Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurovirol ; 30(1): 39-51, 2024 02.
Article in English | MEDLINE | ID: mdl-38172412

ABSTRACT

Sarbecoviruses such as SARS and SARS-CoV-2 have been responsible for two major outbreaks in humans, the latter resulting in a global pandemic. While sarbecoviruses primarily cause an acute respiratory infection, they have been shown to infect the nervous system. However, mechanisms of sarbecovirus neuroinvasion and neuropathogenesis remain unclear. In this study, we examined the infectivity and trans-synaptic transmission potential of the sarbecoviruses SARS and SARS-CoV-2 in human stem cell-derived neural model systems. We demonstrated limited ability of sarbecoviruses to infect and replicate in human stem cell-derived neurons. Furthermore, we demonstrated an inability of sarbecoviruses to transmit between synaptically connected human stem cell-derived neurons. Finally, we determined an absence of SARS-CoV-2 infection in olfactory neurons in experimentally infected ferrets. Collectively, this study indicates that sarbecoviruses exhibit low potential to infect human stem cell-derived neurons, lack an ability to infect ferret olfactory neurons, and lack an inbuilt molecular mechanism to utilise retrograde axonal trafficking and trans-synaptic transmission to spread within the human nervous system.


Subject(s)
Axons , COVID-19 , Ferrets , SARS-CoV-2 , Severe acute respiratory syndrome-related coronavirus , Humans , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Animals , COVID-19/virology , COVID-19/transmission , Axons/virology , Ferrets/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Neurons/virology , Virus Replication , Chlorocebus aethiops , Neural Stem Cells/virology , Vero Cells
2.
J Virol ; 96(20): e0115222, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36173189

ABSTRACT

Bats are recognized as important reservoirs of viruses deadly to other mammals, including humans. These infections are typically nonpathogenic in bats, raising questions about host response differences that might exist between bats and other mammals. Tetherin is a restriction factor which inhibits the release of a diverse range of viruses from host cells, including retroviruses, coronaviruses, filoviruses, and paramyxoviruses, some of which are deadly to humans and transmitted by bats. Here, we characterize the tetherin genes from 27 bat species, revealing that they have evolved under strong selective pressure, and that fruit bats and vesper bats express unique structural variants of the tetherin protein. Tetherin was widely and variably expressed across fruit bat tissue types and upregulated in spleen tissue when stimulated with Toll-like receptor agonists. The expression of two computationally predicted splice isoforms of fruit bat tetherin was verified. We identified an additional third unique splice isoform which includes a C-terminal region that is not homologous to known mammalian tetherin variants but was functionally capable of restricting the release of filoviral virus-like particles. We also report that vesper bats possess and express at least five tetherin genes, including structural variants, more than any other mammal reported to date. These findings support the hypothesis of differential antiviral gene evolution in bats relative to other mammals. IMPORTANCE Bats are an important host of various viruses which are deadly to humans and other mammals but do not cause outward signs of illness in bats. Furthering our understanding of the unique features of the immune system of bats will shed light on how they tolerate viral infections, potentially informing novel antiviral strategies in humans and other animals. This study examines the antiviral protein tetherin, which prevents viral particles from escaping their host cell. Analysis of tetherin from 27 bat species reveals that it is under strong evolutionary pressure, and we show that multiple bat species have evolved to possess more tetherin genes than other mammals, some of which encode structurally unique tetherins capable of activity against different viral particles. These data suggest that bat tetherin plays a potentially broad and important role in the management of viral infections in bats.


Subject(s)
Chiroptera , Virus Diseases , Viruses , Humans , Animals , Bone Marrow Stromal Antigen 2/genetics , Antiviral Agents , Toll-Like Receptors
3.
PLoS Pathog ; 17(6): e1009636, 2021 06.
Article in English | MEDLINE | ID: mdl-34166464

ABSTRACT

Many viruses target signal transducers and activators of transcription (STAT) 1 and 2 to antagonise antiviral interferon signalling, but targeting of signalling by other STATs/cytokines, including STAT3/interleukin 6 that regulate processes important to Ebola virus (EBOV) haemorrhagic fever, is poorly defined. We report that EBOV potently inhibits STAT3 responses to interleukin-6 family cytokines, and that this is mediated by the interferon-antagonist VP24. Mechanistic analysis indicates that VP24 effects a unique strategy combining distinct karyopherin-dependent and karyopherin-independent mechanisms to antagonise STAT3-STAT1 heterodimers and STAT3 homodimers, respectively. This appears to reflect distinct mechanisms of nuclear trafficking of the STAT3 complexes, revealed for the first time by our analysis of VP24 function. These findings are consistent with major roles for global inhibition of STAT3 signalling in EBOV infection, and provide new insights into the molecular mechanisms of STAT3 nuclear trafficking, significant to pathogen-host interactions, cell physiology and pathologies such as cancer.


Subject(s)
Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/physiology , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , Ebolavirus , HEK293 Cells , Humans , Vero Cells
4.
PLoS Pathog ; 17(7): e1009759, 2021 07.
Article in English | MEDLINE | ID: mdl-34320031

ABSTRACT

The host response to SARS-CoV-2 infection provide insights into both viral pathogenesis and patient management. The host-encoded microRNA (miRNA) response to SARS-CoV-2 infection, however, remains poorly defined. Here we profiled circulating miRNAs from ten COVID-19 patients sampled longitudinally and ten age and gender matched healthy donors. We observed 55 miRNAs that were altered in COVID-19 patients during early-stage disease, with the inflammatory miR-31-5p the most strongly upregulated. Supervised machine learning analysis revealed that a three-miRNA signature (miR-423-5p, miR-23a-3p and miR-195-5p) independently classified COVID-19 cases with an accuracy of 99.9%. In a ferret COVID-19 model, the three-miRNA signature again detected SARS-CoV-2 infection with 99.7% accuracy, and distinguished SARS-CoV-2 infection from influenza A (H1N1) infection and healthy controls with 95% accuracy. Distinct miRNA profiles were also observed in COVID-19 patients requiring oxygenation. This study demonstrates that SARS-CoV-2 infection induces a robust host miRNA response that could improve COVID-19 detection and patient management.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , COVID-19/genetics , MicroRNAs/genetics , SARS-CoV-2 , Adult , Aged , Animals , COVID-19/blood , Case-Control Studies , Diagnosis, Differential , Disease Models, Animal , Female , Ferrets , Gene Expression , Host Microbial Interactions/genetics , Humans , Influenza A Virus, H1N1 Subtype , Longitudinal Studies , Male , MicroRNAs/blood , Middle Aged , Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae Infections/genetics , Pandemics , Supervised Machine Learning
5.
Proc Natl Acad Sci U S A ; 117(17): 9529-9536, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32284399

ABSTRACT

Bats are reservoirs of emerging viruses that are highly pathogenic to other mammals, including humans. Despite the diversity and abundance of bat viruses, to date they have not been shown to harbor exogenous retroviruses. Here we report the discovery and characterization of a group of koala retrovirus-related (KoRV-related) gammaretroviruses in Australian and Asian bats. These include the Hervey pteropid gammaretrovirus (HPG), identified in the scat of the Australian black flying fox (Pteropus alecto), which is the first reproduction-competent retrovirus found in bats. HPG is a close relative of KoRV and the gibbon ape leukemia virus (GALV), with virion morphology and Mn2+-dependent virion-associated reverse transcriptase activity typical of a gammaretrovirus. In vitro, HPG is capable of infecting bat and human cells, but not mouse cells, and displays a similar pattern of cell tropism as KoRV-A and GALV. Population studies reveal the presence of HPG and KoRV-related sequences in several locations across northeast Australia, as well as serologic evidence for HPG in multiple pteropid bat species, while phylogenetic analysis places these bat viruses as the basal group within the KoRV-related retroviruses. Taken together, these results reveal bats to be important reservoirs of exogenous KoRV-related gammaretroviruses.


Subject(s)
Chiroptera/virology , Gammaretrovirus/isolation & purification , Animals , Australia , Disease Reservoirs/veterinary , Disease Reservoirs/virology , Phascolarctidae/virology
6.
J Gen Virol ; 103(8)2022 08.
Article in English | MEDLINE | ID: mdl-35972225

ABSTRACT

Bats have been implicated as the reservoir hosts of filoviruses in Africa, with serological evidence of filoviruses in various bat species identified in other countries. Here, serum samples from 190 bats, comprising 12 different species, collected in Australia were evaluated for filovirus antibodies. An in-house indirect microsphere assay to detect antibodies that cross-react with Ebola virus (Zaire ebolavirus; EBOV) nucleoprotein (NP) followed by an immunofluorescence assay (IFA) were used to confirm immunoreactivity to EBOV and Reston virus (Reston ebolavirus; RESTV). We found 27 of 102 Yinpterochiroptera and 19 of 88 Yangochiroptera samples were positive to EBOV NP in the microsphere assay. Further testing of these NP positive samples by IFA revealed nine bat sera that showed binding to ebolavirus-infected cells. This is the first report of filovirus-reactive antibodies detected in Australian bat species and suggests that novel filoviruses may be circulating in Australian bats.


Subject(s)
Chiroptera , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Antibodies, Viral , Australia , Hemorrhagic Fever, Ebola/veterinary , Nucleoproteins
7.
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
8.
J Gen Virol ; 100(3): 403-413, 2019 03.
Article in English | MEDLINE | ID: mdl-30688635

ABSTRACT

Bats are the reservoir hosts for multiple viruses with zoonotic potential, including coronaviruses, paramyxoviruses and filoviruses. Urine collected from Australian pteropid bats was assessed for the presence of paramyxoviruses. One of the viruses isolated was Teviot virus (TevPV), a novel rubulavirus previously isolated from pteropid bat urine throughout the east coast of Australia. Here, we further characterize TevPV through analysis of whole-genome sequencing, growth kinetics, antigenic relatedness and the experimental infection of ferrets and mice. TevPV is phylogenetically and antigenically most closely related to Tioman virus (TioPV). Unlike many other rubulaviruses, cell receptor attachment by TevPV does not appear to be sialic acid-dependent, with the receptor for host cell entry being unknown. The infection of ferrets and mice suggested that TevPV has a low pathogenic potential in mammals. Infected ferrets seroconverted by 10 days post-infection without clinical signs of disease. Furthermore, infected ferrets did not shed virus in any respiratory secretions, suggesting a low risk of onward transmission of TevPV. No productive infection was observed in the mouse infection study.


Subject(s)
Chiroptera/virology , Paramyxoviridae Infections/veterinary , Paramyxovirinae/isolation & purification , Animals , Australia , Ferrets , Genome, Viral , Mice , Paramyxoviridae Infections/virology , Paramyxovirinae/genetics , Paramyxovirinae/pathogenicity , Paramyxovirinae/physiology , Phylogeny , Virulence
9.
J Virol ; 91(23)2017 12 01.
Article in English | MEDLINE | ID: mdl-28931675

ABSTRACT

Ebolavirus and Marburgvirus comprise two genera of negative-sense single-stranded RNA viruses that cause severe hemorrhagic fevers in humans. Despite considerable research efforts, the molecular events following Ebola virus (EBOV) infection are poorly understood. With the view of identifying host factors that underpin EBOV pathogenesis, we compared the transcriptomes of EBOV-infected human, pig, and bat kidney cells using a transcriptome sequencing (RNA-seq) approach. Despite a significant difference in viral transcription/replication between the cell lines, all cells responded to EBOV infection through a robust induction of extracellular growth factors. Furthermore, a significant upregulation of activator protein 1 (AP1) transcription factor complex members FOS and JUN was observed in permissive cell lines. Functional studies focusing on human cells showed that EBOV infection induces protein expression, phosphorylation, and nuclear accumulation of JUN and, to a lesser degree, FOS. Using a luciferase-based reporter, we show that EBOV infection induces AP1 transactivation activity within human cells at 48 and 72 h postinfection. Finally, we show that JUN knockdown decreases the expression of EBOV-induced host gene expression. Taken together, our study highlights the role of AP1 in promoting the host gene expression profile that defines EBOV pathogenesis.IMPORTANCE Many questions remain about the molecular events that underpin filovirus pathophysiology. The rational design of new intervention strategies, such as postexposure therapeutics, will be significantly enhanced through an in-depth understanding of these molecular events. We believe that new insights into the molecular pathogenesis of EBOV may be possible by examining the transcriptomic response of taxonomically diverse cell lines (derived from human, pig, and bat). We first identified the responsive pathways using an RNA-seq-based transcriptomics approach. Further functional and computational analysis focusing on human cells highlighted an important role for the AP1 transcription factor in mediating the transcriptional response to EBOV infection. Our study sheds new light on how host transcription factors respond to and promote the transcriptional landscape that follows viral infection.


Subject(s)
Gene Expression Profiling , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions , Transcription Factor AP-1/metabolism , Animals , Cell Line , Chiroptera , Ebolavirus/pathogenicity , Genes, fos , Genes, jun , High-Throughput Nucleotide Sequencing , Humans , Kidney/cytology , Kidney/virology , Phosphorylation , Swine , Transcription Factor AP-1/genetics , Viral Proteins , Virus Replication
10.
PLoS Pathog ; 12(3): e1005478, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27010548

ABSTRACT

Hendra and Nipah viruses (genus Henipavirus, family Paramyxoviridae) are highly pathogenic bat-borne viruses. The need for high biocontainment when studying henipaviruses has hindered the development of therapeutics and knowledge of the viral infection cycle. We have performed a genome-wide siRNA screen at biosafety level 4 that identified 585 human proteins required for henipavirus infection. The host protein with the largest impact was fibrillarin, a nucleolar methyltransferase that was also required by measles, mumps and respiratory syncytial viruses for infection. While not required for cell entry, henipavirus RNA and protein syntheses were greatly impaired in cells lacking fibrillarin, indicating a crucial role in the RNA replication phase of infection. During infection, the Hendra virus matrix protein co-localized with fibrillarin in cell nucleoli, and co-associated as a complex in pulldown studies, while its nuclear import was unaffected in fibrillarin-depleted cells. Mutagenesis studies showed that the methyltransferase activity of fibrillarin was required for henipavirus infection, suggesting that this enzyme could be targeted therapeutically to combat henipavirus infections.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Henipavirus Infections/virology , Nipah Virus/enzymology , Animals , Chlorocebus aethiops , Chromosomal Proteins, Non-Histone/genetics , HeLa Cells , Hendra Virus/metabolism , Humans , Mutation , Nipah Virus/genetics , Nipah Virus/pathogenicity , RNA, Small Interfering , Vero Cells , Viral Matrix Proteins/metabolism
11.
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
12.
Virol J ; 15(1): 75, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29685158

