established that fusion protein-specific antibodies can block particle attachment and/or entry into the cell (canonical virus neutralization), their capacity to inhibit cell-cell fusion and the consequences of this neutralization for the control of infection are not well characterized, in part because of the lack of specific tools to assay and quantify this activity. Using an adapted bimolecular fluorescence complementation assay, based on a split GFP-Renilla luciferase reporter, we have established a micro-fusion inhibition test (mFIT) that allows the identification and quantification of these neutralizing antibodies. This assay has been optimized for high-throughput use and its applicability has been demonstrated by screening monoclonal antibody (mAb)-mediated inhibition of RSV and NiV fusion and, separately, the development of fusion-inhibitory antibodies following NiV vaccine immunization in pigs. In light of the recent emergence of coronavirus disease 2019 (COVID-19), a similar assay was developed for SARS-CoV-2 and used to screen mAbs and convalescent patient plasma for fusion-inhibitory antibodies. Using mFITs to assess antibody responses following natural infection or vaccination is favourable, as this assay can be performed entirely at low biocontainment, without the need for live virus. In addition, the repertoire of antibodies that inhibit cell-cell fusion may be different to those that inhibit particle entry, shedding light on the mechanisms underpinning antibody-mediated neutralization of viral spread.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , COVID-19/diagnosis , Henipavirus Infections/diagnosis , High-Throughput Screening Assays , Respiratory Syncytial Virus Infections/diagnosis , Viral Fusion Proteins/antagonists & inhibitors , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/isolation & purification , Antibodies, Viral/metabolism , COVID-19/immunology , COVID-19/virology , Cell Fusion , Convalescence , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Immune Sera/chemistry , Luciferases/genetics , Luciferases/metabolism , Models, Molecular , Nipah Virus/immunology , Nipah Virus/pathogenicity , Protein Conformation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Swine , Viral Fusion Protein Inhibitors/chemistry , Viral Fusion Protein Inhibitors/metabolism , Viral Fusion Protein Inhibitors/pharmacology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
17.
mSphere ; 5(6)2020 12 16.
Article in English | MEDLINE | ID: mdl-33328346

ABSTRACT

The Nipah virus (NiV) phosphoprotein (P) gene encodes four proteins. Three of these-P, V, and W-possess a common N-terminal domain but distinct C termini. These proteins interact with immune modulators. Previous studies demonstrated that P, V, and W bind STAT1 and STAT4 and that V also interacts with STAT2 but not with STAT3. The STAT1 and STAT2 interactions block interferon (IFN)-induced STAT tyrosine phosphorylation. To more fully characterize the interactions of P, V, and W with the STATs, we screened for interaction of each viral protein with STATs 1 to 6 by coimmunoprecipitation. We demonstrate that NiV P, V, and W interact with STAT4 through their common N-terminal domain and block STAT4 activity, based on a STAT4 response element reporter assay. Although none of the NiV proteins interact with STAT3 or STAT6, NiV V, but not P or W, interacts with STAT5 through its unique C terminus. Furthermore, the interaction of NiV V with STAT5 was not disrupted by overexpression of the N-terminal binding STAT1 or the C-terminal binding MDA5. NiV V also inhibits a STAT5 response element reporter assay. Residues 114 to 140 of the common N-terminal domain of the NiV P gene products were found to be sufficient to bind STAT1 and STAT4. Analysis of STAT1-STAT3 chimeras suggests that the P gene products target the STAT1 SH2 domain. When fused to GST, the 114-140 peptide is sufficient to decrease STAT1 phosphorylation in IFN-ß-stimulated cells, suggesting that this peptide could potentially be fused to heterologous proteins to confer inhibition of STAT1- and STAT4-dependent responses.IMPORTANCE How Nipah virus (NiV) antagonizes innate immune responses is incompletely understood. The P gene of NiV encodes the P, V, and W proteins. These proteins have a common N-terminal sequence that is sufficient to bind to STAT1 and STAT2 and block IFN-induced signal transduction. This study sought to more fully understand how P, V, and W engage with the STAT family of transcription factors to influence their functions. The results identify a novel interaction of V with STAT5 and demonstrate V inhibition of STAT5 function. We also demonstrate that the common N-terminal residues 114 to 140 of P, V, and W are critical for inhibition of STAT1 and STAT4 function, map the interaction to the SH2 region of STAT1, and show that a fusion construct with this peptide significantly inhibits cytokine-induced STAT1 phosphorylation. These data clarify how these important virulence factors modulate innate antiviral defenses.


