Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Front Immunol ; 15: 1384417, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38726013

RESUMO

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.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Infecções por Henipavirus , Vírus Nipah , Vacinas Virais , Animais , Vírus Nipah/imunologia , Vírus Nipah/genética , Suínos , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Doenças dos Suínos/imunologia , Doenças dos Suínos/prevenção & controle , Doenças dos Suínos/virologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Imunogenicidade da Vacina , Imunização Secundária , Citocinas/imunologia , Vacinas Sintéticas/imunologia , Lipossomos , Nanopartículas
2.
PLoS One ; 19(5): e0300507, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38728300

RESUMO

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.


Assuntos
Glicoproteínas , Vírus Nipah , Vacinas Virais , Vírus Nipah/imunologia , Vacinas Virais/imunologia , Glicoproteínas/imunologia , Glicoproteínas/química , Humanos , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/imunologia , Simulação por Computador , Epitopos/imunologia , Epitopos/química , Simulação de Dinâmica Molecular , Nucleocapsídeo/imunologia , Simulação de Acoplamento Molecular
3.
Trop Med Int Health ; 29(5): 354-364, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38415314

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Vírus Hendra , Infecções por Henipavirus , Vírus Nipah , Vacinas Virais , Humanos , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/imunologia , Anticorpos Monoclonais/uso terapêutico , Vírus Hendra/imunologia , Vírus Nipah/imunologia , Vacinas Virais/imunologia , Vacinas Virais/uso terapêutico , Ensaios Clínicos como Assunto , Animais
4.
Int J Mol Sci ; 22(17)2021 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-34502239

RESUMO

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.


Assuntos
Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Infecções por Henipavirus/prevenção & controle , Vírus Nipah/imunologia , Proteoma/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Biologia Computacional , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Humanos , Simulação de Acoplamento Molecular , Conformação Proteica , Proteoma/análise , Proteoma/metabolismo , Vacinas de Subunidades Antigênicas/imunologia
5.
Cell Rep ; 36(9): 109628, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34469726

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Antivirais/farmacologia , Efrina-B2/antagonistas & inibidores , Efrina-B3/antagonistas & inibidores , Infecções por Henipavirus/prevenção & controle , Henipavirus/patogenicidade , Receptores Virais/antagonistas & inibidores , Animais , Especificidade de Anticorpos , Chlorocebus aethiops , Reações Cruzadas , Modelos Animais de Doenças , Quimioterapia Combinada , Efrina-B2/imunologia , Efrina-B2/metabolismo , Efrina-B3/imunologia , Efrina-B3/metabolismo , Feminino , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/metabolismo , Infecções por Henipavirus/virologia , Interações Hospedeiro-Patógeno , Humanos , Mesocricetus , Receptores Virais/imunologia , Receptores Virais/metabolismo , Células Vero
6.
Immunol Res ; 69(5): 457-460, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34357535

RESUMO

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.


Assuntos
COVID-19/imunologia , Citocinas/imunologia , Doença pelo Vírus Ebola/imunologia , Infecções por Henipavirus/imunologia , Síndrome da Imunodeficiência Adquirida/imunologia , Quimiocinas/imunologia , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/imunologia , Doença pelo Vírus Ebola/tratamento farmacológico , Infecções por Henipavirus/tratamento farmacológico , Humanos , Fatores Imunológicos/uso terapêutico , Linfócitos/imunologia , SARS-CoV-2/fisiologia , Síndrome Respiratória Aguda Grave/tratamento farmacológico , Síndrome Respiratória Aguda Grave/imunologia , Tratamento Farmacológico da COVID-19
7.
Viruses ; 13(7)2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34372504

RESUMO

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.


Assuntos
Quirópteros/virologia , Saúde Global , Infecções por Henipavirus/prevenção & controle , Henipavirus/patogenicidade , Pandemias/prevenção & controle , Viroses/prevenção & controle , Zoonoses/virologia , Animais , Infecções por Henipavirus/epidemiologia , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/transmissão , Especificidade de Hospedeiro , Humanos , Imunidade Inata , Vírus Nipah/patogenicidade , Viroses/imunologia , Viroses/transmissão , Zoonoses/prevenção & controle , Zoonoses/transmissão
8.
Cells ; 10(6)2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34070626

RESUMO

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.


Assuntos
Células Endoteliais , Infecções por Henipavirus , Miócitos de Músculo Liso , Animais , Chlorocebus aethiops , Suscetibilidade a Doenças , Células Endoteliais/imunologia , Células Endoteliais/virologia , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Humanos , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/virologia , Vírus Nipah , Células Vero , Replicação Viral
9.
Nat Struct Mol Biol ; 28(5): 426-434, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33927387

RESUMO

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.