ABSTRACT

BACKGROUND: The 2014/2015 Ebolavirus outbreak resulted in more than 28,000 cases and 11,323 reported deaths, as of March 2016. Domestic transmission of the Guinea strain associated with the outbreak occurred mainly in six African countries, and international transmission was reported in four countries. Outbreak management was limited by the inability to rapidly diagnose infected cases. A further fifteen countries in Africa are predicted to be at risk of Ebolavirus outbreaks in the future as a consequence of climate change and urbanization. Early detection of cases and reduction of transmission rates is critical to prevent and manage future severe outbreaks. We designed a rapid assay for detection of Ebolavirus using recombinase polymerase amplification, a rapid isothermal amplification technology that can be combined with portable lateral flow detection technology. The developed rapid assay operates in 30 min and was comparable with real-time TaqMan™ PCR. METHODS: Designed, screened, selected and optimized oligonucleotides using the NP coding region from Ebola Zaire virus (Guinea strain). We determined the analytical sensitivity of our Ebola rapid molecular test by testing selected primers and probe with tenfold serial dilutions (1.34 × 1010- 1.34 × 101 copies/µL) of cloned NP gene from Mayinga strain of Zaire ebolavirus in pCAGGS vector, and serially diluted cultured Ebolavirus as established by real-time TaqMan™ PCR that was performed using ABI7500 in Fast Mode. We tested extracted and reverse transcribed RNA from cultured Zaire ebolavirus strains - Mayinga, Gueckedou C05, Gueckedou C07, Makona, Kissidougou and Kiwit. We determined the analytical specificity of our assay with related viruses: Marburg, Ebola Reston and Ebola Sudan. We further tested for Dengue virus 1-4, Plasmodium falciparum and West Nile Virus (Kunjin strain). RESULTS: The assay had a detection limit of 134 copies per µL of plasmid containing the NP gene of Ebolavirus Mayinga, and cultured Ebolavirus and was highly specific for the Zaire ebolavirus species, including the Guinea strain responsible for the 2014/2015 outbreak. The assay did not detect related viruses like Marburg, Reston, or Sudan viruses, and other pathogens likely to be isolated from clinical samples. CONCLUSIONS: Our assay could be suitable for implementation in district and primary health laboratories, as only a heating block and centrifuge is required for operation. The technique could provide a pathway for rapid screening of patients and animals for improved management of outbreaks.


Subject(s)
Ebolavirus/genetics , Hemorrhagic Fever, Ebola/diagnosis , Molecular Diagnostic Techniques/methods , Nucleic Acid Amplification Techniques , Recombinases , Cell Line , Hemorrhagic Fever, Ebola/virology , Humans , Nucleocapsid Proteins/genetics , RNA, Viral/analysis , RNA, Viral/genetics , Reverse Transcription , Sensitivity and Specificity
13.
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
14.
J Gen Virol ; 97(3): 581-592, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26703878

ABSTRACT

IFN-antagonist function is a major determinant of pathogenicity and cross-species infection by viruses, but remains poorly defined for many potentially zoonotic viruses resident in animal species. The paramyxovirus family contains several zoonotic viruses, including highly pathogenic viruses such as Nipah virus and Hendra virus, and an increasing number of largely uncharacterized animal viruses. Here, we report the characterization of IFN antagonism by the rodent viruses J virus (JPV) and Beilong virus (BeiPV) of the proposed genus Jeilongvirus of the paramyxoviruses. Infection of cells by JPV and BeiPV was found to inhibit IFN-activated nuclear translocation of signal transducer and activator of transcription 1 (STAT1). However, in contrast to most other paramyxoviruses, the JPV and BeiPV V proteins did not interact with or inhibit signalling by STAT1 or STAT2, suggesting that JPV/BeiPV use an atypical V protein-independent strategy to target STATs, consistent with their inclusion in a separate genus. Nevertheless, the V proteins of both viruses interacted with melanoma differentiation-associated protein 5 (MDA5) and robustly inhibited MDA5-dependent activation of the IFN-ß promoter. This supports a growing body of evidence that MDA5 is a universal target of paramyxovirus V proteins, such that the V-MDA5 interaction represents a potential target for broad-spectrum antiviral approaches.


