Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Methods Mol Biol ; 2682: 281-299, 2023.
Article in English | MEDLINE | ID: mdl-37610589

ABSTRACT

Ferrets are commonly used as experimental models of infection for a variety of viruses due to their susceptibility to human respiratory viruses and the close resemblance of pathological outcomes found in human infections. Even though ferret-specific reagents are limited, the use of ferrets as a preclinical experimental model of infection has gained considerable interest since the publication of the ferret transcriptome and draft ferret genome. These advances have made it feasible to easily perform whole-genome gene expression analysis in the ferret infection model. Here, we describe methods for genome-wide gene expression analysis using RNA sequence (RNAseq) data obtained from the lung and brain tissues obtained from experimental infections of Hendra (HeV) and Nipah (NiV) viruses in ferrets. We provide detailed methods for RNAseq and representative data for host gene expression profiles of the lung tissues that show early activation of interferon pathways and later activation of inflammation-related pathways.


Subject(s)
Coronavirus , Henipavirus Infections , Animals , Humans , Ferrets , Henipavirus Infections/genetics , Gene Expression Profiling , Transcriptome
2.
Sci Adv ; 9(31): eadh4057, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37540755

ABSTRACT

Nipah virus (NiV) causes a highly lethal disease in humans who present with acute respiratory or neurological signs. No vaccines against NiV have been approved to date. Here, we report on the clinical impact of a novel NiV-derived nonspreading replicon particle lacking the fusion (F) protein gene (NiVΔF) as a vaccine in three small animal models of disease. A broad antibody response was detected that included immunoglobulin G (IgG) and IgA subtypes with demonstrable Fc-mediated effector function targeting multiple viral antigens. Single-dose intranasal vaccination up to 3 days before challenge prevented clinical signs and reduced virus levels in hamsters and immunocompromised mice; decreases were seen in tissues and mucosal secretions, critically decreasing potential for virus transmission. This virus replicon particle system provides a vital tool to the field and demonstrates utility as a highly efficacious and safe vaccine candidate that can be administered parenterally or mucosally to protect against lethal Nipah disease.


Subject(s)
Henipavirus Infections , Nipah Virus , Viral Vaccines , Cricetinae , Humans , Animals , Mice , Henipavirus Infections/prevention & control , Henipavirus Infections/genetics , Vaccination , Disease Models, Animal , Nipah Virus/genetics , Replicon
3.
Biosensors (Basel) ; 13(2)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36832018

ABSTRACT

Nipah virus (NiV) is a zoonotic RNA virus which infects humans and animals in Asian countries. Infection in humans occurs in different forms, from asymptomatic infection to fatal encephalitis, and death occurred in 40-70% of those infected in outbreaks that occurred between 1998 and 2018. Modern diagnostics is carried out by real-time PCR to identify pathogens or by ELISA to detect antibodies. Both technologies are labor-intensive and require the use of expensive stationary equipment. Thus, there is a need to develop alternative simple, fast and accurate test systems for virus detection. The aim of this study was to develop a highly specific and easily standardized system for the detection of Nipah virus RNA. In our work, we have developed a design for a Dz_NiV biosensor based on a split catalytic core of deoxyribozyme 10-23. It was shown that the assembly of active 10-23 DNAzymes occurred only in the presence of synthetic target Nipah virus RNA and that this was accompanied by stable fluorescence signals from the cleaved fluorescent substrates. This process was realized at 37 °C, pH 7.5, and in the presence of magnesium ions, with a 10 nM limit of detection achieved for the synthetic target RNA. Constructed via a simple and easily modifiable process, our biosensor may be used for the detection of other RNA viruses.


Subject(s)
DNA, Catalytic , Henipavirus Infections , Nipah Virus , Animals , Humans , Nipah Virus/genetics , Henipavirus Infections/epidemiology , Henipavirus Infections/genetics , RNA, Viral , Enzyme-Linked Immunosorbent Assay
4.
FEBS Lett ; 595(23): 2854-2871, 2021 12.
Article in English | MEDLINE | ID: mdl-34757622

ABSTRACT

SARS-CoV-2 has infected hundreds of millions of people with over four million dead, resulting in one of the worst global pandemics in recent history. Neurological symptoms associated with COVID-19 include anosmia, ageusia, headaches, confusion, delirium, and strokes. These may manifest due to viral entry into the central nervous system (CNS) through the blood-brain barrier (BBB) by means of ill-defined mechanisms. Here, we summarize the abilities of SARS-CoV-2 and other neurotropic RNA viruses, including Zika virus and Nipah virus, to cross the BBB into the CNS, highlighting the role of magnetic resonance imaging (MRI) in assessing presence and severity of brain structural changes in COVID-19 patients. We present new insight into key mutations in SARS-CoV-2 variants B.1.1.7 (P681H) and B.1.617.2 (P681R), which may impact on neuropilin 1 (NRP1) binding and CNS invasion. We postulate that SARS-CoV-2 may infect both peripheral cells capable of crossing the BBB and brain endothelial cells to traverse the BBB and spread into the brain. COVID-19 patients can be followed up with MRI modalities to better understand the long-term effects of COVID-19 on the brain.


Subject(s)
Blood-Brain Barrier , Henipavirus Infections , Nipah Virus , SARS-CoV-2 , Zika Virus Infection , Zika Virus , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/virology , COVID-19/epidemiology , COVID-19/genetics , COVID-19/metabolism , COVID-19/physiopathology , Henipavirus Infections/epidemiology , Henipavirus Infections/genetics , Henipavirus Infections/metabolism , Henipavirus Infections/physiopathology , Humans , Mutation , Nipah Virus/genetics , Nipah Virus/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Zika Virus/genetics , Zika Virus/metabolism , Zika Virus Infection/epidemiology , Zika Virus Infection/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/physiopathology
5.
Nat Commun ; 11(1): 3849, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737300

ABSTRACT

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.


Subject(s)
Antibodies, Viral/biosynthesis , Disease Resistance/genetics , Epstein-Barr Virus Infections/genetics , Henipavirus Infections/genetics , Host-Pathogen Interactions/genetics , Sarcoma, Kaposi/genetics , Adolescent , Adult , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Coinfection , Epstein-Barr Virus Infections/epidemiology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/immunology , Female , Gene Expression , Genome-Wide Association Study , HIV/genetics , HIV/immunology , HIV/pathogenicity , HLA-DQ alpha-Chains/genetics , HLA-DQ alpha-Chains/immunology , Henipavirus Infections/epidemiology , Henipavirus Infections/immunology , Henipavirus Infections/virology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions/immunology , Humans , Incidence , Male , Middle Aged , Rural Population , Sarcoma, Kaposi/epidemiology , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/virology , Uganda/epidemiology , Urban Population
6.
J Virol ; 94(14)2020 07 01.
Article in English | MEDLINE | ID: mdl-32321809

ABSTRACT

Nipah virus (NiV) and Hendra virus (HeV), members of the Henipavirus genus in the Paramyxoviridae family, are recently emerged, highly lethal zoonotic pathogens. The NiV and HeV nonsegmented, negative-sense RNA genomes encode nine proteins, including the W protein. Expressed from the P gene through mRNA editing, W shares a common N-terminus with P and V but has a unique C-terminus. Expressed alone, W modulates innate immune responses by several mechanisms, and elimination of W from NiV alters the course of infection in experimentally infected ferrets. However, the specific host interactions that allow W to modulate innate immunity are incompletely understood. This study demonstrates that the NiV and HeV W proteins interact with all seven isoforms of the 14-3-3 family, regulatory molecules that preferentially bind phosphorylated target proteins to regulate a wide range of cellular functions. The interaction is dependent on the penultimate amino acid residue in the W sequence, a conserved, phosphorylated serine. The cocrystal structure of the W C-terminal binding motif with 14-3-3 provides only the second structure of a complex containing a mode III interactor, which is defined as a 14-3-3 interaction with a phosphoserine/phosphothreonine at the C-termini of the target protein. Transcriptomic analysis of inducible cell lines infected with an RNA virus and expressing either wild-type W or W lacking 14-3-3 binding, identifies new functions for W. These include the regulation of cellular metabolic processes, extracellular matrix organization, and apoptosis.IMPORTANCE Nipah virus (NiV) and Hendra virus (HeV), members of the Henipavirus genus, are recently emerged, highly lethal zoonotic pathogens that cause yearly outbreaks. NiV and HeV each encode a W protein that has roles in regulating host signaling pathways, including antagonism of the innate immune response. However, the mechanisms used by W to regulate these host responses are not clear. Here, characterization of the interaction of NiV and HeV W with 14-3-3 identifies modulation of 14-3-3-regulated host signaling pathways not previously associated with W, suggesting new avenues of research. The cocrystal structure of the NiV W:14-3-3 complex, as only the second structure of a 14-3-3 mode III interactor, provides further insight into this less-well-understood 14-3-3 binding motif.


Subject(s)
14-3-3 Proteins/metabolism , Gene Expression Regulation , Hendra Virus/metabolism , Henipavirus Infections/metabolism , Nipah Virus/metabolism , Viral Proteins/metabolism , 14-3-3 Proteins/genetics , HEK293 Cells , Hendra Virus/genetics , Henipavirus Infections/genetics , Humans , Nipah Virus/genetics , Viral Proteins/genetics
7.
PLoS Pathog ; 16(3): e1008412, 2020 03.
Article in English | MEDLINE | ID: mdl-32226041

ABSTRACT

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.


Subject(s)
Antigens, Viral/immunology , Hendra Virus/immunology , Henipavirus Infections/immunology , Immunity, Cellular , Immunity, Innate , Lung/immunology , Models, Immunological , Animals , Antigens, Viral/genetics , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chiroptera , Ferrets , Hendra Virus/genetics , Henipavirus Infections/genetics , Henipavirus Infections/pathology , Interferons/genetics , Interferons/immunology , Lung/pathology , Lung/virology , Species Specificity
8.
Proc Natl Acad Sci U S A ; 116(41): 20707-20715, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548390

ABSTRACT

Cedar virus (CedV) is a bat-borne henipavirus related to Nipah virus (NiV) and Hendra virus (HeV), zoonotic agents of fatal human disease. CedV receptor-binding protein (G) shares only ∼30% sequence identity with those of NiV and HeV, although they can all use ephrin-B2 as an entry receptor. We demonstrate that CedV also enters cells through additional B- and A-class ephrins (ephrin-B1, ephrin-A2, and ephrin-A5) and report the crystal structure of the CedV G ectodomain alone and in complex with ephrin-B1 or ephrin-B2. The CedV G receptor-binding site is structurally distinct from other henipaviruses, underlying its capability to accommodate additional ephrin receptors. We also show that CedV can enter cells through mouse ephrin-A1 but not human ephrin-A1, which differ by 1 residue in the key contact region. This is evidence of species specific ephrin receptor usage by a henipavirus, and implicates additional ephrin receptors in potential zoonotic transmission.


Subject(s)
Ephrin-B1/metabolism , Ephrin-B2/metabolism , Ephrin-B3/metabolism , Henipavirus Infections/virology , Henipavirus/physiology , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Animals , Cell Fusion , Ephrin-B1/genetics , Ephrin-B2/genetics , Ephrin-B3/genetics , Henipavirus Infections/genetics , Henipavirus Infections/metabolism , Humans , Mice , Mutation , Protein Binding , Protein Conformation , Receptors, Virus/genetics , Species Specificity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
9.
PLoS Negl Trop Dis ; 12(11): e0006978, 2018 11.
Article in English | MEDLINE | ID: mdl-30462637

ABSTRACT

Nipah virus (NiV) infection can lead to severe respiratory or neurological disease in humans. Transmission of NiV has been shown to occur through contact with virus contaminated fomites or consumption of contaminated food. Previous results using the African green monkey (AGM) model of NiV infection identified aspects of infection that, while similar to humans, don't fully recapitulate disease. Previous studies also demonstrate near uniform lethality that is not consistent with human NiV infection. In these studies, aerosol exposure using an intermediate particle size (7µm) was used to mimic potential human exposure by facilitating virus deposition in the upper respiratory tract. Computed tomography evaluation found some animals developed pulmonary parenchymal disease including consolidations, ground-glass opacities, and reactive adenopathy. Despite the lack of neurological signs, magnetic resonance imaging identified distinct brain lesions in three animals, similar to those previously reported in NiV-infected patients. Immunological characterization of tissues collected at necropsy suggested a local pulmonary inflammatory response with increased levels of macrophages in the lung, but a limited neurologic response. These data provide the first clear evidence of neurological involvement in the AGM that recapitulates human disease. With the development of a disease model that is more representative of human disease, these data suggest that NiV infection in the AGM may be appropriate for evaluating therapeutic countermeasures directed at virus-induced neuropathogenesis.


Subject(s)
Aerosols/adverse effects , Henipavirus Infections/virology , Nervous System Diseases/virology , Nipah Virus/physiology , Aerosols/chemistry , Animals , Chlorocebus aethiops , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Henipavirus Infections/genetics , Henipavirus Infections/immunology , Humans , Lung/immunology , Lung/virology , Male , Nervous System Diseases/etiology , Nervous System Diseases/genetics , Nervous System Diseases/immunology , Nipah Virus/chemistry , Particle Size , Respiratory Tract Infections/etiology , Respiratory Tract Infections/genetics , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology
10.
Sci Rep ; 8(1): 15994, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30375468

ABSTRACT

Continued outbreaks of Henipaviruses in South Asia and Australia cause severe and lethal disease in both humans and animals. Together, with evidence of human to human transmission for Nipah virus and the lack of preventative or therapeutic measures, its threat to cause a widespread outbreak and its potential for weaponization has increased. In this study we demonstrate how overexpression of the Nipah virus nucleocapsid protein regulates viral polymerase activity and viral RNA production. By overexpressing the Nipah virus nucleocapsid protein in trans viral transcription was inhibited; however, an increase in viral genome synthesis was observed. Together, the bias of polymerase activity towards genome production led to the severe inhibition of viral progeny. We identified two domains within the nucleocapsid protein, which were each independently capable of binding the viral phosphoprotein. Evident by our data, we propose that the nucleocapsid protein's ability to interact with the phosphoprotein of the polymerase complex causes a change in polymerase activity and subsequent deficiency in viral replication. This study not only provides insights into the dynamics of Henipavirus RNA synthesis and replication, but also provides insight into potential targets for antiviral drug development.


Subject(s)
Henipavirus Infections/genetics , Nipah Virus/genetics , Nucleocapsid Proteins/genetics , Virus Replication/genetics , Henipavirus Infections/transmission , Henipavirus Infections/virology , Humans , Nipah Virus/pathogenicity , Phosphoproteins/genetics , Protein Binding/genetics , RNA, Viral/genetics , Viral Proteins/genetics , Virion/genetics , Virion/pathogenicity
11.
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
12.
Sci Rep ; 8(1): 358, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321677

ABSTRACT

Hendra virus (HeV) is a paramyxovirus that causes lethal disease in humans, for which no vaccine or antiviral agent is available. HeV V protein is central to pathogenesis through its ability to interact with cytoplasmic host proteins, playing key antiviral roles. Here we use immunoprecipitation, siRNA knockdown and confocal laser scanning microscopy to show that HeV V shuttles to and from the nucleus through specific host nuclear transporters. Spectroscopic and small angle X-ray scattering studies reveal HeV V undergoes a disorder-to-order transition upon binding to either importin α/ß1 or exportin-1/Ran-GTP, dependent on the V N-terminus. Importantly, we show that specific inhibitors of nuclear transport prevent interaction with host transporters, and reduce HeV infection. These findings emphasize the critical role of host-virus interactions in HeV infection, and potential use of compounds targeting nuclear transport, such as the FDA-approved agent ivermectin, as anti-HeV agents.


Subject(s)
Hendra Virus/physiology , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Host-Pathogen Interactions , Nucleocytoplasmic Transport Proteins/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Nucleus/metabolism , Drug Discovery , Gene Knockdown Techniques , Hendra Virus/drug effects , Henipavirus Infections/genetics , Humans , Karyopherins/chemistry , Karyopherins/genetics , Karyopherins/metabolism , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/metabolism , Exportin 1 Protein
13.
Curr Top Microbiol Immunol ; 419: 191-213, 2018.
Article in English | MEDLINE | ID: mdl-28674944

ABSTRACT

Hendra and Nipah viruses (family Paramyxoviridae, genus Henipavirus) are zoonotic RNA viruses that cause lethal disease in humans and are designated as Biosafety Level 4 (BSL4) agents. Moreover, henipaviruses belong to the same group of viruses that cause disease more commonly in humans such as measles, mumps and respiratory syncytial virus. Due to the relatively recent emergence of the henipaviruses and the practical constraints of performing functional genomics studies at high levels of containment, our understanding of the henipavirus infection cycle is incomplete. In this chapter we describe recent loss-of-function (i.e. RNAi) functional genomics screens that shed light on the henipavirus-host interface at a genome-wide level. Further to this, we cross-reference RNAi results with studies probing host proteins targeted by henipavirus proteins, such as nuclear proteins and immune modulators. These functional genomics studies join a growing body of evidence demonstrating that nuclear and nucleolar host proteins play a crucial role in henipavirus infection. Furthermore these studies will underpin future efforts to define the role of nucleolar host-virus interactions in infection and disease.


Subject(s)
Genomics , Hendra Virus/immunology , Henipavirus Infections/genetics , Henipavirus Infections/immunology , Host-Pathogen Interactions , MicroRNAs/metabolism , Nipah Virus/immunology , Nuclear Proteins/metabolism , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics
14.
Sci Rep ; 7(1): 7431, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785041

ABSTRACT

Hendra virus (HeV) is an emerging zoonotic pathogen harbored by Australian mainland flying foxes. HeV infection can cause lethal disease in humans and horses, and to date all cases of human HeV disease have resulted from contact with infected horses. Currently, diagnosis of acute HeV infections in horses relies on the productive phase of infection when virus shedding may occur. An assay that identifies infected horses during the preclinical phase of infection would reduce the risk of zoonotic viral transmission during management of HeV outbreaks. Having previously shown that the host microRNA (miR)-146a is upregulated in the blood of HeV-infected horses days prior to the detection of viremia, we have profiled miRNAs at the transcriptome-wide level to comprehensively assess differences between infected and uninfected horses. Next-generation sequencing and the miRDeep2 algorithm identified 742 mature miRNA transcripts corresponding to 593 miRNAs in whole blood of six horses (three HeV-infected, three uninfected). Thirty seven miRNAs were differentially expressed in infected horses, two of which were validated by qRT-PCR. This study describes a methodology for the transcriptome-wide profiling of miRNAs in whole blood and supports the notion that measuring host miRNA expression levels may aid infectious disease diagnosis in the future.


Subject(s)
Circulating MicroRNA/genetics , Gene Expression Profiling/veterinary , Henipavirus Infections/veterinary , Horse Diseases/diagnosis , Horses/genetics , Animals , Australia , Circulating MicroRNA/blood , Early Diagnosis , Gene Expression Regulation , Hendra Virus/pathogenicity , Henipavirus Infections/blood , Henipavirus Infections/diagnosis , Henipavirus Infections/genetics , High-Throughput Nucleotide Sequencing/veterinary , Horse Diseases/blood , Horse Diseases/genetics , Horses/blood , Sequence Analysis, RNA/veterinary
15.
PLoS Pathog ; 12(10): e1005974, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783670

