Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Virol ; 98(3): e0183823, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38426726

ABSTRACT

Nipah virus (NiV) is a highly lethal, zoonotic Henipavirus (HNV) that causes respiratory and neurological signs and symptoms in humans. Similar to other paramyxoviruses, HNVs mediate entry into host cells through the concerted actions of two surface glycoproteins: a receptor-binding protein (RBP) that mediates attachment and a fusion glycoprotein (F) that triggers fusion in an RBP-dependent manner. NiV uses ephrin-B2 (EFNB2) and ephrin-B3 (EFNB3) as entry receptors. Ghana virus (GhV), a novel HNV identified in a Ghanaian bat, uses EFNB2 but not EFNB3. In this study, we employ a structure-informed approach to identify receptor-interfacing residues and systematically introduce GhV-RBP residues into a NiV-RBP backbone to uncover the molecular determinants of EFNB3 usage. We reveal two regions that severely impair EFNB3 binding by NiV-RBP and EFNB3-mediated entry by NiV pseudotyped viral particles. Further analyses uncovered two-point mutations (NiVN557SGhV and NiVY581TGhV) pivotal for this phenotype. Moreover, we identify NiV interaction with Y120 of EFNB3 as important for the usage of this receptor. Beyond these EFNB3-related findings, we reveal two domains that restrict GhV binding of EFNB2, confirm the HNV-head as an immunodominant target for polyclonal and monoclonal antibodies, and describe putative epitopes for GhV- and NiV-specific monoclonal antibodies. Cumulatively, the work presented here generates useful reagents and tools that shed insight to residues important for NiV usage of EFNB3, reveal regions critical for GhV binding of EFNB2, and describe putative HNV antibody-binding epitopes. IMPORTANCE: Hendra virus and Nipah virus (NiV) are lethal, zoonotic Henipaviruses (HNVs) that cause respiratory and neurological clinical features in humans. Since their initial outbreaks in the 1990s, several novel HNVs have been discovered worldwide, including Ghana virus. Additionally, there is serological evidence of zoonotic transmission, lending way to concerns about future outbreaks. HNV infection of cells is mediated by the receptor-binding protein (RBP) and the Fusion protein (F). The work presented here identifies NiV RBP amino acids important for the usage of ephrin-B3 (EFNB3), a receptor highly expressed in neurons and predicted to be important for neurological clinical features caused by NiV. This study also characterizes epitopes recognized by antibodies against divergent HNV RBPs. Together, this sheds insight to amino acids critical for HNV receptor usage and antibody binding, which is valuable for future studies investigating determinants of viral pathogenesis and developing antibody therapies.


Subject(s)
Henipavirus Infections , Henipavirus , Receptors, Virus , Humans , Amino Acids/genetics , Antibodies, Monoclonal/metabolism , Carrier Proteins/metabolism , Ephrin-B3/genetics , Ephrin-B3/chemistry , Ephrin-B3/metabolism , Epitopes/genetics , Epitopes/metabolism , Ghana , Hendra Virus/metabolism , Henipavirus/classification , Henipavirus/genetics , Henipavirus/metabolism , Mutagenesis , Nipah Virus/metabolism , Viral Envelope Proteins/genetics , Virus Internalization , Receptors, Virus/metabolism
2.
Nat Commun ; 15(1): 812, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38280880

ABSTRACT

Langya Henipavirus (LayV) infection is an emerging zoonotic disease that has been causing respiratory symptoms in China since 2019. For virus entry, LayV's genome encodes the fusion protein F and the attachment glycoprotein G. However, the structural and functional information regarding LayV-G remains unclear. In this study, we revealed that LayV-G cannot bind to the receptors found in other HNVs, such as ephrin B2/B3, and it shows different antigenicity from HeV-G and NiV-G. Furthermore, we determined the near full-length structure of LayV-G, which displays a distinct mushroom-shaped configuration, distinguishing it from other attachment glycoproteins of HNV. The stalk and transmembrane regions resemble the stem and root of mushroom and four downward-tilted head domains as mushroom cap potentially interact with the F protein and influence membrane fusion process. Our findings enhance the understanding of emerging HNVs that cause human diseases through zoonotic transmission and provide implication for LayV related vaccine development.


Subject(s)
Henipavirus Infections , Henipavirus , Nipah Virus , Humans , Cryoelectron Microscopy , Henipavirus/genetics , Glycoproteins/metabolism , China , Nipah Virus/metabolism , Virus Internalization , Viral Envelope Proteins/metabolism
3.
Methods Mol Biol ; 2682: 33-58, 2023.
Article in English | MEDLINE | ID: mdl-37610572

ABSTRACT

Henipaviruses possess two envelope glycoproteins, the attachment (G) and the fusion (F) proteins that mediate cellular entry and are the major targets of virus-neutralizing antibody responses. Recombinant expression technologies have been used to produce soluble G and F proteins (sG and sF) that retain native-like oligomeric conformations and epitopes, which are advantageous for the development and characterization of vaccines and antiviral antibody therapeutics. In addition to Hendra virus and Nipah virus tetrameric sG and trimeric sF production, we also describe the expression and purification of Cedar virus tetrameric sG and Ghana virus trimeric sF glycoproteins. These henipavirus glycoproteins were also used as immunizing antigens to generate monoclonal antibodies, and binding was demonstrated with a pan-henipavirus multiplex microsphere immunoassay.


Subject(s)
Henipavirus , Henipavirus/genetics , Antibodies, Blocking , Antibodies, Monoclonal
5.
J Virol ; 96(18): e0092122, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36040175

ABSTRACT

The genus Henipavirus (family Paramyxoviridae) currently comprises seven viruses, four of which have demonstrated prior evidence of zoonotic capacity. These include the biosafety level 4 agents Hendra (HeV) and Nipah (NiV) viruses, which circulate naturally in pteropodid fruit bats. Here, we describe and characterize Angavokely virus (AngV), a divergent henipavirus identified in urine samples from wild, Madagascar fruit bats. We report the nearly complete 16,740-nucleotide genome of AngV, which encodes the six major henipavirus structural proteins (nucleocapsid, phosphoprotein, matrix, fusion, glycoprotein, and L polymerase). Within the phosphoprotein (P) gene, we identify an alternative start codon encoding the AngV C protein and a putative mRNA editing site where the insertion of one or two guanine residues encodes, respectively, additional V and W proteins. In other paramyxovirus systems, C, V, and W are accessory proteins involved in antagonism of host immune responses during infection. Phylogenetic analysis suggests that AngV is ancestral to all four previously described bat henipaviruses-HeV, NiV, Cedar virus (CedV), and Ghanaian bat virus (GhV)-but evolved more recently than rodent- and shrew-derived henipaviruses, Mojiang (MojV), Gamak (GAKV), and Daeryong (DARV) viruses. Predictive structure-based alignments suggest that AngV is unlikely to bind ephrin receptors, which mediate cell entry for all other known bat henipaviruses. Identification of the AngV receptor is needed to clarify the virus's potential host range. The presence of V and W proteins in the AngV genome suggest that the virus could be pathogenic following zoonotic spillover. IMPORTANCE Henipaviruses include highly pathogenic emerging zoonotic viruses, derived from bat, rodent, and shrew reservoirs. Bat-borne Hendra (HeV) and Nipah (NiV) are the most well-known henipaviruses, for which no effective antivirals or vaccines for humans have been described. Here, we report the discovery and characterization of a novel henipavirus, Angavokely virus (AngV), isolated from wild fruit bats in Madagascar. Genomic characterization of AngV reveals all major features associated with pathogenicity in other henipaviruses, suggesting that AngV could be pathogenic following spillover to human hosts. Our work suggests that AngV is an ancestral bat henipavirus that likely uses viral entry pathways distinct from those previously described for HeV and NiV. In Madagascar, bats are consumed as a source of human food, presenting opportunities for cross-species transmission. Characterization of novel henipaviruses and documentation of their pathogenic and zoonotic potential are essential to predicting and preventing the emergence of future zoonoses that cause pandemics.


Subject(s)
Chiroptera , Genome, Viral , Henipavirus Infections , Henipavirus , Nipah Virus , Animals , Chiroptera/genetics , Genome, Viral/genetics , Glycoproteins/genetics , Henipavirus/classification , Henipavirus/genetics , Henipavirus Infections/virology , Humans , Madagascar , Nipah Virus/genetics , Phylogeny , Urine/virology , Zoonoses/genetics
6.
Viruses ; 13(10)2021 10 07.
Article in English | MEDLINE | ID: mdl-34696450

ABSTRACT

Paramyxoviruses, negative-sense single-stranded RNA viruses, pose a critical threat to human public health. Currently, 78 species, 17 genera, and 4 subfamilies of paramyxoviruses are harbored by multiple natural reservoirs, including rodents, bats, birds, reptiles, and fish. Henipaviruses are critical zoonotic pathogens that cause severe acute respiratory distress and neurological diseases in humans. Using reverse transcription-polymerase chain reaction, 115 Crocidura species individuals were examined for the prevalence of paramyxovirus infections. Paramyxovirus RNA was observed in 26 (22.6%) shrews collected at five trapping sites, Republic of Korea. Herein, we report two genetically distinct novel paramyxoviruses (genus: Henipavirus): Gamak virus (GAKV) and Daeryong virus (DARV) isolated from C. lasiura and C. shantungensis, respectively. Two GAKVs and one DARV were nearly completely sequenced using next-generation sequencing. GAKV and DARV contain six genes (3'-N-P-M-F-G-L-5') with genome sizes of 18,460 nucleotides and 19,471 nucleotides, respectively. The phylogenetic inference demonstrated that GAKV and DARV form independent genetic lineages of Henipavirus in Crocidura species. GAKV-infected human lung epithelial cells elicited the induction of type I/III interferons, interferon-stimulated genes, and proinflammatory cytokines. In conclusion, this study contributes further understandings of the molecular prevalence, genetic characteristics and diversity, and zoonotic potential of novel paramyxoviruses in shrews.


Subject(s)
Henipavirus/classification , Henipavirus/genetics , Paramyxovirinae/classification , Paramyxovirinae/genetics , Phylogeny , Shrews/virology , Animals , Biodiversity , Birds/virology , Chiroptera/virology , Fishes/virology , Henipavirus/isolation & purification , High-Throughput Nucleotide Sequencing , Interferons , Paramyxovirinae/isolation & purification , RNA Viruses/classification , Reptiles/virology , Republic of Korea , Rodentia/virology , Viral Zoonoses/virology
7.
J Virol ; 95(20): e0066621, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34288734

ABSTRACT

Cedar virus (CedV) is a nonpathogenic member of the Henipavirus (HNV) genus of emerging viruses, which includes the deadly Nipah (NiV) and Hendra (HeV) viruses. CedV forms syncytia, a hallmark of henipaviral and paramyxoviral infections and pathogenicity. However, the intrinsic fusogenic capacity of CedV relative to NiV or HeV remains unquantified. HNV entry is mediated by concerted interactions between the attachment (G) and fusion (F) glycoproteins. Upon receptor binding by the HNV G head domain, a fusion-activating G stalk region is exposed and triggers F to undergo a conformational cascade that leads to viral entry or cell-cell fusion. Here, we demonstrate quantitatively that CedV is inherently significantly less fusogenic than NiV at equivalent G and F cell surface expression levels. We then generated and tested six headless CedV G mutants of distinct C-terminal stalk lengths, surprisingly revealing highly hyperfusogenic cell-cell fusion phenotypes 3- to 4-fold greater than wild-type CedV levels. Additionally, similarly to NiV, a headless HeV G mutant yielded a less pronounced hyperfusogenic phenotype compared to wild-type HeV. Further, coimmunoprecipitation and cell-cell fusion assays revealed heterotypic NiV/CedV functional G/F bidentate interactions, as well as evidence of HNV G head domain involvement beyond receptor binding or G stalk exposure. All evidence points to the G head/stalk junction being key to modulating HNV fusogenicity, supporting the notion that head domains play several distinct and central roles in modulating stalk domain fusion promotion. Further, this study exemplifies how CedV may help elucidate important mechanistic underpinnings of HNV entry and pathogenicity. IMPORTANCE The Henipavirus genus in the Paramyxoviridae family includes the zoonotic Nipah (NiV) and Hendra (HeV) viruses. NiV and HeV infections often cause fatal encephalitis and pneumonia, but no vaccines or therapeutics are currently approved for human use. Upon viral entry, Henipavirus infections yield the formation of multinucleated cells (syncytia). Viral entry and cell-cell fusion are mediated by the attachment (G) and fusion (F) glycoproteins. Cedar virus (CedV), a nonpathogenic henipavirus, may be a useful tool to gain knowledge on henipaviral pathogenicity. Here, using homotypic and heterotypic full-length and headless CedV, NiV, and HeV G/F combinations, we discovered that CedV G/F are significantly less fusogenic than NiV or HeV G/F, and that the G head/stalk junction is key to modulating cell-cell fusion, refining the mechanism of henipaviral membrane fusion events. Our study exemplifies how CedV may be a useful tool to elucidate broader mechanistic understanding for the important henipaviruses.


Subject(s)
Henipavirus/metabolism , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Giant Cells/metabolism , Glycoproteins/genetics , HEK293 Cells , Henipavirus/genetics , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Humans , Membrane Fusion/physiology , Receptors, Virus/metabolism , Viral Envelope Proteins/genetics , Viral Fusion Proteins/physiology , Virus Attachment , Virus Internalization
8.
Viruses ; 12(4)2020 04 23.
Article in English | MEDLINE | ID: mdl-32340278

ABSTRACT

The genus Henipavirus (HNVs) includes two fatal viruses, namely Nipah virus (NiV) and Hendra virus (HeV). Since 1994, NiV and HeV have been endemic to the Asia-Pacific region and responsible for more than 600 cases of infections. Two emerging HNVs, Ghana virus (GhV) and Mojiang virus (MojV), are speculated to be associated with unrecognized human diseases in Africa and China, respectively. Despite many efforts to develop vaccines against henipaviral diseases, there is presently no licensed human vaccine. As HNVs are highly pathogenic and diverse, it is necessary to develop universal vaccines to prevent future outbreaks. The attachment enveloped glycoprotein (G protein) of HNVs mediates HNV attachment to the host cell's surface receptors. G proteins have been used as a protective antigen in many vaccine candidates for HNVs. We performed quantitative studies on the antibody responses elicited by the G proteins of NiV, HeV, GhV, and MojV. We found that the G proteins of NiV and HeV elicited only a limited cross-reactive antibody response. Further, there was no cross-protection between MojV, GhV, and highly pathogenic HNVs. We then constructed a bivalent vaccine where the G proteins of NiV and HeV were fused with the human IgG1 Fc domain. The immunogenicity of the bivalent vaccine was compared with that of monovalent vaccines. Our results revealed that the Fc-based bivalent vaccine elicited a potent antibody response against both NiV and HeV. We also constructed a tetravalent Fc heterodimer fusion protein that contains the G protein domains of four HNVs. Immunization with the tetravalent vaccine elicited broad antibody responses against NiV, HeV, GhV, and MojV in mice, indicating compatibility among the four antigens in the Fc-fusion protein. These data suggest that our novel bivalent and tetravalent Fc-fusion proteins may be efficient candidates to prevent HNV infection.


Subject(s)
Broadly Neutralizing Antibodies/immunology , Henipavirus Infections/prevention & control , Henipavirus/genetics , Henipavirus/immunology , Immunoglobulin Fc Fragments/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hendra Virus/immunology , Henipavirus/classification , Mice , Neutralization Tests , Nipah Virus/immunology , Phylogeny , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
9.
J Infect Dis ; 221(Suppl 4): S383-S388, 2020 05 11.
Article in English | MEDLINE | ID: mdl-31784761

ABSTRACT

Viruses in the genus Henipavirus encompass 2 highly pathogenic emerging zoonotic pathogens, Hendra virus (HeV) and Nipah virus (NiV). Despite the impact on human health, there is currently limited full-genome sequence information available for henipaviruses. This lack of full-length genomes hampers our ability to understand the molecular drivers of henipavirus emergence. Furthermore, rapidly deployable viral genome sequencing can be an integral part of outbreak response and epidemiological investigations to study transmission chains. In this study, we describe the development of a reverse-transcription, long-range polymerase chain reaction (LRPCR) assay for efficient genome amplification of NiV, HeV, and a related non-pathogenic henipavirus, Cedar virus (CedPV). We then demonstrated the utility of our method by amplifying partial viral genomes from 6 HeV-infected tissue samples from Syrian hamsters and 4 tissue samples from a NiV-infected African green monkey with viral loads as low as 52 genome copies/mg. We subsequently sequenced the amplified genomes on the portable Oxford Nanopore MinION platform and analyzed the data using a newly developed field-deployable bioinformatic pipeline. Our LRPCR assay allows amplification and sequencing of 2 or 4 amplicons in semi-nested reactions. Coupled with an easy-to-use bioinformatics pipeline, this method is particularly useful in the field during outbreaks in resource-poor environments.


Subject(s)
Henipavirus/genetics , High-Throughput Nucleotide Sequencing/methods , Polymerase Chain Reaction/methods , Genome, Viral , RNA, Viral
10.
Viruses ; 11(9)2019 08 29.
Article in English | MEDLINE | ID: mdl-31470664

ABSTRACT

The Ghana virus (GhV) is phylogenetically related to the zoonotic henipaviruses Nipah (NiV) and Hendra virus. Although GhV uses the highly conserved receptor ephrin-B2, the fusogenicity is restricted to cell lines of bat origin. Furthermore, the surface expression of the GhV attachment glycoprotein (G) is reduced compared to NiV and most of this protein is retained in the endoplasmic reticulum (ER). Here, we generated truncated as well as chimeric GhV G proteins and investigated the influence of the structural domains (cytoplasmic tail, transmembrane domain, ectodomain) of this protein on the intracellular transport and the fusogenicity following coexpression with the GhV fusion protein (F). We demonstrate that neither the cytoplasmic tail nor the transmembrane domain is responsible for the intracellular retention of GhV G. Furthermore, the cytoplasmic tail of GhV G modulates the fusogenicity of GhV F and therefore controls the species-restricted fusogenicity of the GhV surface glycoproteins.


Subject(s)
Cell Fusion , Henipavirus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , Animals , Cell Line , Chiroptera , Chlorocebus aethiops , HEK293 Cells , Henipavirus/genetics , Host Specificity , Humans , Protein Domains , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Vero Cells , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics
11.
Viruses ; 10(11)2018 11 01.
Article in English | MEDLINE | ID: mdl-30388838

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are among a group of emerging bat-borne paramyxoviruses that have crossed their species-barrier several times by infecting several hosts with a high fatality rate in human beings. Despite the fatal nature of their infection, a comprehensive study to explore their evolution and adaptation in different hosts is lacking. A study of codon usage patterns in henipaviruses may provide some fruitful insight into their evolutionary processes of synonymous codon usage and host-adapted evolution. Here, we performed a systematic evolutionary and codon usage bias analysis of henipaviruses. We found a low codon usage bias in the coding sequences of henipaviruses and that natural selection, mutation pressure, and nucleotide compositions shapes the codon usage patterns of henipaviruses, with natural selection being more important than the others. Also, henipaviruses showed the highest level of adaptation to bats of the genus Pteropus in the codon adaptation index (CAI), relative to the codon de-optimization index (RCDI), and similarity index (SiD) analyses. Furthermore, a comparison to recently identified henipa-like viruses indicated a high tRNA adaptation index of henipaviruses for human beings, mainly due to F, G and L proteins. Consequently, the study concedes the substantial emergence of henipaviruses in human beings, particularly when paired with frequent exposure to direct/indirect bat excretions.


Subject(s)
Codon , Evolution, Molecular , Henipavirus Infections/virology , Henipavirus/genetics , Host Specificity , Host-Pathogen Interactions , Selection, Genetic , Adaptation, Biological , Animals , Chiroptera/virology , Genome, Viral , Genomics/methods , Henipavirus/classification , Humans , Phylogeny
12.
Nat Commun ; 9(1): 3057, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30076298

ABSTRACT

Recent studies indicate that nucleoli play critical roles in the DNA-damage response (DDR) via interaction of DDR machinery including NBS1 with nucleolar Treacle protein, a key mediator of ribosomal RNA (rRNA) transcription and processing. Here, using proteomics, confocal and single molecule super-resolution imaging, and infection under biosafety level-4 containment, we show that this nucleolar DDR pathway is targeted by infectious pathogens. We find that the matrix proteins of Hendra virus and Nipah virus, highly pathogenic viruses of the Henipavirus genus in the order Mononegavirales, interact with Treacle and inhibit its function, thereby silencing rRNA biogenesis, consistent with mimicking NBS1-Treacle interaction during a DDR. Furthermore, inhibition of Treacle expression/function enhances henipavirus production. These data identify a mechanism for viral modulation of host cells by appropriating the nucleolar DDR and represent, to our knowledge, the first direct intranucleolar function for proteins of any mononegavirus.


Subject(s)
Cell Nucleolus/physiology , Cell Nucleolus/virology , DNA Damage/physiology , Hendra Virus/physiology , Nipah Virus/physiology , Cell Cycle Proteins/metabolism , HEK293 Cells , HeLa Cells , Henipavirus/genetics , Henipavirus Infections , Host-Pathogen Interactions/physiology , Humans , Mononegavirales/genetics , Nuclear Proteins/metabolism , Nucleoproteins/metabolism , Proteomics , RNA, Ribosomal/biosynthesis , Viral Proteins/metabolism
13.
Vet Microbiol ; 218: 90-97, 2018 May.
Article in English | MEDLINE | ID: mdl-29685227

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are highly pathogenic henipaviruses originating from fruit bats in Australia and Asia that can cause severe infections in livestock and humans. In recent years, also African bat henipaviruses were identified at the nucleic acid level. To assess their potential to replicate in non-bat species, several studies were performed to characterize the two surface glycoproteins required for virus entry and spread by cell-cell fusion. It has been shown that surface expression and fusion-helper function of the receptor-binding G protein of Kumasi virus (KV), the prototypic Ghanaian bat henipavirus, is reduced compared to other non-African henipavirus G proteins. Immunostainings and pulse-chase analysis revealed a delayed export of KV G from the ER. As defects in oligomerization of viral glycoproteins can be responsible for limited surface transport thereby restricting the bioactivity, we analyzed the oligomerization pattern of KV G. In contrast to HeV and NiV whose G proteins are known to be expressed at a dimer-tetramer ratio of 1:1, KV G almost exclusively formed stable tetramers or higher oligomers. KV G also showed less stringent requirements for defined stalk cysteines to form dimers and tetramers. Interestingly, any changes in the oligomeric forms negatively affected the fusion-helper activity although surface expression and receptor binding was unchanged. This clearly indicates that the formation of mostly higher oligomeric KV G forms is not a deficiency responsible for ER retention, but is rather a basic structural feature essential for the bioactivity of this African bat henipavirus glycoprotein.


Subject(s)
Chiroptera/virology , GTP-Binding Proteins/chemistry , Henipavirus/metabolism , Membrane Glycoproteins/chemistry , Viral Envelope Proteins , Animals , Endoplasmic Reticulum/virology , GTP-Binding Proteins/metabolism , Ghana/epidemiology , Henipavirus/chemistry , Henipavirus/genetics , Henipavirus Infections/epidemiology , Henipavirus Infections/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization
14.
PLoS Negl Trop Dis ; 12(3): e0006343, 2018 03.
Article in English | MEDLINE | ID: mdl-29538374

ABSTRACT

Henipavirus infection causes severe respiratory and neurological disease in humans that can be fatal. To characterize the pathogenic mechanisms of henipavirus infection in vivo, we performed experimental infections in ferrets followed by genome-wide gene expression analysis of lung and brain tissues. The Hendra, Nipah-Bangladesh, and Nipah-Malaysia strains caused severe respiratory and neurological disease with animals succumbing around 7 days post infection. Despite the presence of abundant viral shedding, animal-to-animal transmission did not occur. The host gene expression profiles of the lung tissue showed early activation of interferon responses and subsequent expression of inflammation-related genes that coincided with the clinical deterioration. Additionally, the lung tissue showed unchanged levels of lymphocyte markers and progressive downregulation of cell cycle genes and extracellular matrix components. Infection in the brain resulted in a limited breadth of the host responses, which is in accordance with the immunoprivileged status of this organ. Finally, we propose a model of the pathogenic mechanisms of henipavirus infection that integrates multiple components of the host responses.


Subject(s)
Henipavirus Infections/genetics , Henipavirus Infections/immunology , Henipavirus/physiology , Host-Pathogen Interactions , Transcriptome , Animals , Brain/metabolism , Brain/virology , Cell Cycle , Disease Models, Animal , Extracellular Matrix/genetics , Ferrets/virology , Hendra Virus/immunology , Hendra Virus/pathogenicity , Henipavirus/genetics , Henipavirus Infections/virology , Humans , Inflammation , Interferons/genetics , Lung/metabolism , Lung/virology , Nipah Virus/immunology , Nipah Virus/pathogenicity , Virus Shedding
15.
Virol J ; 15(1): 56, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29587789

ABSTRACT

BACKGROUND: Hendra virus and Nipah virus are zoonotic viruses that have caused severe to fatal disease in livestock and human populations. The isolation of Cedar virus, a non-pathogenic virus species in the genus Henipavirus, closely-related to the highly pathogenic Hendra virus and Nipah virus offers an opportunity to investigate differences in pathogenesis and receptor tropism among these viruses. METHODS: We constructed full-length cDNA clones of Cedar virus from synthetic oligonucleotides and rescued two replication-competent, recombinant Cedar virus variants: a recombinant wild-type Cedar virus and a recombinant Cedar virus that expresses a green fluorescent protein from an open reading frame inserted between the phosphoprotein and matrix genes. Replication kinetics of both viruses and stimulation of the interferon pathway were characterized in vitro. Cellular tropism for ephrin-B type ligands was qualitatively investigated by microscopy and quantitatively by a split-luciferase fusion assay. RESULTS: Successful rescue of recombinant Cedar virus expressing a green fluorescent protein did not significantly affect virus replication compared to the recombinant wild-type Cedar virus. We demonstrated that recombinant Cedar virus stimulated the interferon pathway and utilized the established Hendra virus and Nipah virus receptor, ephrin-B2, but not ephrin-B3 to mediate virus entry. We further characterized virus-mediated membrane fusion kinetics of Cedar virus with the known henipavirus receptors ephrin-B2 and ephrin-B3. CONCLUSIONS: The recombinant Cedar virus platform may be utilized to characterize the determinants of pathogenesis across the henipaviruses, investigate their receptor tropisms, and identify novel pan-henipavirus antivirals. Moreover, these experiments can be conducted safely under BSL-2 conditions.


Subject(s)
Ephrin-B2/metabolism , Henipavirus Infections/virology , Henipavirus/physiology , Receptors, Virus/metabolism , Cell Fusion , Cell Line , Cytopathogenic Effect, Viral , Genes, Reporter , Green Fluorescent Proteins/genetics , Henipavirus/genetics , Henipavirus/metabolism , Henipavirus/pathogenicity , Henipavirus Infections/metabolism , Interferon Type I/genetics , Neutralization Tests , Protein Binding , Recombination, Genetic , Reverse Genetics , Viral Envelope Proteins/metabolism , Viral Tropism , Virus Internalization , Virus Replication
16.
PLoS One ; 13(2): e0191933, 2018.
Article in English | MEDLINE | ID: mdl-29390028

ABSTRACT

In 2011, an unusually large number of independent Hendra virus outbreaks were recorded on horse properties in Queensland and New South Wales, Australia. Urine from bat colonies adjacent to the outbreak sites were sampled and screened for Hendra and other viruses. Several novel paramyxoviruses were also isolated at different locations. Here one of the novel viruses, named Hervey virus (HerPV), is fully characterized by genome sequencing, annotation, phylogeny and in vitro host range, and its serological cross-reactivity and neutralization patterns are examined. HerPV may have ecological and spatial and temporal patterns similar to Hendra virus and could serve as a sentinel virus for the surveillance of this highly pathogenic virus. The suitability of HerPV as potential sentinel virus is further assessed by determining the serological prevalence of HerPV antibodies in fruit-eating bats from Australia, Indonesia, Papua New Guinea, Tanzania and the Gulf of Guinea, indicating the presence of similar viruses in regions beyond the Australian border.


Subject(s)
Chiroptera/virology , Henipavirus/isolation & purification , Paramyxovirinae/isolation & purification , Africa/epidemiology , Animals , Antibodies, Viral/immunology , Australia/epidemiology , Cell Line , Disease Outbreaks , Henipavirus/genetics , Henipavirus/immunology , Henipavirus Infections/epidemiology , Henipavirus Infections/virology , High-Throughput Nucleotide Sequencing , Indonesia/epidemiology , Microscopy, Confocal , Neutralization Tests , Papua New Guinea/epidemiology , Paramyxovirinae/genetics , Paramyxovirinae/immunology
17.
J Gen Virol ; 98(4): 563-576, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28056216

ABSTRACT

Viruses of the genus Henipavirus of the family Paramyxoviridae are zoonotic pathogens, which have emerged in Southeast Asia, Australia and Africa. Nipah virus (NiV) and Hendra virus are highly virulent pathogens transmitted from bats to animals and humans, while the henipavirus Cedar virus seems to be non-pathogenic in infection studies. The full replication cycle of the Paramyxoviridae occurs in the host cell's cytoplasm, where viral assembly is orchestrated by the matrix (M) protein. Unexpectedly, the NiV-M protein traffics through the nucleus as an essential step to engage the plasma membrane in preparation for viral budding/release. Comparative studies were performed to assess whether M protein nuclear localization is a common feature of the henipaviruses, including the recently sequenced (although not yet isolated) Ghanaian bat henipavirus (Kumasi virus, GH-M74a virus) and Mojiang virus. Live-cell confocal microscopy revealed that nuclear translocation of GFP-fused M protein is conserved between henipaviruses in both human- and bat-derived cell lines. However, the efficiency of M protein nuclear localization and virus-like particle budding competency varied. Additionally, Cedar virus-, Kumasi virus- and Mojiang virus-M proteins were mutated in a bipartite nuclear localization signal, indicating that a key lysine residue is essential for nuclear import, export and induction of budding events, as previously reported for NiV-M. The results of this study suggest that the M proteins of henipaviruses may utilize a similar nucleocytoplasmic trafficking pathway as an essential step during viral replication in both humans and bats.


Subject(s)
Active Transport, Cell Nucleus , Henipavirus/genetics , Henipavirus/physiology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Animals , Henipavirus/isolation & purification , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Localization Signals , Protein Transport , Virosomes/genetics , Virosomes/metabolism
19.
J Gen Virol ; 97(5): 1066-1076, 2016 05.
Article in English | MEDLINE | ID: mdl-26813519

ABSTRACT

The amino-terminal cytoplasmic domains of paramyxovirus attachment glycoproteins include trafficking signals that influence protein processing and cell surface expression. To characterize the role of the cytoplasmic domain in protein expression, fusion support and particle assembly in more detail, we constructed chimeric Nipah virus (NiV) glycoprotein (G) and canine distemper virus (CDV) haemagglutinin (H) proteins carrying the respective heterologous cytoplasmic domain, as well as a series of mutants with progressive deletions in this domain. CDV H retained fusion function and was normally expressed on the cell surface with a heterologous cytoplasmic domain, while the expression and fusion support of NiV G was dramatically decreased when its cytoplasmic domain was replaced with that of CDV H. The cell surface expression and fusion support functions of CDV H were relatively insensitive to cytoplasmic domain deletions, while short deletions in the corresponding region of NiV G dramatically decreased both. In addition, the first 10 residues of the CDV H cytoplasmic domain strongly influence its incorporation into virus-like particles formed by the CDV matrix (M) protein, while the co-expression of NiV M with NiV G had no significant effect on incorporation of G into particles. The cytoplasmic domains of both the CDV H and NiV G proteins thus contribute differently to the virus life cycle.


Subject(s)
Gene Expression Regulation, Viral/physiology , Henipavirus/metabolism , Morbillivirus/metabolism , Viral Envelope Proteins/metabolism , Virus Assembly/physiology , Virus Internalization , Amino Acid Sequence , Henipavirus/genetics , Morbillivirus/genetics , Viral Envelope Proteins/genetics
20.
J Infect Dis ; 213(3): 448-55, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26357909

ABSTRACT

Henipaviruses are zoonotic viruses that can cause severe and acute respiratory diseases and encephalitis in humans. To date, no vaccine or treatments are approved for human use. The presence of neutralizing antibodies is a strong correlate of protection against lethal disease in animals. However, since RNA viruses are prone to high mutation rates, the possibility that these viruses will escape neutralization remains a potential concern. In the present study, we generated neutralization-escape mutants, using 6 different monoclonal antibodies, and studied the effect of these neutralization-escape mutations on in vitro and in vivo fitness. These data provide a mechanism for overcoming neutralization escape by use of cocktails of cross-neutralizing monoclonal antibodies that recognize residues within the glycoprotein that are important for virus replication and virulence.


Subject(s)
Antibodies, Monoclonal/immunology , Henipavirus Infections/virology , Henipavirus/genetics , Henipavirus/physiology , Animals , Antibodies, Neutralizing , Antibodies, Viral/immunology , Cricetinae , Female , Genetic Fitness , Henipavirus/immunology , Henipavirus Infections/immunology , Humans , Mesocricetus , Mice , Mutation , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...