Subject(s)
Immune Evasion , Paramyxoviridae Infections/immunology , Paramyxovirinae/immunology , Viral Proteins/immunology , Animals , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , HEK293 Cells , Humans , Interferon-Induced Helicase, IFIH1 , Interferon-alpha/genetics , Interferon-alpha/immunology , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/virology , Paramyxovirinae/classification , Paramyxovirinae/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/immunology , Signal Transduction , Viral Proteins/genetics
16.
Emerg Infect Dis ; 21(2): 328-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25626011

ABSTRACT

During 2014, henipavirus infection caused severe illness among humans and horses in southern Philippines; fatality rates among humans were high. Horse-to-human and human-to-human transmission occurred. The most likely source of horse infection was fruit bats. Ongoing surveillance is needed for rapid diagnosis, risk factor investigation, control measure implementation, and further virus characterization.


Subject(s)
Disease Outbreaks , Henipavirus Infections/epidemiology , Henipavirus/classification , Adolescent , Adult , Animal Diseases/epidemiology , Animal Diseases/virology , Animals , Base Sequence , Child , Child, Preschool , Female , Henipavirus/genetics , Henipavirus Infections/diagnosis , Henipavirus Infections/history , History, 21st Century , Humans , Male , Middle Aged , Molecular Sequence Data , Molecular Typing , Philippines/epidemiology , Phylogeny , Population Surveillance , Sequence Alignment , Serotyping , Viral Proteins/chemistry , Viral Proteins/genetics , Young Adult
17.
Proc Biol Sci ; 282(1798): 20142124, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25392474

ABSTRACT

Viruses that originate in bats may be the most notorious emerging zoonoses that spill over from wildlife into domestic animals and humans. Understanding how these infections filter through ecological systems to cause disease in humans is of profound importance to public health. Transmission of viruses from bats to humans requires a hierarchy of enabling conditions that connect the distribution of reservoir hosts, viral infection within these hosts, and exposure and susceptibility of recipient hosts. For many emerging bat viruses, spillover also requires viral shedding from bats, and survival of the virus in the environment. Focusing on Hendra virus, but also addressing Nipah virus, Ebola virus, Marburg virus and coronaviruses, we delineate this cross-species spillover dynamic from the within-host processes that drive virus excretion to land-use changes that increase interaction among species. We describe how land-use changes may affect co-occurrence and contact between bats and recipient hosts. Two hypotheses may explain temporal and spatial pulses of virus shedding in bat populations: episodic shedding from persistently infected bats or transient epidemics that occur as virus is transmitted among bat populations. Management of livestock also may affect the probability of exposure and disease. Interventions to decrease the probability of virus spillover can be implemented at multiple levels from targeting the reservoir host to managing recipient host exposure and susceptibility.


Subject(s)
Chiroptera/virology , Models, Biological , RNA Virus Infections/transmission , RNA Viruses/physiology , Zoonoses/transmission , Animals , Humans , Queensland , RNA Virus Infections/virology , RNA Viruses/isolation & purification , Zoonoses/virology
18.
BMC Genomics ; 15: 682, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25128405

ABSTRACT

BACKGROUND: Bats are a major source of new and emerging viral diseases. Despite the fact that bats carry and shed highly pathogenic viruses including Ebola, Nipah and SARS, they rarely display clinical symptoms of infection. Host factors influencing viral replication are poorly understood in bats and are likely to include both pre- and post-transcriptional regulatory mechanisms. MicroRNAs are a major mechanism of post-transcriptional gene regulation, however very little is known about them in bats. RESULTS: This study describes 399 microRNAs identified by deep sequencing of small RNA isolated from tissues of the Black flying fox, Pteropus alecto, a confirmed natural reservoir of the human pathogens Hendra virus and Australian bat lyssavirus. Of the microRNAs identified, more than 100 are unique amongst vertebrates, including a subset containing mutations in critical seed regions. Clusters of rapidly-evolving microRNAs were identified, as well as microRNAs predicted to target genes involved in antiviral immunity, the DNA damage response, apoptosis and autophagy. Closer inspection of the predicted targets for several highly supported novel miRNA candidates suggests putative roles in host-virus interaction. CONCLUSIONS: MicroRNAs are likely to play major roles in regulating virus-host interaction in bats, via dampening of inflammatory responses (limiting the effects of immunopathology), and directly limiting the extent of viral replication, either through restricting the availability of essential factors or by controlling apoptosis. Characterisation of the bat microRNA repertoire is an essential step towards understanding transcriptional regulation during viral infection, and will assist in the identification of mechanisms that enable bats to act as natural virus reservoirs. This in turn will facilitate the development of antiviral strategies for use in humans and other species.