ABSTRACT

Hendra and Nipah viruses (family Paramyxoviridae, genus Henipavirus) are bat-borne viruses that cause fatal disease in humans and a range of other mammalian species. Gaining a deeper understanding of host pathways exploited by henipaviruses for infection may identify targets for new anti-viral therapies. Here we have performed genome-wide high-throughput agonist and antagonist screens at biosafety level 4 to identify host-encoded microRNAs (miRNAs) impacting henipavirus infection in human cells. Members of the miR-181 and miR-17~93 families strongly promoted Hendra virus infection. miR-181 also promoted Nipah virus infection, but did not affect infection by paramyxoviruses from other genera, indicating specificity in the virus-host interaction. Infection promotion was primarily mediated via the ability of miR-181 to significantly enhance henipavirus-induced membrane fusion. Cell signalling receptors of ephrins, namely EphA5 and EphA7, were identified as novel negative regulators of henipavirus fusion. The expression of these receptors, as well as EphB4, were suppressed by miR-181 overexpression, suggesting that simultaneous inhibition of several Ephs by the miRNA contributes to enhanced infection and fusion. Immune-responsive miR-181 levels was also up-regulated in the biofluids of ferrets and horses infected with Hendra virus, suggesting that the host innate immune response may promote henipavirus spread and exacerbate disease severity. This study is the first genome-wide screen of miRNAs influencing infection by a clinically significant mononegavirus and nominates select miRNAs as targets for future anti-viral therapy development.


Subject(s)
Henipavirus Infections/genetics , MicroRNAs/genetics , Virus Internalization , Animals , Ferrets , Fluorescent Antibody Technique , Genome-Wide Association Study , Henipavirus , High-Throughput Nucleotide Sequencing , Horses , Humans , Real-Time Polymerase Chain Reaction
16.
PLoS Pathog ; 12(9): e1005880, 2016 09.
Article in English | MEDLINE | ID: mdl-27622505

ABSTRACT

For efficient replication, viruses have developed mechanisms to evade innate immune responses, including the antiviral type-I interferon (IFN-I) system. Nipah virus (NiV), a highly pathogenic member of the Paramyxoviridae family (genus Henipavirus), is known to encode for four P gene-derived viral proteins (P/C/W/V) with IFN-I antagonist functions. Here we report that NiV matrix protein (NiV-M), which is important for virus assembly and budding, can also inhibit IFN-I responses. IFN-I production requires activation of multiple signaling components including the IκB kinase epsilon (IKKε). We previously showed that the E3-ubiquitin ligase TRIM6 catalyzes the synthesis of unanchored K48-linked polyubiquitin chains, which are not covalently attached to any protein, and activate IKKε for induction of IFN-I mediated antiviral responses. Using co-immunoprecipitation assays and confocal microscopy we show here that the NiV-M protein interacts with TRIM6 and promotes TRIM6 degradation. Consequently, NiV-M expression results in reduced levels of unanchored K48-linked polyubiquitin chains associated with IKKε leading to impaired IKKε oligomerization, IKKε autophosphorylation and reduced IFN-mediated responses. This IFN antagonist function of NiV-M requires a conserved lysine residue (K258) in the bipartite nuclear localization signal that is found in divergent henipaviruses. Consistent with this, the matrix proteins of Ghana, Hendra and Cedar viruses were also able to inhibit IFNß induction. Live NiV infection, but not a recombinant NiV lacking the M protein, reduced the levels of endogenous TRIM6 protein expression. To our knowledge, matrix proteins of paramyxoviruses have never been reported to be involved in innate immune antagonism. We report here a novel mechanism of viral innate immune evasion by targeting TRIM6, IKKε and unanchored polyubiquitin chains. These findings expand the universe of viral IFN antagonism strategies and provide a new potential target for development of therapeutic interventions against NiV infections.


Subject(s)
Henipavirus Infections/immunology , I-kappa B Kinase/immunology , Immune Evasion , Interferon Type I/immunology , Nipah Virus/immunology , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Viral Proteins/immunology , A549 Cells , Animals , Chlorocebus aethiops , HeLa Cells , Henipavirus Infections/genetics , Humans , I-kappa B Kinase/genetics , Immunity, Innate , Interferon Type I/genetics , Nipah Virus/genetics , Polyubiquitin/genetics , Polyubiquitin/immunology , Protein Multimerization/genetics , Protein Multimerization/immunology , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology , Vero Cells , Viral Proteins/genetics
17.
J Gen Virol ; 97(4): 839-843, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26781134

ABSTRACT

Incorporation of reporter genes within virus genomes is an indispensable tool for interrogation of virus biology and pathogenesis. In previous work, we incorporated a fluorophore into a viral ORF by attaching it to the viral gene via a P2A ribosomal skipping sequence. This recombinant Nipah virus, however, was attenuated in vitro relative to WT virus. In this work, we determined that inefficient ribosomal skipping was a major contributing factor to this attenuation. Inserting a GSG linker before the P2A sequence resulted in essentially complete skipping, significantly improved growth in vitro, and WT lethality in vivo. To the best of our knowledge, this represents the first time a recombinant virus of Mononegavirales with integration of a reporter into a viral ORF has been compared with the WT virus in vivo. Incorporating the GSG linker for improved skipping efficiency whenever functionally important is a critical consideration for recombinant virus design.


Subject(s)
Genes, Reporter , Genetic Engineering/methods , Henipavirus Infections/genetics , Nipah Virus/genetics , Ribosomal Proteins/genetics , Amino Acid Sequence , Animals , Female , Gene Expression Regulation , Henipavirus Infections/mortality , Henipavirus Infections/pathology , Henipavirus Infections/virology , Human Umbilical Vein Endothelial Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mesocricetus , Molecular Sequence Data , Mutagenesis, Insertional , Nipah Virus/pathogenicity , Phalloidine/genetics , Phalloidine/metabolism , Ribosomal Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Survival Analysis , Transcription, Genetic , Virus Replication/genetics , Red Fluorescent Protein
18.
J Virol ; 89(15): 7550-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972557

ABSTRACT

UNLABELLED: Bats are important reservoirs for several viruses, many of which cause lethal infections in humans but have reduced pathogenicity in bats. As the innate immune response is critical for controlling viruses, the nature of this response in bats and how it may differ from that in other mammals are of great interest. Using next-generation transcriptome sequencing (mRNA-seq), we profiled the transcriptional response of Pteropus vampyrus bat kidney (PVK) cells to Newcastle disease virus (NDV), an avian paramyxovirus known to elicit a strong innate immune response in mammalian cells. The Pteropus genus is a known reservoir of Nipah virus (NiV) and Hendra virus (HeV). Analysis of the 200 to 300 regulated genes showed that genes for interferon (IFN) and antiviral pathways are highly upregulated in NDV-infected PVK cells, including genes for beta IFN, RIG-I, MDA5, ISG15, and IRF1. NDV-infected cells also upregulated several genes not previously characterized to be antiviral, such as RND1, SERTAD1, CHAC1, and MORC3. In fact, we show that MORC3 is induced by both IFN and NDV infection in PVK cells but is not induced by either stimulus in human A549 cells. In contrast to NDV infection, HeV and NiV infection of PVK cells failed to induce these innate immune response genes. Likewise, an attenuated response was observed in PVK cells infected with recombinant NDVs expressing the NiV IFN antagonist proteins V and W. This study provides the first global profile of a robust virus-induced innate immune response in bats and indicates that henipavirus IFN antagonist mechanisms are likely active in bat cells. IMPORTANCE: Bats are the reservoir host for many highly pathogenic human viruses, including henipaviruses, lyssaviruses, severe acute respiratory syndrome coronavirus, and filoviruses, and many other viruses have also been isolated from bats. Viral infections are reportedly asymptomatic or heavily attenuated in bat populations. Despite their ecological importance to viral maintenance, research into their immune system and mechanisms for viral control has only recently begun. Nipah virus and Hendra virus are two paramyxoviruses associated with high mortality rates in humans and whose reservoir is the Pteropus genus of bats. Greater knowledge of the innate immune response of P. vampyrus bats to viral infection may elucidate how bats serve as a reservoir for so many viruses.


Subject(s)
Chiroptera/immunology , Disease Reservoirs/virology , Gene Expression Profiling , Henipavirus Infections/immunology , Immunity, Innate , Interferons/immunology , Nipah Virus/immunology , Animals , Chiroptera/genetics , Chiroptera/virology , Hendra Virus/genetics , Hendra Virus/immunology , Hendra Virus/physiology , Henipavirus Infections/genetics , Henipavirus Infections/virology , Humans , Immune Evasion , Interferons/genetics , Newcastle disease virus/genetics , Newcastle disease virus/immunology , Newcastle disease virus/physiology , Nipah Virus/genetics , Nipah Virus/physiology
19.
Proc Natl Acad Sci U S A ; 112(17): E2156-65, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25825759

ABSTRACT

The discovery of African henipaviruses (HNVs) related to pathogenic Hendra virus (HeV) and Nipah virus (NiV) from Southeast Asia and Australia presents an open-ended health risk. Cell receptor use by emerging African HNVs at the stage of host-cell entry is a key parameter when considering the potential for spillover and infection of human populations. The attachment glycoprotein from a Ghanaian bat isolate (GhV-G) exhibits <30% sequence identity with Asiatic NiV-G/HeV-G. Here, through functional and structural analysis of GhV-G, we show how this African HNV targets the same human cell-surface receptor (ephrinB2) as the Asiatic HNVs. We first characterized this virus-receptor interaction crystallographically. Compared with extant HNV-G-ephrinB2 structures, there was significant structural variation in the six-bladed ß-propeller scaffold of the GhV-G receptor-binding domain, but not the Greek key fold of the bound ephrinB2. Analysis revealed a surprisingly conserved mode of ephrinB2 interaction that reflects an ongoing evolutionary constraint among geographically distal and phylogenetically divergent HNVs to maintain the functionality of ephrinB2 recognition during virus-host entry. Interestingly, unlike NiV-G/HeV-G, we could not detect binding of GhV-G to ephrinB3. Comparative structure-function analysis further revealed several distinguishing features of HNV-G function: a secondary ephrinB2 interaction site that contributes to more efficient ephrinB2-mediated entry in NiV-G relative to GhV-G and cognate residues at the very C terminus of GhV-G (absent in Asiatic HNV-Gs) that are vital for efficient receptor-induced fusion, but not receptor binding per se. These data provide molecular-level details for evaluating the likelihood of African HNVs to spill over into human populations.


Subject(s)
Ephrin-B2 , Henipavirus Infections/metabolism , Henipavirus , Viral Proteins , Virus Internalization , Ephrin-B2/chemistry , Ephrin-B2/genetics , Ephrin-B2/metabolism , Ephrin-B3/chemistry , Ephrin-B3/genetics , Ephrin-B3/metabolism , HEK293 Cells , Henipavirus/chemistry , Henipavirus/physiology , Henipavirus Infections/genetics , Humans , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Genome Biol ; 15(11): 532, 2014.
Article in English | MEDLINE | ID: mdl-25398248

ABSTRACT

BACKGROUND: Bats are a major reservoir of emerging infectious viruses. Many of these viruses are highly pathogenic to humans however bats remain asymptomatic. The mechanism by which bats control viral replication is unknown. Here we utilize an integrated approach of proteomics informed by transcriptomics to compare the response of immortalized bat and human cells following infection with the highly pathogenic bat-borne Hendra virus (HeV). RESULTS: The host response between the cell lines was significantly different at both the mRNA and protein levels. Human cells demonstrated minimal response eight hours post infection, followed by a global suppression of mRNA and protein abundance. Bat cells demonstrated a robust immune response eight hours post infection, which led to the up-regulation of apoptosis pathways, mediated through the tumor necrosis factor-related apoptosis inducing ligand (TRAIL). HeV sensitized bat cells to TRAIL-mediated apoptosis, by up-regulating death receptor transcripts. At 48 and 72 hours post infection, bat cells demonstrated a significant increase in apoptotic cell death. CONCLUSIONS: This is the first study to comprehensively compare the response of bat and human cells to a highly pathogenic zoonotic virus. An early induction of innate immune processes followed by apoptosis of virally infected bat cells highlights the possible involvement of programmed cell death in the host response. Our study shows for the first time a side-by-side high-throughput analysis of a dangerous zoonotic virus in cell lines derived from humans and the natural bat host. This enables a way to search for divergent mechanisms at a molecular level that may influence host pathogenesis.


Subject(s)
Hendra Virus/genetics , Henipavirus Infections/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Transcriptome/genetics , Animals , Apoptosis/genetics , Chiroptera/genetics , Chiroptera/virology , Hendra Virus/pathogenicity , Henipavirus Infections/transmission , Henipavirus Infections/virology , Host-Pathogen Interactions/genetics , Humans , Proteomics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...