Assuntos
Anticorpos Antivirais/imunologia , Anticorpos Amplamente Neutralizantes/imunologia , Vírus Hendra/imunologia , Vírus Nipah/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Monoclonais Humanizados/imunologia , Células CHO , Cricetulus , Reações Cruzadas , Células HEK293 , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/prevenção & controle , Humanos , Camundongos , Internalização do Vírus
10.
J Gen Virol ; 102(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33054904

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , COVID-19/diagnóstico , Infecções por Henipavirus/diagnóstico , Ensaios de Triagem em Larga Escala , Infecções por Vírus Respiratório Sincicial/diagnóstico , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/metabolismo , COVID-19/imunologia , COVID-19/virologia , Fusão Celular , Convalescença , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Humanos , Soros Imunes/química , Luciferases/genética , Luciferases/metabolismo , Modelos Moleculares , Vírus Nipah/imunologia , Vírus Nipah/patogenicidade , Conformação Proteica , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Suínos , Inibidores de Proteínas Virais de Fusão/química , Inibidores de Proteínas Virais de Fusão/metabolismo , Inibidores de Proteínas Virais de Fusão/farmacologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
11.
mSphere ; 5(6)2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33328346

RESUMO

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.


Assuntos
Núcleo Celular/química , Infecções por Henipavirus/metabolismo , Vírus Nipah/fisiologia , Fatores de Transcrição STAT/metabolismo , Proteínas Virais/metabolismo , Células HEK293 , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Humanos , Imunidade Inata/imunologia , Fosforilação , Fatores de Transcrição STAT/genética , Transdução de Sinais , Transativadores/metabolismo , Proteínas Virais/genética
12.
Sci Rep ; 10(1): 18256, 2020 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-33106487

RESUMO

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.


Assuntos
Alanina/genética , Anticorpos Monoclonais/metabolismo , Simulação por Computador , Glicoproteínas/metabolismo , Infecções por Henipavirus/virologia , Vírus Nipah/metabolismo , Proteínas do Envelope Viral/metabolismo , Alanina/química , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/metabolismo , Epitopos/imunologia , Glicoproteínas/química , Glicoproteínas/genética , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Vírus Nipah/imunologia , Vírus Nipah/isolamento & purificação , Elementos Estruturais de Proteínas/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
13.
Annu Rev Virol ; 7(1): 447-473, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32991264

RESUMO

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.


Assuntos
Doenças Transmissíveis Emergentes/prevenção & controle , Vírus Hendra/imunologia , Infecções por Henipavirus/prevenção & controle , Vírus Nipah/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Quirópteros/virologia , Doenças Transmissíveis Emergentes/imunologia , Doenças Transmissíveis Emergentes/virologia , Modelos Animais de Doenças , Infecções por Henipavirus/imunologia , Humanos , Camundongos , Zoonoses Virais/prevenção & controle , Zoonoses Virais/transmissão
14.
Cells ; 9(8)2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32824665

RESUMO

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.


Assuntos
Transporte Ativo do Núcleo Celular/imunologia , Vírus Hendra/metabolismo , Infecções por Henipavirus/imunologia , Proteínas Intrinsicamente Desordenadas/metabolismo , Vírus Nipah/metabolismo , Membrana Nuclear/metabolismo , Transdução de Sinais/imunologia , Proteínas Virais/metabolismo , Infecções por Henipavirus/metabolismo , Infecções por Henipavirus/virologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade Inata , Interferons/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Receptores de Reconhecimento de Padrão/metabolismo , Proteínas Virais/química
15.
Nat Commun ; 11(1): 3849, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32737300

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr Virus (EBV) establish life-long infections and are associated with malignancies. Striking geographic variation in incidence and the fact that virus alone is insufficient to cause disease, suggests other co-factors are involved. Here we present epidemiological analysis and genome-wide association study (GWAS) in 4365 individuals from an African population cohort, to assess the influence of host genetic and non-genetic factors on virus antibody responses. EBV/KSHV co-infection (OR = 5.71(1.58-7.12)), HIV positivity (OR = 2.22(1.32-3.73)) and living in a more rural area (OR = 1.38(1.01-1.89)) are strongly associated with immunogenicity. GWAS reveals associations with KSHV antibody response in the HLA-B/C region (p = 6.64 × 10-09). For EBV, associations are identified for VCA (rs71542439, p = 1.15 × 10-12). Human leucocyte antigen (HLA) and trans-ancestry fine-mapping substantiate that distinct variants in HLA-DQA1 (p = 5.24 × 10-44) are driving associations for EBNA-1 in Africa. This study highlights complex interactions between KSHV and EBV, in addition to distinct genetic architectures resulting in important differences in pathogenesis and transmission.


Assuntos
Anticorpos Antivirais/biossíntese , Resistência à Doença/genética , Infecções por Vírus Epstein-Barr/genética , Infecções por Henipavirus/genética , Interações Hospedeiro-Patógeno/genética , Sarcoma de Kaposi/genética , Adolescente , Adulto , Antígenos Virais/genética , Antígenos Virais/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Coinfecção , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Antígenos Nucleares do Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Feminino , Expressão Gênica , Estudo de Associação Genômica Ampla , HIV/genética , HIV/imunologia , HIV/patogenicidade , Cadeias alfa de HLA-DQ/genética , Cadeias alfa de HLA-DQ/imunologia , Infecções por Henipavirus/epidemiologia , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/imunologia , Herpesvirus Humano 8/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , População Rural , Sarcoma de Kaposi/epidemiologia , Sarcoma de Kaposi/imunologia , Sarcoma de Kaposi/virologia , Uganda/epidemiologia , População Urbana
16.
J Immunol Res ; 2020: 2567957, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32377531

RESUMO

BACKGROUND: Nipah belongs to the genus Henipavirus and the Paramyxoviridae family. It is an endemic most commonly found at South Asia and has first emerged in Malaysia in 1998. Bats are found to be the main reservoir for this virus, causing disease in both humans and animals. The last outbreak has occurred in May 2018 in Kerala. It is characterized by high pathogenicity and fatality rates which varies from 40% to 70% depending on the severity of the disease and on the availability of adequate healthcare facilities. Currently, there are no antiviral drugs available for NiV disease and the treatment is just supportive. Clinical presentations for this virus range from asymptomatic infection to fatal encephalitis. OBJECTIVE: This study is aimed at predicting an effective epitope-based vaccine against glycoprotein G of Nipah henipavirus, using immunoinformatics approaches. METHODS AND MATERIALS: Glycoprotein G of the Nipah virus sequence was retrieved from NCBI. Different prediction tools were used to analyze the epitopes, namely, BepiPred-2.0: Sequential B Cell Epitope Predictor for B cell and T cell MHC classes II and I. Then, the proposed peptides were docked using Autodock 4.0 software program. Results and Conclusions. The two peptides TVYHCSAVY and FLIDRINWI have showed a very strong binding affinity to MHC class I and MHC class II alleles. Furthermore, considering the conservancy, the affinity, and the population coverage, the peptide FLIDRINWIT is highly suitable to be utilized to formulate a new vaccine against glycoprotein G of Nipah henipavirus. An in vivo study for the proposed peptides is also highly recommended.


Assuntos
Antígenos Virais/genética , Epitopos/genética , Glicosídeo Hidrolases/genética , Infecções por Henipavirus/imunologia , Vírus Nipah/fisiologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Virais/imunologia , Antígenos Virais/metabolismo , Sudeste Asiático/epidemiologia , Biologia Computacional , Doenças Endêmicas , Mapeamento de Epitopos , Epitopos/imunologia , Epitopos/metabolismo , Glicosídeo Hidrolases/metabolismo , Antígenos HLA/metabolismo , Infecções por Henipavirus/epidemiologia , Humanos , Malásia/epidemiologia , Simulação de Acoplamento Molecular , Ligação Proteica , Infecções Respiratórias , Vacinação
17.
PLoS Pathog ; 16(3): e1008412, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32226041

RESUMO

Bats are the natural reservoir host for a number of zoonotic viruses, including Hendra virus (HeV) which causes severe clinical disease in humans and other susceptible hosts. Our understanding of the ability of bats to avoid clinical disease following infection with viruses such as HeV has come predominantly from in vitro studies focusing on innate immunity. Information on the early host response to infection in vivo is lacking and there is no comparative data on responses in bats compared with animals that succumb to disease. In this study, we examined the sites of HeV replication and the immune response of infected Australian black flying foxes and ferrets at 12, 36 and 60 hours post exposure (hpe). Viral antigen was detected at 60 hpe in bats and was confined to the lungs whereas in ferrets there was evidence of widespread viral RNA and antigen by 60 hpe. The mRNA expression of IFNs revealed antagonism of type I and III IFNs and a significant increase in the chemokine, CXCL10, in bat lung and spleen following infection. In ferrets, there was an increase in the transcription of IFN in the spleen following infection. Liquid chromatography tandem mass spectrometry (LC-MS/MS) on lung tissue from bats and ferrets was performed at 0 and 60 hpe to obtain a global overview of viral and host protein expression. Gene Ontology (GO) enrichment analysis of immune pathways revealed that six pathways, including a number involved in cell mediated immunity were more likely to be upregulated in bat lung compared to ferrets. GO analysis also revealed enrichment of the type I IFN signaling pathway in bats and ferrets. This study contributes important comparative data on differences in the dissemination of HeV and the first to provide comparative data on the activation of immune pathways in bats and ferrets in vivo following infection.


Assuntos
Antígenos Virais/imunologia , Vírus Hendra/imunologia , Infecções por Henipavirus/imunologia , Imunidade Celular , Imunidade Inata , Pulmão/imunologia , Modelos Imunológicos , Animais , Antígenos Virais/genética , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Quirópteros , Furões , Vírus Hendra/genética , Infecções por Henipavirus/genética , Infecções por Henipavirus/patologia , Interferons/genética , Interferons/imunologia , Pulmão/patologia , Pulmão/virologia , Especificidade da Espécie
18.
J Infect Dis ; 221(Suppl 4): S401-S406, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-31853535

RESUMO

Interferon (IFN) type I plays a critical role in the protection of mice from lethal Nipah virus (NiV) infection, but mechanisms responsible for IFN-I induction remain unknown. In the current study, we demonstrated the critical role of the mitochondrial antiviral signaling protein signaling pathway in IFN-I production and NiV replication in murine embryonic fibroblasts in vitro, and the redundant but essential roles of both mitochondrial antiviral signaling protein and myeloid differentiation primary response 88 adaptors, but not toll/interleukin-1 receptor/resistance [TIR] domain-containing adaptor-inducing IFN-ß (TRIF), in the control of NiV infection in mice. These results reveal potential novel targets for antiviral intervention and help in understanding NiV immunopathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Fator 88 de Diferenciação Mieloide/metabolismo , Vírus Nipah , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , Regulação da Expressão Gênica/imunologia , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
19.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-31656582

RESUMO

Nipah virus (NiV) is a highly lethal zoonotic paramyxovirus that emerged at the end of last century as a human pathogen capable of causing severe acute respiratory infection and encephalitis. Although NiV provokes serious diseases in numerous mammalian species, the infection seems to be asymptomatic in NiV natural hosts, the fruit bats, which provide a continuous virus source for further outbreaks. Consecutive human-to-human transmission has been frequently observed during outbreaks in Bangladesh and India. NiV was shown to interfere with the innate immune response and interferon type I signaling, restraining the anti-viral response and permitting viral spread. Studies of adaptive immunity in infected patients and animal models have suggested an unbalanced immune response during NiV infection. Here, we summarize some of the recent studies of NiV pathogenesis and NiV-induced modulation of both innate and adaptive immune responses, as well as the development of novel prophylactic and therapeutic approaches, necessary to control this highly lethal emerging infection.


Assuntos
Imunidade Adaptativa , Infecções por Henipavirus/imunologia , Imunidade Inata , Vírus Nipah/patogenicidade , Animais , Bangladesh , Humanos , Índia , Vírus Nipah/imunologia
20.
PLoS Negl Trop Dis ; 13(6): e0007454, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31166946

RESUMO

The ability to appropriately mimic human disease is critical for using animal models as a tool for understanding virus pathogenesis. In the case of Nipah virus (NiV), infection of humans appears to occur either through inhalation, contact with or consumption of infected material. In two of these circumstances, respiratory or sinusoidal exposure represents a likely route of infection. In this study, intermediate-size aerosol particles (~7 µm) of NiV-Malaysia were used to mimic potential routes of exposure by focusing viral deposition in the upper respiratory tract. Our previous report showed this route of exposure extended the disease course and a single animal survived the infection. Here, analysis of the peripheral immune response found minimal evidence of systemic inflammation and depletion of B cells during acute disease. However, the animal that survived infection developed an early IgM response with rapid development of neutralizing antibodies that likely afforded protection. The increase in NiV-specific antibodies correlated with an expansion of the B cell population in the survivor. Cell-mediated immunity was not clearly apparent in animals that succumbed during the acute phase of disease. However, CD4+ and CD8+ effector memory cells increased in the survivor with correlating increases in cytokines and chemokines associated with cell-mediated immunity. Interestingly, kinetic changes of the CD4+ and CD8bright T cell populations over the course of acute disease were opposite from animals that succumbed to infection. In addition, increases in NK cells and basophils during convalescence of the surviving animal were also evident, with viral antigen found in NK cells. These data suggest that a systemic inflammatory response and "cytokine storm" are not major contributors to NiV-Malaysia pathogenesis in the AGM model using this exposure route. Further, these data demonstrate that regulation of cell-mediated immunity, in addition to rapid production of NiV specific antibodies, may be critical for surviving NiV infection.


Assuntos
Aerossóis , Infecções por Henipavirus/imunologia , Imunidade Humoral , Exposição por Inalação , Vírus Nipah/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Chlorocebus aethiops , Citocinas/análise , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Celular , Imunoglobulina M/sangue , Células Matadoras Naturais/imunologia , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...