Subject(s)
Cell Nucleus/chemistry , Henipavirus Infections/metabolism , Nipah Virus/physiology , STAT Transcription Factors/metabolism , Viral Proteins/metabolism , HEK293 Cells , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Immunity, Innate/immunology , Phosphorylation , STAT Transcription Factors/genetics , Signal Transduction , Trans-Activators/metabolism , Viral Proteins/genetics
18.
Sci Rep ; 10(1): 18256, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33106487

ABSTRACT

Nipah Virus (NiV) has been designated as a priority disease with an urgent need for therapeutic development by World Health Organization. The monoclonal antibody m102.4 binds to the immunodominant NiV receptor-binding glycoprotein (GP), and potently neutralizes NiV, indicating its potential as a therapeutic agent. Although the co-crystal structure of m102.3, an m102.4 derivative, in complex with the GP of the related Hendra Virus (HeV) has been solved, the structural interaction between m102.4 and NiV is uncharacterized. Herein, we used structure-guided alanine-scanning mutagenesis to map the functional epitope and paratope residues that govern the antigen-antibody interaction. Our results revealed that the binding of m102.4 is mediated predominantly by two residues in the HCDR3 region, which is unusually small for an antibody-antigen interaction. We performed computational docking to generate a structural model of m102.4-NiV interaction. Our model indicates that m102.4 targets the common hydrophobic central cavity and a hydrophilic rim on the GP, as observed for the m102.3-HeV co-crystal, albeit with Fv orientation differences. In summary, our study provides insight into the m102.4-NiV interaction, demonstrating that structure-guided alanine-scanning and computational modeling can serve as the starting point for additional antibody reengineering (e.g. affinity maturation) to generate potential therapeutic candidates.


Subject(s)
Alanine/genetics , Antibodies, Monoclonal/metabolism , Computer Simulation , Glycoproteins/metabolism , Henipavirus Infections/virology , Nipah Virus/metabolism , Viral Envelope Proteins/metabolism , Alanine/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Epitopes/immunology , Glycoproteins/chemistry , Glycoproteins/genetics , Henipavirus Infections/immunology , Henipavirus Infections/metabolism , Humans , Mutagenesis, Site-Directed , Nipah Virus/immunology , Nipah Virus/isolation & purification , Protein Structural Elements/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
19.
Annu Rev Virol ; 7(1): 447-473, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32991264

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are bat-borne zoonotic para-myxoviruses identified in the mid- to late 1990s in outbreaks of severe disease in livestock and people in Australia and Malaysia, respectively. HeV repeatedly re-emerges in Australia while NiV continues to cause outbreaks in South Asia (Bangladesh and India), and these viruses have remained transboundary threats. In people and several mammalian species, HeV and NiV infections present as a severe systemic and often fatal neurologic and/or respiratory disease. NiV stands out as a potential pandemic threat because of its associated high case-fatality rates and capacity for human-to-human transmission. The development of effective vaccines, suitable for people and livestock, against HeV and NiV has been a research focus. Here, we review the progress made in NiV and HeV vaccine development, with an emphasis on those approaches that have been tested in established animal challenge models of NiV and HeV infection and disease.


Subject(s)
Communicable Diseases, Emerging/prevention & control , Hendra Virus/immunology , Henipavirus Infections/prevention & control , Nipah Virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Chiroptera/virology , Communicable Diseases, Emerging/immunology , Communicable Diseases, Emerging/virology , Disease Models, Animal , Henipavirus Infections/immunology , Humans , Mice , Viral Zoonoses/prevention & control , Viral Zoonoses/transmission
20.
Cells ; 9(8)2020 08 18.
Article in English | MEDLINE | ID: mdl-32824665

ABSTRACT

Nipah and Hendra viruses are highly pathogenic, zoonotic henipaviruses that encode proteins that inhibit the host's innate immune response. The W protein is one of four products encoded from the P gene and binds a number of host proteins to regulate signalling pathways. The W protein is intrinsically disordered, a structural attribute that contributes to its diverse host protein interactions. Here, we review the role of W in innate immune suppression through inhibition of both pattern recognition receptor (PRR) pathways and interferon (IFN)-responsive signalling. PRR stimulation leading to activation of IRF-3 and IFN release is blocked by henipavirus W, and unphosphorylated STAT proteins are sequestered within the nucleus of host cells by W, thereby inhibiting the induction of IFN stimulated genes. We examine the critical role of nuclear transport in multiple functions of W and how specific binding of importin-alpha (Impα) isoforms, and the 14-3-3 group of regulatory proteins suggests further modulation of these processes. Overall, the disordered nature and multiple functions of W warrant further investigation to understand henipavirus pathogenesis and may reveal insights aiding the development of novel therapeutics.


Subject(s)
Active Transport, Cell Nucleus/immunology , Hendra Virus/metabolism , Henipavirus Infections/immunology , Intrinsically Disordered Proteins/metabolism , Nipah Virus/metabolism , Nuclear Envelope/metabolism , Signal Transduction/immunology , Viral Proteins/metabolism , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Host Microbial Interactions/immunology , Humans , Immunity, Innate , Interferons/metabolism , Intrinsically Disordered Proteins/chemistry , Receptors, Pattern Recognition/metabolism , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL