Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
J Med Virol ; 96(7): e29788, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982767

ABSTRACT

Molecular surveillance is vital for monitoring arboviruses, often employing genus-specific quantitative reverse-transcription polymerase chain reaction (RT-qPCR). Despite this, an overlooked chikungunya fever outbreak occurred in Yunnan province, China, in 2019 and false negatives are commonly encountered during alphaviruses screening practice, highlighting the need for improved detection methods. In this study, we developed an improved alphaviruses-specific RT-qPCR capable of detecting chikungunya virus, eastern equine encephalitis virus, western equine encephalitis virus, Venezuelan equine encephalitis virus, Sindbis virus, Mayaro virus, and Ross River virus with high sensitivity and specificity. The assay identified three chikungunya virus-positive cases out of 188 sera retrospectively. Later genetic characterization suggested that imported cases from neighboring countries may be responsible for the neglected chikungunya fever outbreak of 2019 in Yunnan. Our findings underscore the value of improved alphaviruses-specific RT-qPCR in bolstering alphaviruses surveillance and informing preventive strategies.


Subject(s)
Alphavirus Infections , Alphavirus , Chikungunya virus , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Humans , Alphavirus/genetics , Alphavirus/isolation & purification , Alphavirus Infections/diagnosis , Alphavirus Infections/virology , Alphavirus Infections/prevention & control , Alphavirus Infections/epidemiology , China/epidemiology , Real-Time Polymerase Chain Reaction/methods , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Retrospective Studies , Chikungunya Fever/diagnosis , Chikungunya Fever/prevention & control , Chikungunya Fever/virology , Chikungunya Fever/epidemiology , Encephalitis Virus, Eastern Equine/genetics , Disease Outbreaks/prevention & control , Sindbis Virus/genetics , Encephalitis Virus, Western Equine/genetics , Ross River virus/genetics , Ross River virus/isolation & purification , Encephalitis Virus, Venezuelan Equine/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , RNA, Viral/genetics
2.
Parasit Vectors ; 16(1): 186, 2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37280650

ABSTRACT

BACKGROUND: Ross River virus (RRV) is Australia's most common and widespread mosquito-transmitted arbovirus and is of significant public health concern. With increasing anthropogenic impacts on wildlife and mosquito populations, it is important that we understand how RRV circulates in its endemic hotspots to determine where public health efforts should be directed. Current surveillance methods are effective in locating the virus but do not provide data on the circulation of the virus and its strains within the environment. This study examined the ability to identify single nucleotide polymorphisms (SNPs) within the variable E2/E3 region by generating full-length haplotypes from a range of mosquito trap-derived samples. METHODS: A novel tiled primer amplification workflow for amplifying RRV was developed with analysis using Oxford Nanopore Technology's MinION and a custom ARTIC/InterARTIC bioinformatic protocol. By creating a range of amplicons across the whole genome, fine-scale SNP analysis was enabled by specifically targeting the variable region that was amplified as a single fragment and established haplotypes that informed spatial-temporal variation of RRV in the study site in Victoria. RESULTS: A bioinformatic and laboratory pipeline was successfully designed and implemented on mosquito whole trap homogenates. Resulting data showed that genotyping could be conducted in real time and that whole trap consensus of the viruses (with major SNPs) could be determined in a timely manner. Minor variants were successfully detected from the variable E2/E3 region of RRV, which allowed haplotype determination within complex mosquito homogenate samples. CONCLUSIONS: The novel bioinformatic and wet laboratory methods developed here will enable fast detection and characterisation of RRV isolates. The concepts presented in this body of work are transferable to other viruses that exist as quasispecies in samples. The ability to detect minor SNPs, and thus haplotype strains, is critically important for understanding the epidemiology of viruses their natural environment.


Subject(s)
Alphavirus Infections , Culicidae , Nanopore Sequencing , Animals , Humans , Ross River virus/genetics , Genomics
3.
mBio ; 14(2): e0058823, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37036079

ABSTRACT

Arthritogenic alphaviruses such as Ross River virus (RRV) and Chikungunya virus (CHIKV) are responsible for large-scale epidemics that cause debilitating acute and chronic musculoskeletal diseases. MXRA8 was recently discovered as an entry receptor for multiple alphaviruses including CHIKV, RRV, Mayaro virus (MAYV), and O'nyong-nyong virus (ONNV). However, the role of MXRA8 in the development of alphavirus-induced musculoskeletal inflammation has not yet been fully studied. Here, we attempt to fully characterize the contribution of MXRA8 to RRV disease in an established mouse model. MXRA8 knockout (MXRA8-/-) mice generated on a C57BL/6J background, showed abrogated disease signs and reduced viral replication, which correlated with lower viral load, diminished proinflammatory cytokines, and limited cell infiltrates in inflamed tissues. Immunomodulation genes were upregulated to higher levels in RRV-infected wild-type (WT) mice than in MXRA8-/- mice. Intriguingly, Cdkn1a and Ifi44 genes in blood and CD127/IL7RA, CD45, BatF3, IFNGR, Ly6G/Ly6C, CD40, CD127, F4/80, and MHC-II genes in quadriceps were found to be upregulated in RRV-infected MXRA8-/- mice compared to WT mice. Our results showed an essential role of MXRA8 in the immune response of mice infected with RRV and, more importantly, demonstrated novel changes in immunomodulation genes, which shed light on the immunopathogenesis of alphavirus-induced disease. IMPORTANCE Previous studies have shown the importance of the cell surface protein MXRA8 as an entry receptor for several different prominent alphaviruses such as CHIKV, RRV, MAYV, and ONNV. In particular, the role of MXRA8 in the tissue tropism, viral pathogenesis, and immune response of a CHIKV mouse model have already been briefly characterized. However, the role of MXRA8 warrants further characterization in RRV disease background, since there are noticeable differences in the disease profile between CHIKV and RRV. For example, patients infected with CHIKV are usually affected by sudden onset of severe arthritis and fever, whereas RRV-infected patients generally only have minor joint pain and mild fever. Here, we characterized the role of MXRA8 in RRV disease and assessed several key mechanisms of MXRA8 that may contribute to the disease progression.


Subject(s)
Alphavirus Infections , Arthritis , Chikungunya virus , Animals , Mice , Ross River virus/genetics , Mice, Inbred C57BL , Chikungunya virus/genetics , Immunoglobulins , Membrane Proteins/metabolism
4.
Front Cell Infect Microbiol ; 12: 849662, 2022.
Article in English | MEDLINE | ID: mdl-35223559

ABSTRACT

We collected 5,500 mosquitoes belonging to six species in three locations in China. Their viromes were tested using metagenomic sequencing and bioinformatic analysis. The affluent viral sequences that were detected and annotated belong to 22 viral taxonomic families. Then, PCR was performed to confirm the results, followed by phylogenetic analysis. Herein, part of mosquito virome was identified, including chikungunya virus (CHIKV), Getah virus (GETV), and Ross river virus (RRV). After metagenomic analysis, seven CHIKV sequences were verified by PCR amplification, among which CHIKV-China/YN2018-1 had the highest homology with the CHIKV isolated in Senegal, 1983, with a nucleotide (nt) identity of at least 81%, belonging to genotype West Africa viral genes. Five GETV sequences were identified, which had a high homology with the GETV sequences isolated from Equus caballus in Japan, 1978, with a (nt) identity of at least 97%. The newly isolated virus CHIKV-China/YN2018-1 became more infectious after passage of the BHK-21 cell line to the Vero cell line. The newly identified RRV gene had the highest homology with the 2006 RRV isolate from Australia, with a (nt) identity of at least 94%. In addition, numerous known and unknown viruses have also been detected in mosquitoes from Yunnan province, China, and propagation tests will be carried out.


Subject(s)
Chikungunya Fever , Culicidae , Viruses , Animals , China , Horses , Humans , Phylogeny , Ross River virus/genetics , Togaviridae , Virome , Viruses/genetics
5.
PLoS Negl Trop Dis ; 16(2): e0009848, 2022 02.
Article in English | MEDLINE | ID: mdl-35143495

ABSTRACT

Across the Pacific, and including in the Solomon Islands, outbreaks of arboviruses such as dengue, chikungunya, and Zika are increasing in frequency, scale and impact. Outbreaks of mosquito-borne disease have the potential to overwhelm the health systems of small island nations. This study mapped the seroprevalence of dengue, Zika, chikungunya and Ross River viruses in 5 study sites in the Solomon Islands. Serum samples from 1,021 participants were analysed by ELISA. Overall, 56% of participants were flavivirus-seropositive for dengue (28%), Zika (1%) or both flaviviruses (27%); and 53% of participants were alphavirus-seropositive for chikungunya (3%), Ross River virus (31%) or both alphaviruses (18%). Seroprevalence for both flaviviruses and alphaviruses varied by village and age of the participant. The most prevalent arboviruses in the Solomon Islands were dengue and Ross River virus. The high seroprevalence of dengue suggests that herd immunity may be a driver of dengue outbreak dynamics in the Solomon Islands. Despite being undetected prior to this survey, serology results suggest that Ross River virus transmission is endemic. There is a real need to increase the diagnostic capacities for each of the arboviruses to support effective case management and to provide timely information to inform vector control efforts and other outbreak mitigation interventions.


Subject(s)
Alphavirus Infections/blood , Chikungunya Fever/blood , Chikungunya virus/immunology , Dengue Virus/immunology , Dengue/blood , Ross River virus/immunology , Zika Virus Infection/blood , Zika Virus/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Alphavirus Infections/epidemiology , Alphavirus Infections/virology , Antibodies, Viral/blood , Chikungunya Fever/epidemiology , Chikungunya Fever/virology , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Child , Child, Preschool , Dengue/epidemiology , Dengue/virology , Dengue Virus/genetics , Dengue Virus/isolation & purification , Female , Humans , Male , Melanesia/epidemiology , Middle Aged , Ross River virus/genetics , Ross River virus/isolation & purification , Seroepidemiologic Studies , Young Adult , Zika Virus/genetics , Zika Virus/isolation & purification , Zika Virus Infection/epidemiology , Zika Virus Infection/virology
6.
mBio ; 13(1): e0336321, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35089088

ABSTRACT

Ross River virus (RRV) is the major mosquito-borne virus in the South Pacific region. RRV infections are characterized by arthritic symptoms, which can last from several weeks to months. Type I interferon (IFN), the primary antiviral innate immune response, is able to modulate adaptive immune responses. The relationship between the protective role of type I IFN and the induction of signaling proteins that drive RRV disease pathogenesis remains poorly understood. In the present study, the role of TIR-domain-containing adapter-inducing interferon-ß (TRIF), an essential signaling adaptor protein downstream of Toll-like receptor (TLR) 3, a key single-stranded RNA (ssRNA)-sensing receptor, was investigated. We found that TRIF-/- mice were highly susceptible to RRV infection, with severe disease, high viremia, and a low type I IFN response early during disease development, which suggests the TLR3-TRIF axis may engage early in response to RRV infection. The number and the activation level of CD4+ T cells, CD8+ T cells, and NK cells were reduced in TRIF-/- mice compared to those in infected wild-type (WT) mice. In addition, the number of germinal center B cells was lower in TRIF-/- mice than WT mice following RRV infection, with lower titers of IgG antibodies detected in infected TRIF-/- mice compared to WT. Interestingly, the requirement for TRIF to promote immunoglobulin class switch recombination was at the level of the local immune microenvironment rather than B cells themselves. The slower resolution of RRV disease in TRIF-/- mice was associated with persistence of the RRV genome in muscle tissue and a continuing IFN response. IMPORTANCE RRV has been prevalent in the South Pacific region for decades and causes substantial economic and social costs. Though RRV is geographically restricted, a number of other alphaviruses have spread globally due to expansion of the mosquito vectors and increased international travel. Since over 30 species of mosquitoes have been implicated as potent vectors for RRV dissemination, RRV has the potential to further expand its distribution. In the pathogenesis of RRV disease, it is still not clear how innate immune responses synergize with adaptive immune responses. Type I IFN is crucial for bridging innate to adaptive immune responses to viral invasion. Hence, key signaling proteins in type I IFN induction pathways, which are important for type I IFN modulation, may also play critical roles in viral pathogenesis. This study provides insight into the role of TRIF in RRV disease development.


Subject(s)
Alphavirus Infections , Interferon Type I , Mice , Animals , Antiviral Agents , Ross River virus/genetics , CD8-Positive T-Lymphocytes/metabolism , Mosquito Vectors , Adaptor Proteins, Vesicular Transport/metabolism , Interferon-beta , Mice, Knockout
7.
Viruses ; 15(1)2022 12 31.
Article in English | MEDLINE | ID: mdl-36680176

ABSTRACT

Old world alphaviruses, such as Ross River virus (RRV), cause debilitating arthralgia during acute and chronic stages of the disease. RRV-induced cartilage degradation has been implicated as a cause of joint pain felt by RRV patients. Chondrocytes are a major cell type of cartilage and are involved in the production and maintenance of the cartilage matrix. It is thought that these cells may play a vital role in RRV disease pathogenesis. In this study, we used RNA-sequencing (RNA-Seq) to examine the transcriptomes of RRV-infected and bystander chondrocytes in the same environment. RRV containing green fluorescent protein (GFP) allowed for the separation of RRV-infected (GFP+) and bystander uninfected cells (GFP-). We found that whereas GFP+ and GFP- populations commonly presented similar gene expression profiles during infection, there were also unique signatures. For example, RIMS2 and FOXJ1 were unique to GFP+ cells, whilst Aim2 and CCL8 were only found in bystander chondrocytes. This indicates that careful selection of potential therapeutic targets is important to minimise adverse effects to the neighbouring uninfected cell populations. Our study serves as a resource to provide more information about the pathways and responses elicited by RRV in cells which are both infected and stimulated because of neighbouring infected cells.


Subject(s)
Alphavirus Infections , Alphavirus , Humans , Chondrocytes/metabolism , Alphavirus/genetics , Ross River virus/genetics , Ross River virus/metabolism
8.
J Microbiol ; 59(11): 1044-1055, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34570337

ABSTRACT

Getah virus (GETV), which was first isolated in Malaysia in 1955, and Sagiyama virus (SAGV), isolated in Japan in 1956, are members of the genus Alphavirus in the family Togaviridae. It is a consensus view that SAGV is a variant of GETV. In the present study, we determined the complete sequences of the prototype GETV MM2021 and SAGV M6-Mag132 genomic RNA extracted from plaque-purified viruses. The MM2021 genome was 11,692 nucleotides (nt) in length in the absence of 3' poly(A) tail, and the length of M6-Mag132 genome was 11,698 nt. Through sequence alignment of MM2021 and M6-Mag132, we located all the amino acid differences between these two strains, which were scattered in all the encoded proteins. Subsequently, we validated the close evolutionary relationship between GETV and SAGV by constructing phylogenetic trees based on either complete genomes or structural genomes. We eventually analyzed the growth kinetics of GETV and SAGV as well as other representative alphaviruses in various mammalian and insect cell lines. It was shown that human-oriented cell lines such as HEK-293T and Hela cells were relatively resistant to GETV and SAGV infection due to absence of proviral factors or species-specific barrier. On the other hand, both GETV and SAGV replicated efficiently in non-human cell lines. Our results provide essential genetic information for future epidemiological surveillance on Alphaviruses and lay the foundation for developing effective interventions against GETV and SAGV.


Subject(s)
Alphavirus/genetics , Genome, Viral , Host Specificity , Ross River virus/genetics , Alphavirus/classification , Alphavirus/isolation & purification , Alphavirus/physiology , Animals , Cell Line , Humans , Phylogeny , RNA, Viral/genetics , Ross River virus/classification , Ross River virus/isolation & purification , Ross River virus/physiology , Sequence Analysis, DNA
9.
Viruses ; 13(8)2021 07 31.
Article in English | MEDLINE | ID: mdl-34452382

ABSTRACT

Alphaviruses are small enveloped viruses with positive-sense RNA genomes. During infection, the alphavirus capsid protein (Cp) selectively packages and assembles with the viral genomic RNA to form the nucleocapsid core, a process critical to the production of infectious virus. Prior studies of the alphavirus Semliki Forest virus (SFV) showed that packaging and assembly are promoted by Cp binding to multiple high affinity sites on the genomic RNA. Here, we developed an in vitro Cp binding assay based on fluorescently labeled RNA oligos. We used this assay to explore the RNA sequence and structure requirements for Cp binding to site #1, the top binding site identified on the genomic RNA during all stages of virus assembly. Our results identify a stem-loop structure that promotes specific binding of the SFV Cp to site #1 RNA. This structure is also recognized by the Cps of the related alphaviruses chikungunya virus and Ross River virus.


Subject(s)
Alphavirus/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Genome, Viral/physiology , Inverted Repeat Sequences/genetics , RNA, Viral/metabolism , Alphavirus/metabolism , Binding Sites , Capsid/metabolism , Cell Line , Chikungunya virus/genetics , Chikungunya virus/metabolism , Genome, Viral/genetics , Inverted Repeat Sequences/physiology , Protein Binding , RNA, Viral/genetics , RNA-Binding Motifs , Ross River virus/genetics , Ross River virus/metabolism , Semliki forest virus/genetics , Semliki forest virus/metabolism , Virus Assembly
10.
Viruses ; 13(3)2021 03 15.
Article in English | MEDLINE | ID: mdl-33804215

ABSTRACT

Ross River virus (RRV) is the most medically significant mosquito-borne virus of Australia, in terms of human morbidity. RRV cases, characterised by febrile illness and potentially persistent arthralgia, have been reported from all Australian states and territories. RRV was the cause of a large-scale epidemic of multiple Pacific Island countries and territories (PICTs) from 1979 to 1980, involving at least 50,000 cases. Historical evidence of RRV seropositivity beyond Australia, in populations of Papua New Guinea (PNG), Indonesia and the Solomon Islands, has been documented. We describe the genomic characterisation and timescale analysis of the first isolate of RRV to be sampled from PNG to date. Our analysis indicates that RRV has evolved locally within PNG, independent of Australian lineages, over an approximate 40 year period. The mean time to most recent common ancestor (tMRCA) of the unique PNG clade coincides with the initiation of the PICTs epidemic in mid-1979. This may indicate that an ancestral variant of the PNG clade was seeded into the region during the epidemic, a period of high RRV transmission. Further epidemiological and molecular-based surveillance is required in PNG to better understand the molecular epidemiology of RRV in the general Australasian region.


Subject(s)
Culicidae/virology , Evolution, Molecular , Genome, Viral , Ross River virus/genetics , Sequence Analysis , Alphavirus Infections/virology , Animals , Humans , Papua New Guinea , Phylogeny , Ross River virus/classification , Ross River virus/isolation & purification
11.
J Virol ; 95(6)2021 02 24.
Article in English | MEDLINE | ID: mdl-33361425

ABSTRACT

Ross River virus (RRV) is a mosquito-borne alphavirus that causes epidemics of debilitating musculoskeletal disease. To define the innate immune mechanisms that mediate control of RRV infection, we studied a RRV strain encoding 6 nonsynonymous mutations in nsP1 (RRV-T48-nsP16M) that is attenuated in wild-type (WT) mice and Rag1-/- mice, which are unable to mount adaptive immune responses, but not in mice that lack the capacity to respond to type I interferon (IFN) (Ifnar1-/- mice). Utilizing this attenuated strain, our prior studies revealed that mitochondrial antiviral signaling (MAVS)-dependent production of type I IFN by Ly6Chi monocytes is critical for control of acute RRV infection. Here, we infected Mavs-/- mice with either WT RRV or RRV-T48-nsP16M to elucidate MAVS-independent protective mechanisms. Mavs-/- mice infected with WT RRV developed severe disease and succumbed to infection, whereas those infected with RRV-T48-nsP16M exhibited minimal disease signs. Mavs-/- mice infected with RRV-T48-nsP16M had higher levels of systemic type I IFN than Mavs-/- mice infected with WT virus, and treatment of Mavs-/- mice infected with the attenuated nsP1 mutant virus with an IFNAR1-blocking antibody resulted in a lethal infection. In vitro, type I IFN expression was induced in plasmacytoid dendritic cells (pDCs) cocultured with RRV-infected cells in a MAVS-independent manner, and depletion of pDCs in Mavs-/- mice resulted in increased viral burdens in joint and muscle tissues, suggesting that pDCs are a source of the protective IFN in Mavs-/- mice. These data suggest that pDC production of type I IFN through a MAVS-independent pathway contributes to control of RRV infection.IMPORTANCE Arthritogenic alphaviruses, including Ross River virus (RRV), are human pathogens that cause debilitating acute and chronic musculoskeletal disease and are a significant public health burden. Using an attenuated RRV with enhanced susceptibility to host innate immune responses has revealed key cellular and molecular mechanisms that can mediate control of attenuated RRV infection and that are evaded by more virulent RRV strains. In this study, we found that pDCs contribute to the protective type I interferon response during RRV infection through a mechanism that is independent of the mitochondrial antiviral signaling (MAVS) adaptor protein. These findings highlight a key innate immune mechanism that contributes to control of alphavirus infections.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Alphavirus Infections/immunology , Antiviral Agents/metabolism , Dendritic Cells/immunology , Interferon Type I/metabolism , Ross River virus/pathogenicity , Adaptor Proteins, Signal Transducing/metabolism , Alphavirus Infections/virology , Animals , Dendritic Cells/metabolism , Immunity, Innate , Mice , Mutation , Ross River virus/genetics , Signal Transduction , Viral Load , Viral Nonstructural Proteins/genetics , Virulence/genetics
12.
Viruses ; 12(7)2020 06 27.
Article in English | MEDLINE | ID: mdl-32605094

ABSTRACT

Alphaviruses are globally distributed and predominately transmitted by mosquitoes. Aedes species are common vectors for the clinically important alphaviruses-Chikungunya, Sindbis, and Ross River (RRV) viruses-with Aedes aegypti also being a vector for the flaviviruses dengue, Yellow Fever, and Zika viruses. Ae. aegypti was putatively implicated in the large 1979-1980 South Pacific Islands outbreak of RRV-the leading cause of arboviral disease in Australia today. The RNA interference (RNAi) defense response in mosquitoes involves a number of small RNAs, with their kinetics induced by alphaviruses being poorly understood, particularly at the tissue level. We compared the small RNA profiles between RRV-infected and noninfected Ae. aegypti midgut and fat body tissues at 2, 6, and 12 days post-inoculation (dpi). RRV induced an incremental RNAi response, yielding short interfering and P-element-induced-wimpy-testis (PIWI)-interacting RNAs. Fourteen host microRNAs were differentially expressed due to RRV with the majority in the fat body at 2 dpi. The largely congruent pattern of microRNA regulation with previous reports for alphaviruses and divergence from those for flaviviruses suggests a degree of conservation, whereas patterns of microRNA expression unique to this study provide novel insights into the tissuespecific hostvirus attributes of Ae. aegypti responses to this previously unexplored oldworld alphavirus.


Subject(s)
Aedes/genetics , Aedes/virology , Host Microbial Interactions/genetics , MicroRNAs/genetics , RNA Interference , Ross River virus/genetics , Animals , Female , Gene Expression Profiling , Mosquito Vectors/genetics , Mosquito Vectors/virology , Virus Replication/genetics
13.
J Virol ; 94(8)2020 03 31.
Article in English | MEDLINE | ID: mdl-31996431

ABSTRACT

Ross River virus (RRV) belongs to the genus Alphavirus and is prevalent in Australia. RRV infection can cause arthritic symptoms in patients and may include rash, fever, arthralgia, and myalgia. Type I interferons (IFN) are the primary antiviral cytokines and trigger activation of the host innate immune system to suppress the replication of invading viruses. Alphaviruses are able to subvert the type I IFN system, but the mechanisms used are ill defined. In this study, seven RRV field strains were analyzed for induction of and sensitivity to type I IFN. The sensitivities of these strains to human IFN-ß varied significantly and were highest for the RRV 2548 strain. Compared to prototype laboratory strain RRV-T48, RRV 2548 also induced higher type I IFN levels both in vitro and in vivo and caused milder disease. To identify the determinants involved in type I IFN modulation, the region encoding the nonstructural proteins (nsPs) of RRV 2548 was sequenced, and 42 amino acid differences from RRV-T48 were identified. Using fragment swapping and site-directed mutagenesis, we discovered that substitutions E402A and R522Q in nsP1 as well as Q619R in nsP2 were responsible for increased sensitivity of RRV 2548 to type I IFN. In contrast, substitutions A31T, N219T, S580L, and Q619R in nsP2 led to induction of higher levels of type I IFN. With exception of E402A, all these variations are common for naturally occurring RRV strains. However, they are different from all known determinants of type I IFN modulation reported previously in nsPs of alphaviruses.IMPORTANCE By identifying natural Ross River virus (RRV) amino acid determinants for type I interferon (IFN) modulation, this study gives further insight into the mechanism of type I IFN modulation by alphaviruses. Here, the crucial role of type I IFN in the early stages of RRV disease pathogenesis is further demonstrated. This study also provides a comparison of the roles of different parts of the RRV nonstructural region in type I IFN modulation, highlighting the importance of nonstructural protein 1 (nsP1) and nsP2 in this process. Three substitutions in nsP1 and nsP2 were found to be independently associated with enhanced type I IFN sensitivity, and four independent substitutions in nsP2 were important in elevated type I IFN induction. Such evidence has clear implications for RRV immunobiology, persistence, and pathology. The identification of viral proteins that modulate type I IFN may also have importance for the pathogenesis of other alphaviruses.


Subject(s)
Antiviral Agents/pharmacology , Interferon Type I/immunology , Interferon Type I/pharmacology , Ross River virus/drug effects , Ross River virus/immunology , Alphavirus/genetics , Alphavirus/immunology , Alphavirus Infections/virology , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Cytokines , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Ross River virus/genetics , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Virulence , Virus Replication/drug effects
14.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31666378

ABSTRACT

Ross River virus (RRV), an alphavirus of the Togaviridae family, is the most medically significant mosquito-borne virus of Australia. Past RRV phylogenetic and evolutionary analyses have been based on partial genome analyses only. Three geographically distinct RRV lineages, the Eastern, the Western, and the supposedly extinct North-Eastern lineage, were classified previously. We sought to expand on past phylogenies through robust genome-scale phylogeny to better understand RRV genetic diversity and evolutionary dynamics. We analyzed 106 RRV complete coding sequences, which included 13 genomes available on NCBI and 94 novel sequences derived for this study, sampled throughout Western Australia (1977-2014) and during the substantial Pacific Islands RRV epidemic (1979-1980). Our final data set comprised isolates sampled over 59 years (1959-2018) from a range of locations. Four distinct genotypes were defined, with the newly described genotype 4 (G4) found to be the contemporary lineage circulating in Western Australia. The prior geographical classification of RRV lineages was not supported by our findings, with evidence of geographical and temporal cocirculation of distinct genetic groups. Bayesian Markov chain Monte Carlo (MCMC) analysis revealed that RRV lineages diverged from a common ancestor approximately 94 years ago, with distinct lineages emerging roughly every 10 years over the past 50 years in periodic bursts of genetic diversity. Our study has enabled a more robust analysis of RRV evolutionary history and resolved greater genetic diversity that had been previously defined by partial E2 gene analysis.IMPORTANCE Ross River virus (RRV) causes the most common mosquito-borne infection in Australia and causes a significant burden of suffering to infected individuals as well as being a large burden to the Australian economy. The genetic diversity of RRV and its evolutionary history have so far only been studied using partial E2 gene analysis with a limited number of isolates. Robust whole-genome analysis has not yet been conducted. This study generated 94 novel near-whole-genome sequences to investigate the evolutionary history of RRV to better understand its genetic diversity through comprehensive whole-genome phylogeny. A better understanding of RRV genetic diversity will enable better diagnostics, surveillance, and potential future vaccine design.


Subject(s)
Alphavirus Infections , Epidemics , Evolution, Molecular , Phylogeny , Ross River virus/genetics , Alphavirus Infections/epidemiology , Alphavirus Infections/genetics , Animals , Humans , Ross River virus/classification , Western Australia/epidemiology
15.
Sci Rep ; 9(1): 19398, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852942

ABSTRACT

The ability to identify all the viruses within a sample makes metatranscriptomic sequencing an attractive tool to screen mosquitoes for arboviruses. Practical application of this technique, however, requires a clear understanding of its analytical sensitivity and specificity. To assess this, five dilutions (1:1, 1:20, 1:400, 1:8,000 and 1:160,000) of Ross River virus (RRV) and Umatilla virus (UMAV) isolates were spiked into subsamples of a pool of 100 Culex australicus mosquitoes. The 1:1 dilution represented the viral load of one RRV-infected mosquito in a pool of 100 mosquitoes. The subsamples underwent nucleic acid extraction, mosquito-specific ribosomal RNA depletion, and Illumina HiSeq sequencing. The viral load of the subsamples was also measured using reverse transcription droplet digital PCR (RT-ddPCR) and quantitative PCR (RT-qPCR). Metatranscriptomic sequencing detected both RRV and UMAV in the 1:1, 1:20 and 1:400 subsamples. A high specificity was achieved, with 100% of RRV and 99.6% of UMAV assembled contigs correctly identified. Metatranscriptomic sequencing was not as sensitive as RT-qPCR or RT-ddPCR; however, it recovered whole genome information and detected 19 other viruses, including four first detections for Australia. These findings will assist arbovirus surveillance programs in utilising metatranscriptomics in routine surveillance activities to enhance arbovirus detection.


Subject(s)
Arboviruses/genetics , Culicidae/virology , Metagenome/genetics , Transcriptome/genetics , Animals , Arboviruses/isolation & purification , Australia/epidemiology , Culex/genetics , Culex/virology , Culicidae/genetics , Humans , Mosquito Vectors/genetics , Mosquito Vectors/virology , RNA, Viral/genetics , RNA, Viral/isolation & purification , Ross River virus/genetics , Ross River virus/isolation & purification , Sensitivity and Specificity
16.
Emerg Infect Dis ; 25(12): 2243-2252, 2019 12.
Article in English | MEDLINE | ID: mdl-31742522

ABSTRACT

Australia experienced its largest recorded outbreak of Ross River virus (RRV) during the 2014-15 reporting year, comprising >10,000 reported cases. We investigated epidemiologic, entomologic, and virologic factors that potentially contributed to the scale of the outbreak in Queensland, the state with the highest number of notifications (6,371). Spatial analysis of human cases showed that notifications were geographically widespread. In Brisbane, human case notifications and virus detections in mosquitoes occurred across inland and coastal locations. Viral sequence data demonstrated 2 RRV lineages (northeastern genotypes I and II) were circulating, and a new strain containing 3 unique amino acid changes in the envelope 2 protein was identified. Longitudinal mosquito collections demonstrated unusually high relative abundance of Culex annulirostris and Aedes procax mosquitoes, attributable to extensive freshwater larval habitats caused by early and persistent rainfall during the reporting year. Increased prevalence of these mosquitoes probably contributed to the scale of this outbreak.


Subject(s)
Alphavirus Infections/epidemiology , Alphavirus Infections/virology , Ross River virus , Alphavirus Infections/history , Alphavirus Infections/transmission , Disease Outbreaks , Genes, Viral , Geography, Medical , History, 21st Century , Humans , Mosquito Vectors/virology , Phylogeny , Public Health Surveillance , Queensland/epidemiology , Ross River virus/classification , Ross River virus/genetics , Ross River virus/immunology
17.
J Am Mosq Control Assoc ; 35(3): 220-223, 2019 09.
Article in English | MEDLINE | ID: mdl-31647704

ABSTRACT

An arbovirus surveillance military exercise was conducted to assess the risk of Ross River virus (RRV) and Barmah Forest virus (BFV) in the Australian Defence Force (ADF) Wide Bay training area (WBTA), northeastern Australia, in April 2018. Of the 5,540 female mosquitoes collected, 3,702 were screened for RRV and BFV by quantitative reverse transcription-polymerase chain reaction in a field laboratory. One pool of Verrallina funerea was positive for RRV and 8 pools (7 pools of Aedes vigilax and 1 pool of Culex annulirostris) were positive for BFV. Phylogenetic analysis of the complete nucleotide sequence of the E2 protein subgrouped both RRV and BFV with viruses previously isolated from human infections, indicating the potential risk of RRV and BFV infection to ADF personnel while training in WBTA. This is the 1st time that both RRV and BFV have been detected in a military training area.


Subject(s)
Alphavirus/isolation & purification , Culicidae/virology , Ross River virus/isolation & purification , Alphavirus/genetics , Animals , Female , Military Personnel , Population Surveillance , Queensland , Ross River virus/genetics
18.
Emerg Infect Dis ; 25(10): 1793-1801, 2019 10.
Article in English | MEDLINE | ID: mdl-31538560

ABSTRACT

Two outbreaks of epidemic polyarthritis occurred among Australian Defence Force personnel during and following short military exercises in the Shoalwater Bay Training Area, northeastern Australia, in 2016 and 2017. Ross River virus (RRV) IgM was detected in acute-phase serum samples from most patients (28/28 in 2016 and 25/31 in 2017), and RRV was recovered from 4/38 serum samples assayed (1/21 in 2016 and 3/17 in 2017). Phylogenetic analyses of RRV envelope glycoprotein E2 and nonstructural protein nsP3 nucleotide sequences segregated the RRV isolates obtained in 2016 and 2017 outbreaks into 2 distinct sublineages, suggesting that each outbreak was caused by a different strain of RRV. The spatiotemporal characteristics of the 2016 outbreak suggested that some of the infections involved human-mosquito-human transmission without any intermediate host. These outbreaks highlight the importance of personal protective measures in preventing vectorborne diseases for which no vaccine or specific prophylaxis exists.


Subject(s)
Alphavirus Infections/epidemiology , Arthritis/epidemiology , Epidemics , Military Personnel , Ross River virus , Adult , Alphavirus Infections/virology , Arthritis/virology , Epidemics/statistics & numerical data , Female , Humans , Male , Middle Aged , Military Personnel/statistics & numerical data , Phylogeny , Queensland/epidemiology , Ross River virus/genetics , Young Adult
19.
Viruses ; 11(7)2019 06 27.
Article in English | MEDLINE | ID: mdl-31252609

ABSTRACT

Arboviruses like chikungunya and Ross River (RRV) are responsible for massive outbreaks of viral polyarthritis. There is no effective treatment or vaccine available against these viruses that induce prolonged and disabling arthritis. To explore the physiopathological mechanisms of alphaviral arthritis, we engineered a recombinant RRV expressing a NanoLuc reporter (RRV-NLuc), which exhibited high stability, near native replication kinetics and allowed real time monitoring of viral spread in an albino mouse strain. During the acute phase of the disease, we observed a high bioluminescent signal reflecting viral replication and dissemination in the infected mice. Using Bindarit, an anti-inflammatory drug that inhibits monocyte recruitment, we observed a reduction in viral dissemination demonstrating the important role of monocytes in the propagation of the virus and the adaptation of this model to the in vivo evaluation of treatment strategies. After resolution of the acute symptoms, we observed an increase in the bioluminescent signal in mice subjected to an immunosuppressive treatment 30 days post infection, thus showing active in vivo replication of remnant virus. We show here that this novel reporter virus is suitable to study the alphaviral disease up to the chronic phase, opening new perspectives for the evaluation of therapeutic interventions.


Subject(s)
Alphavirus Infections/virology , Ross River virus/physiology , Alphavirus Infections/diagnostic imaging , Animals , Arthritis/diagnostic imaging , Arthritis/virology , Disease Models, Animal , Genes, Reporter , Humans , Luminescent Measurements , Mice , Mice, Inbred C57BL , Ross River virus/chemistry , Ross River virus/genetics
20.
mBio ; 9(4)2018 08 21.
Article in English | MEDLINE | ID: mdl-30131356

ABSTRACT

Infection with Ross River virus (RRV) causes debilitating polyarthritis and arthralgia in individuals. Alphaviruses are highly sensitive to type I interferon (IFN). Mutations at the conserved P3 position of the cleavage site between nonstructural protein 1 (nsP1) and nsP2 (1/2 site) modulate type I IFN induction for both RRV and Sindbis virus (SINV). We constructed and characterized RRV-T48A534V, a mutant harboring an A534V substitution in the P1 position of the 1/2 site, and compared it to parental RRV-T48 and to RRV-T48A532V, SINVI538 and SINVT538 harboring different substitutions in the same region. A534V substitution resulted in impaired processing of RRV nonstructural polyprotein and in elevated production of replicase-generated pathogen-associated molecular pattern (PAMP) RNAs that induce expression of type I IFN. Both A532V and A534V substitutions affected synthesis of viral RNAs, though the effects of these closely located mutations were drastically different affecting mostly either the viral negative-strand RNA or genomic and subgenomic RNA levels, respectively. Synthesis of PAMP RNAs was also observed for SINV replicase, and it was increased by I538T substitution. In comparison to RRV-T48, RRV-T48A534V was attenuated in vitro and in vivo Interestingly, when type I IFN-deficient cells and type I IFN receptor-deficient mice were infected with RRV-T48 or RRV-T48A534V, differences between these viruses were no longer apparent. Compared to RRV-T48, RRV-T48A534V infection was associated with increased upregulation of type I IFN signaling proteins. We demonstrate novel mechanisms by which the A534V mutation affect viral nonstructural polyprotein processing that can impact PAMP RNA production, type I IFN induction/sensitivity, and disease.IMPORTANCE This study gives further insight into mechanisms of type I IFN modulation by the medically important alphaviruses Ross River virus (RRV) and Sindbis virus (SINV). By characterizing attenuated RRV mutants, the crucial role of amino acid residues in P1 and P3 positions (the first and third amino acid residues preceding the scissile bond) of the cleavage site between nsP1 and nsP2 regions was highlighted. The study uncovers a unique relationship between alphavirus nonstructural polyprotein processing, RNA replication, production of different types of pathogen-associated molecular pattern (PAMP) RNAs, type I IFN induction, and disease pathogenesis. This study also highlights the importance of the host innate immune response in RRV infections. The viral determinants of type I IFN modulation provide potential drug targets for clinical treatment of alphaviral disease and offer new approaches for rational attenuation of alphaviruses for construction of vaccine candidates.


Subject(s)
Interferons/metabolism , Mutant Proteins/immunology , Mutation, Missense , Polyproteins/metabolism , RNA, Viral/immunology , Ross River virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Alphavirus Infections/pathology , Alphavirus Infections/virology , Animals , Antiviral Agents/metabolism , Disease Models, Animal , Mice , Mutant Proteins/genetics , Polyproteins/genetics , RNA, Viral/metabolism , Ross River virus/genetics , Ross River virus/immunology , Sindbis Virus/genetics , Sindbis Virus/immunology , Sindbis Virus/pathogenicity , Viral Nonstructural Proteins/genetics , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...