Subject(s)
Chiroptera/genetics , MicroRNAs/genetics , Animals , Base Sequence , Binding Sites , Gene Ontology , High-Throughput Nucleotide Sequencing , Introns , Inverted Repeat Sequences , Male , Molecular Sequence Annotation , Molecular Sequence Data , Multigene Family , RNA Interference , Sequence Analysis, RNA , Sequence Homology, Nucleic Acid
19.
Emerg Infect Dis ; 20(3): 372-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24572697

ABSTRACT

In recent years, the emergence of several highly pathogenic zoonotic diseases in humans has led to a renewed emphasis on the interconnectedness of human, animal, and environmental health, otherwise known as One Health. For example, Hendra virus (HeV), a zoonotic paramyxovirus, was discovered in 1994, and since then, infections have occurred in 7 humans, each of whom had a strong epidemiologic link to similarly affected horses. As a consequence of these outbreaks, eradication of bat populations was discussed, despite their crucial environmental roles in pollination and reduction of the insect population. We describe the development and evaluation of a vaccine for horses with the potential for breaking the chain of HeV transmission from bats to horses to humans, thereby protecting horse, human, and environmental health. The HeV vaccine for horses is a key example of a One Health approach to the control of human disease.


Subject(s)
Environmental Health , Hendra Virus/immunology , Henipavirus Infections/prevention & control , Horse Diseases/prevention & control , Viral Vaccines/immunology , Zoonoses/prevention & control , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Ferrets , Guinea Pigs , Hendra Virus/genetics , Horse Diseases/pathology , Horse Diseases/virology , Horses , Humans , Immunization , Neutralization Tests , Zoonoses/pathology , Zoonoses/virology
20.
J Virol ; 87(7): 3782-91, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345523

ABSTRACT

Hendra virus is a highly pathogenic zoonotic paramyxovirus in the genus Henipavirus. Thirty-nine outbreaks of Hendra virus have been reported since its initial identification in Queensland, Australia, resulting in seven human infections and four fatalities. Little is known about cellular host factors impacting Hendra virus replication. In this work, we demonstrate that Hendra virus makes use of a microRNA (miRNA) designated miR-146a, an NF-κB-responsive miRNA upregulated by several innate immune ligands, to favor its replication. miR-146a is elevated in the blood of ferrets and horses infected with Hendra virus and is upregulated by Hendra virus in human cells in vitro. Blocking miR-146a reduces Hendra virus replication in vitro, suggesting a role for this miRNA in Hendra virus replication. In silico analysis of miR-146a targets identified ring finger protein (RNF)11, a member of the A20 ubiquitin editing complex that negatively regulates NF-κB activity, as a novel component of Hendra virus replication. RNA interference-mediated silencing of RNF11 promotes Hendra virus replication in vitro, suggesting that increased NF-κB activity aids Hendra virus replication. Furthermore, overexpression of the IκB superrepressor inhibits Hendra virus replication. These studies are the first to demonstrate a host miRNA response to Hendra virus infection and suggest an important role for host miRNAs in Hendra virus disease.


Subject(s)
Hendra Virus/physiology , MicroRNAs/metabolism , Virus Replication/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , DNA-Binding Proteins , HeLa Cells , Humans , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , RNA Interference , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL