Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
J Virol ; 96(17): e0099922, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000846

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Subject(s)
Alphavirus Infections , Arthritis , Myositis , Polysaccharides , Ross River virus , Skin , Alphavirus Infections/complications , Alphavirus Infections/immunology , Animals , Antiviral Agents/immunology , Arthritis/complications , Arthritis/immunology , Culicidae/virology , Dendritic Cells , Disease Models, Animal , Glycosylation , Humans , Mass Spectrometry , Mice , Monocytes , Myositis/complications , Myositis/immunology , Neutrophils , Polysaccharides/chemistry , Polysaccharides/immunology , Ross River virus/immunology , Skin/immunology , Skin/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
2.
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
3.
PLoS Pathog ; 18(2): e1010185, 2022 02.
Article in English | MEDLINE | ID: mdl-35143591

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne viruses that are a major cause of infectious arthropathies worldwide, and recent outbreaks of chikungunya virus and Ross River virus (RRV) infections highlight the need for robust intervention strategies. Alphaviral arthritis can persist for months after the initial acute disease, and is mediated by cellular immune responses. A common strategy to limit inflammation and pathology is to dampen the overwhelming inflammatory responses by modulating proinflammatory cytokine pathways. Here, we investigate the contribution of interleukin-17 (IL-17), a cytokine involved in arthropathies such as rheumatoid arthritis, in the development RRV-induced arthritis and myositis. IL-17 was quantified in serum from RRV-infected patients, and mice were infected with RRV and joints and muscle tissues collected to analyse cellular infiltrates, tissue mRNA, cytokine expression, and joint and muscle histopathology. IL-17 expression was increased in musculoskeletal tissues and serum of RRV-infected mice and humans, respectively. IL-17-producing T cells and neutrophils contributed to the cellular infiltrate in the joint and muscle tissue during acute RRV disease in mice. Blockade of IL-17A/F using a monoclonal antibody (mAb) reduced disease severity in RRV-infected mice and led to decreased proinflammatory proteins, cellular infiltration in synovial tissues and cartilage damage, without affecting viral titers in inflamed tissues. IL-17A/F blockade triggered a shift in transcriptional profile of both leukocyte infiltrates and musculoskeletal stromal cells by downregulating proinflammatory genes. This study highlights a previously uncharacterized role for an effector cytokine in alphaviral pathology and points towards potential therapeutic benefit in targeting IL-17 to treat patients presenting with RRV-induced arthropathy.


Subject(s)
Arthritis, Rheumatoid/immunology , Immunity, Cellular , Inflammation/immunology , Interleukin-17/immunology , Myositis/immunology , Ross River virus/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Animals , Arthritis, Rheumatoid/virology , Chlorocebus aethiops , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Myositis/virology , Vero Cells , Viral Load
4.
PLoS Pathog ; 16(8): e1008743, 2020 08.
Article in English | MEDLINE | ID: mdl-32760128

ABSTRACT

Arthritogenic alphaviruses cause debilitating musculoskeletal disease and historically have circulated in distinct regions. With the global spread of chikungunya virus (CHIKV), there now is more geographic overlap, which could result in heterologous immunity affecting natural infection or vaccination. Here, we evaluated the capacity of a cross-reactive anti-CHIKV monoclonal antibody (CHK-265) to protect against disease caused by the distantly related alphavirus, Ross River virus (RRV). Although CHK-265 only moderately neutralizes RRV infection in cell culture, it limited clinical disease in mice independently of Fc effector function activity. Despite this protective phenotype, RRV escaped from CHK-265 neutralization in vivo, with resistant variants retaining pathogenic potential. Near the inoculation site, CHK-265 reduced viral burden in a type I interferon signaling-dependent manner and limited immune cell infiltration into musculoskeletal tissue. In a parallel set of experiments, purified human CHIKV immune IgG also weakly neutralized RRV, yet when transferred to mice, resulted in improved clinical outcome during RRV infection despite the emergence of resistant viruses. Overall, this study suggests that weakly cross-neutralizing antibodies can protect against heterologous alphavirus disease, even if neutralization escape occurs, through an early viral control program that tempers inflammation.


Subject(s)
Alphavirus Infections/complications , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cross Reactions/immunology , Musculoskeletal Diseases/prevention & control , Ross River virus/isolation & purification , Viral Load/immunology , Alphavirus Infections/virology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Musculoskeletal Diseases/immunology , Musculoskeletal Diseases/virology , Receptors, Fc/physiology , Ross River virus/immunology , Virulence
5.
PLoS One ; 15(7): e0229314, 2020.
Article in English | MEDLINE | ID: mdl-32706777

ABSTRACT

INTRODUCTION: Many arboviruses of public health significance are maintained in zoonotic cycles with complex transmission pathways. The presence of serum antibody against arboviruses in vertebrates provides evidence of their historical exposure but reveals nothing about the vector-reservoir relationship. Moreover, collecting blood or tissue samples from vertebrate hosts is ethically and logistically challenging. We developed a novel approach for screening the immune status of vertebrates against Ross River virus that allows us to implicate the vectors that form the transmission pathways for this commonly notified Australian arboviral disease. METHODS: A micro-plaque reduction neutralisation test (micro-PRNT) was developed and validated on koala (Phascolarctos cinereus) sera against a standard PRNT. The ability of the micro-PRNT to detect RRV antibodies in mosquito blood meals was then tested using two mosquito models. Laboratory-reared Aedes aegypti were fed, via a membrane, on sheep blood supplemented with RRV seropositive and seronegative human sera. Aedes notoscriptus were fed on RRV seropositive and seronegative human volunteers. Blood-fed mosquitoes were harvested at various time points after feeding and their blood meals analysed for the presence of RRV neutralising antibodies using the micro-PRNT. RESULTS: There was significant agreement of the plaque neutralisation resulting from the micro-PRNT and standard PRNT techniques (R2 = 0.65; P<0.0001) when applied to RRV antibody detection in koala sera. Sensitivity and specificity of the micro-PRNT assay were 88.2% and 96%, respectively, in comparison with the standard PRNT. Blood meals from mosquitoes fed on sheep blood supplemented with RRV antibodies, and on blood from RRV seropositive humans neutralised the virus by ≥50% until 48 hr post feeding. The vertebrate origin of the blood meal was also ascertained for the same samples, in parallel, using established molecular techniques. CONCLUSIONS: The small volumes of blood present in mosquito abdomens can be used to identify RRV antibodies and therefore host exposure to arbovirus infection. In tandem with the accurate identification of the mosquito, and diagnostics for the host origin of the blood meal, this technique has tremendous potential for exploring RRV transmission pathways. It can be adapted for similar studies on other mosquito borne zoonoses.


Subject(s)
Aedes/metabolism , Animal Feed/analysis , Antibodies, Viral/analysis , Neutralization Tests/methods , Ross River virus/immunology , Aedes/virology , Alphavirus Infections/diagnosis , Alphavirus Infections/transmission , Alphavirus Infections/veterinary , Animal Feed/virology , Animals , Antibodies, Viral/blood , Disease Vectors , Female , Humans , Phascolarctidae/virology , Sensitivity and Specificity
6.
PLoS Pathog ; 16(5): e1008517, 2020 05.
Article in English | MEDLINE | ID: mdl-32365139

ABSTRACT

Ross River fever is a mosquito-transmitted viral disease that is endemic to Australia and the surrounding Pacific Islands. Ross River virus (RRV) belongs to the arthritogenic group of alphaviruses, which largely cause disease characterized by debilitating polyarthritis, rash, and fever. There is no specific treatment or licensed vaccine available, and the mechanisms of protective humoral immunity in humans are poorly understood. Here, we describe naturally occurring human mAbs specific to RRV, isolated from subjects with a prior natural infection. These mAbs potently neutralize RRV infectivity in cell culture and block infection through multiple mechanisms, including prevention of viral attachment, entry, and fusion. Some of the most potently neutralizing mAbs inhibited binding of RRV to Mxra8, a recently discovered alpahvirus receptor. Epitope mapping studies identified the A and B domains of the RRV E2 protein as the major antigenic sites for the human neutralizing antibody response. In experiments in mice, these mAbs were protective against cinical disease and reduced viral burden in multiple tissues, suggesting a potential therapeutic use for humans.


Subject(s)
Alphavirus Infections/prevention & control , Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Capsid Proteins/immunology , Epitopes/immunology , Ross River virus/immunology , Viral Envelope Proteins/immunology , Alphavirus Infections/immunology , Alphavirus Infections/pathology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Chlorocebus aethiops , Female , Humans , Mice , Middle Aged , Vero Cells
7.
Int J Infect Dis ; 96: 94-96, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32114197

ABSTRACT

BACKGROUND: Ross River virus (RRV) is a zoonotic alphavirus transmitted by several mosquito species. Until recently, endemic transmission was only considered possible in the presence of marsupial reservoirs. METHODS: RRV seroprevalence was investigated in placental mammals (including horses, cows, goats, pigs, dogs, rats, and mice) in Fiji, where there are no marsupials. A total of 302 vertebrate serum samples were collected from 86 households from 10 communities in Western Fiji. RESULTS: Neutralizing antibodies against RRV were detected in 28% to 100% of sera depending on the species, and neutralization was strong even at high dilutions. CONCLUSIONS: These results are unlikely to be due to cross-reactions. Chikungunya is the only other alphavirus known to be present in the Pacific Islands, but it rarely spills over into non-humans, even during epidemics. The study findings, together with a recent report of high RRV seroprevalence in humans, strongly suggest that RRV is circulating in Fiji in the absence of marsupial reservoirs. Considering that all non-human vertebrates present in Fiji are pan-global in distribution, RRV has the potential to further expand its geographic range. Further surveillance of RRV and access to RRV diagnostics will be critical for the early detection of emergence and outbreaks.


Subject(s)
Alphavirus Infections/veterinary , Ross River virus , Alphavirus Infections/epidemiology , Alphavirus Infections/transmission , Animals , Cattle , Dogs , Female , Fiji/epidemiology , Goats/virology , Horses/virology , Humans , Marsupialia , Mice , Pregnancy , Rats , Ross River virus/immunology , Seroepidemiologic Studies , Swine/virology
8.
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
9.
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
10.
Aust J Gen Pract ; 48(9): 645-649, 2019 09.
Article in English | MEDLINE | ID: mdl-31476825

ABSTRACT

BACKGROUND: Ross River virus (RRV) and Barmah Forest virus (BFV) cause approximately 4000 and 1000 cases, respectively, of rheumatic disease in Australia every year. Confirmation of a diagnosis usually involves testing for virus-specific immunoglobulin (Ig) M and IgG by a National Association of Testing Authorities-accredited pathology facility. OBJECTIVE: The aim of the article is to provide a logical framework by which clinicians can interpret paired RRV and BFV serology results in environments in which numerical antibody titres are no longer routinely provided. The traditional recommendation to look for an increase in titres is now largely obsolete. DISCUSSION: Paired serology is clinical best practice but needs to be appropriately interpreted given the false positive and negative rates, the large number of asymptomatic infections and the long-term persistence of IgM in some individuals. An inappropriate interpretation risks a misdiagnosis.


Subject(s)
Alphavirus Infections/immunology , Alphavirus/immunology , Antibodies, Viral/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Rheumatic Diseases/immunology , Ross River virus/immunology , Serologic Tests/methods , Alphavirus Infections/diagnosis , Australia , False Negative Reactions , False Positive Reactions , Humans , Rheumatic Diseases/diagnosis , Rheumatic Diseases/virology
11.
Emerg Infect Dis ; 25(4): 827-830, 2019 04.
Article in English | MEDLINE | ID: mdl-30882332

ABSTRACT

A unique outbreak of Ross River virus (RRV) infection was reported in Fiji in 1979. In 2013, RRV seroprevalence among residents was 46.5% (362/778). Of the residents who were seronegative in 2013 and retested in 2015, 10.9% (21/192) had seroconverted to RRV, suggesting ongoing endemic circulation of RRV in Fiji.


Subject(s)
Alphavirus Infections/diagnosis , Ross River virus/immunology , Alphavirus Infections/blood , Alphavirus Infections/epidemiology , Antibodies, Viral/blood , Fiji/epidemiology , Humans , Ross River virus/isolation & purification , Seroepidemiologic Studies
12.
Viruses ; 11(1)2019 01 18.
Article in English | MEDLINE | ID: mdl-30669393

ABSTRACT

BACKGROUND: Alphaviruses are transmitted by arthropod vectors and can be found worldwide. Alphaviruses of the Semliki Forest complex such as chikungunya virus (CHIKV), Mayaro virus (MAYV) or Ross River virus (RRV) cause acute febrile illness and long-lasting arthralgia in humans, which cannot be clinically discriminated from a dengue virus or Zika virus infection. Alphaviruses utilize a diverse array of mosquito vectors for transmission and spread. For instance, adaptation of CHIKV to transmission by Aedes albopictus has increased its spread and resulted in large outbreaks in the Indian Ocean islands. For many alphaviruses commercial diagnostic tests are not available or show cross-reactivity among alphaviruses. Climate change and globalization will increase the spread of alphaviruses and monitoring of infections is necessary and requires virus-specific methods. METHOD: We established an alphavirus neutralization assay in a 384-well format by using pseudotyped lentiviral vectors. RESULTS: MAYV-specific reactivity could be discriminated from CHIKV reactivity. Human plasma from blood donors infected with RRV could be clearly identified and did not cross-react with other alphaviruses. CONCLUSION: This safe and easy to use multiplex assay allows the discrimination of alphavirus-specific reactivity within a single assay and has potential for epidemiological surveillance. It might also be useful for the development of a pan-alphavirus vaccine.


Subject(s)
Alphavirus Infections/diagnosis , Alphavirus Infections/immunology , Neutralization Tests/methods , Ross River virus/immunology , Animals , Chikungunya virus/immunology , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Humans , Inhibitory Concentration 50 , Mosquito Vectors/virology , Semliki forest virus/immunology , Sensitivity and Specificity
13.
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
14.
Virology ; 515: 250-260, 2018 02.
Article in English | MEDLINE | ID: mdl-29324290

ABSTRACT

Mannose binding lectin (MBL) generally plays a protective role during viral infection, yet MBL-mediated complement activation promotes Ross River virus (RRV)-induced inflammatory tissue destruction, contributing to arthritis and myositis. As MBL binds to carbohydrates, we hypothesized that N-linked glycans on the RRV envelope glycoproteins act as ligands for MBL. Using a panel of RRV mutants lacking the envelope N-linked glycans, we found that MBL deposition onto infected cells was dependent on the E2 glycans. Moreover, the glycan-deficient viruses exhibited reduced disease and tissue damage in a mouse model of RRV-induced myositis compared to wild-type RRV, despite similar viral load and inflammatory infiltrates within the skeletal muscle. Instead, the reduced disease induced by glycan-deficient viruses was linked to decreased MBL deposition and complement activation within inflamed tissues. These results demonstrate that the viral N-linked glycans promote MBL deposition and complement activation onto RRV-infected cells, contributing to the development of RRV-induced myositis.


Subject(s)
Alphavirus Infections/immunology , Complement System Proteins/immunology , Polysaccharides/immunology , Ross River virus/immunology , Viral Envelope Proteins/immunology , Alphavirus Infections/virology , Animals , Complement Activation , Disease Models, Animal , Humans , Mannose-Binding Lectin/immunology , Mice, Inbred C57BL , Polysaccharides/chemistry , Ross River virus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
15.
PLoS Pathog ; 13(12): e1006748, 2017 12.
Article in English | MEDLINE | ID: mdl-29244871

ABSTRACT

Chikungunya virus (CHIKV) and Ross River virus (RRV) are mosquito-transmitted alphaviruses that cause debilitating acute and chronic musculoskeletal disease. Monocytes are implicated in the pathogenesis of these infections; however, their specific roles are not well defined. To investigate the role of inflammatory Ly6ChiCCR2+ monocytes in alphavirus pathogenesis, we used CCR2-DTR transgenic mice, enabling depletion of these cells by administration of diptheria toxin (DT). DT-treated CCR2-DTR mice displayed more severe disease following CHIKV and RRV infection and had fewer Ly6Chi monocytes and NK cells in circulation and muscle tissue compared with DT-treated WT mice. Furthermore, depletion of CCR2+ or Gr1+ cells, but not NK cells or neutrophils alone, restored virulence and increased viral loads in mice infected with an RRV strain encoding attenuating mutations in nsP1 to levels detected in monocyte-depleted mice infected with fully virulent RRV. Disease severity and viral loads also were increased in DT-treated CCR2-DTR+;Rag1-/- mice infected with the nsP1 mutant virus, confirming that these effects are independent of adaptive immunity. Monocytes and macrophages sorted from muscle tissue of RRV-infected mice were viral RNA positive and had elevated expression of Irf7, and co-culture of Ly6Chi monocytes with RRV-infected cells resulted in induction of type I IFN gene expression in monocytes that was Irf3;Irf7 and Mavs-dependent. Consistent with these data, viral loads of the attenuated nsP1 mutant virus were equivalent to those of WT RRV in Mavs-/- mice. Finally, reconstitution of Irf3-/-;Irf7-/- mice with CCR2-DTR bone marrow rescued mice from severe infection, and this effect was reversed by depletion of CCR2+ cells, indicating that CCR2+ hematopoietic cells are capable of inducing an antiviral response. Collectively, these data suggest that MAVS-dependent production of type I IFN by monocytes is critical for control of acute alphavirus infection and that determinants in nsP1, the viral RNA capping protein, counteract this response.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/virology , Monocytes/immunology , Monocytes/virology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Antigens, Ly/metabolism , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/immunology , Humans , Inflammation/virology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/drug effects , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Ross River virus/genetics , Ross River virus/immunology , Ross River virus/pathogenicity , Viral Load , Virulence/genetics , Virulence/immunology
16.
PLoS Pathog ; 13(12): e1006788, 2017 12.
Article in English | MEDLINE | ID: mdl-29281739

ABSTRACT

Chikungunya virus (CHIKV) belongs to a group of mosquito-borne alphaviruses associated with acute and chronic arthropathy, with peripheral and limb joints most commonly affected. Using a mouse model of CHIKV infection and arthritic disease, we show that CHIKV replication and the ensuing foot arthropathy were dramatically reduced when mice were housed at 30°C, rather than the conventional 22°C. The effect was not associated with a detectable fever, but was dependent on type I interferon responses. Bioinformatics analyses of RNA-Seq data after injection of poly(I:C)/jetPEI suggested the unfolded protein response and certain type I interferon responses are promoted when feet are slightly warmer. The ambient temperature thus appears able profoundly to effect anti-viral activity in the periphery, with clear consequences for alphaviral replication and the ensuing arthropathy. These observations may provide an explanation for why alphaviral arthropathies are largely restricted to joints of the limbs and the extremities.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/virology , Arthritis, Experimental/immunology , Arthritis, Experimental/virology , Arthritis, Infectious/immunology , Arthritis, Infectious/virology , Interferon Type I/metabolism , Alphavirus Infections/pathology , Animals , Arthritis, Experimental/pathology , Arthritis, Infectious/pathology , Chikungunya Fever/immunology , Chikungunya Fever/pathology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Chikungunya virus/physiology , Female , Foot , Host-Pathogen Interactions/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Ross River virus/immunology , Ross River virus/pathogenicity , Ross River virus/physiology , Temperature , Viral Load , Virus Replication/immunology , Virus Replication/physiology
17.
Emerg Infect Dis ; 23(10): 1751-1753, 2017 10.
Article in English | MEDLINE | ID: mdl-28930020

ABSTRACT

Ross River virus (RRV), spread by Aedes and Culex mosquitoes, is the most commonly transmitted arbovirus in Australia. A serosurvey of blood donors in French Polynesia during 2011-2013 suggested that RRV circulated without being detected. We report RRV circulation in French Polynesia based on further screening of blood samples collected during 2014-2015.


Subject(s)
Aedes/virology , Alphavirus Infections/epidemiology , Antibodies, Viral/blood , Culex/virology , Immunoglobulin G/blood , Insect Vectors/virology , Ross River virus/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Alphavirus Infections/transmission , Alphavirus Infections/virology , Animals , Asymptomatic Diseases , Blood Donors , Child , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Polynesia/epidemiology , Ross River virus/immunology , Seroepidemiologic Studies
18.
J Virol ; 90(8): 4150-4159, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865723

ABSTRACT

UNLABELLED: The alphaviral6kgene region encodes the two structural proteins 6K protein and, due to a ribosomal frameshift event, the transframe protein (TF). Here, we characterized the role of the6kproteins in the arthritogenic alphavirus Ross River virus (RRV) in infected cells and in mice, using a novel6kin-frame deletion mutant. Comprehensive microscopic analysis revealed that the6kproteins were predominantly localized at the endoplasmic reticulum of RRV-infected cells. RRV virions that lack the6kproteins 6K and TF [RRV-(Δ6K)] were more vulnerable to changes in pH, and the corresponding virus had increased sensitivity to a higher temperature. While the6kdeletion did not reduce RRV particle production in BHK-21 cells, it affected virion release from the host cell. Subsequentin vivostudies demonstrated that RRV-(Δ6K) caused a milder disease than wild-type virus, with viral titers being reduced in infected mice. Immunization of mice with RRV-(Δ6K) resulted in a reduced viral load and accelerated viral elimination upon secondary infection with wild-type RRV or another alphavirus, chikungunya virus (CHIKV). Our results show that the6kproteins may contribute to alphaviral disease manifestations and suggest that manipulation of the6kgene may be a potential strategy to facilitate viral vaccine development. IMPORTANCE: Arthritogenic alphaviruses, such as chikungunya virus (CHIKV) and Ross River virus (RRV), cause epidemics of debilitating rheumatic disease in areas where they are endemic and can emerge in new regions worldwide. RRV is of considerable medical significance in Australia, where it is the leading cause of arboviral disease. The mechanisms by which alphaviruses persist and cause disease in the host are ill defined. This paper describes the phenotypic properties of an RRV6kdeletion mutant. The absence of the6kgene reduced virion release from infected cells and also reduced the severity of disease and viral titers in infected mice. Immunization with the mutant virus protected mice against viremia not only upon exposure to RRV but also upon challenge with CHIKV. These findings could lead to the development of safer and more immunogenic alphavirus vectors for vaccine delivery.


Subject(s)
Alphavirus Infections/virology , Ross River virus/genetics , Ross River virus/immunology , Viral Structural Proteins/genetics , Alphavirus Infections/immunology , Alphavirus Infections/physiopathology , Animals , Cell Line , Cell Line, Tumor , Chikungunya virus/immunology , Chlorocebus aethiops , Cricetinae , Humans , Hydrogen-Ion Concentration , Mice , Mutation , Reading Frames , Ross River virus/pathogenicity , Sequence Deletion , Vero Cells , Viral Load , Viral Structural Proteins/analysis , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Virus Replication
19.
Mol Immunol ; 68(2 Pt B): 465-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26476978

ABSTRACT

Rheumatoid arthritis (RA) develops in response to both genetic and environmental factors. The strongest genetic determinant is HLA-DR, where polymorphisms within the P4 and P6 binding pockets confer elevated risk. However, low disease concordance across monozygotic twin pairs underscores the importance of an environmental factor, probably infectious. The goal of this investigation was to predict the microorganism most likely to interact with HLA-DR to trigger RA under the molecular mimicry hypothesis. A set of 185 structural proteins from viruses or intracellular bacteria was scanned for regions of sequence homology with a collagen peptide that binds preferentially to DR4; candidates were then evaluated against a motif required for T cell cross-reactivity. The plausibility of the predicted agent was evaluated by comparison of microbial prevalence patterns to epidemiological characteristics of RA. Peptides from alphavirus capsid proteins provided the closest fit. Variations in the P6 position suggest that the HLA binding preference may vary by species, with Ross River virus, Chikungunya virus, and Mayaro virus peptides binding preferentially to DR4, and peptides from Sindbis/Ockelbo virus showing stronger affinity to DR1. The predicted HLA preference is supported by epidemiological studies of post-infection chronic arthralgia. Parallels between the cytokine profiles of RA and chronic alphavirus infection are discussed.


Subject(s)
Alphavirus/chemistry , Arthritis, Rheumatoid/virology , Capsid Proteins/immunology , HLA-DR1 Antigen/immunology , HLA-DR4 Antigen/immunology , Alphavirus/genetics , Alphavirus Infections/epidemiology , Amino Acid Motifs , Arthritis, Rheumatoid/immunology , Bacterial Proteins/immunology , Chikungunya virus/immunology , Computer Simulation , Humans , Molecular Sequence Data , Protein Binding/immunology , Ross River virus/immunology , Sindbis Virus/immunology , T-Lymphocytes/immunology , Twins, Monozygotic
20.
PLoS Pathog ; 11(10): e1005191, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26436766

ABSTRACT

Arthritogenic alphaviruses, including Ross River virus (RRV) and chikungunya virus (CHIKV), are responsible for explosive epidemics involving millions of cases. These mosquito-transmitted viruses cause inflammation and injury in skeletal muscle and joint tissues that results in debilitating pain. We previously showed that arginase 1 (Arg1) was highly expressed in myeloid cells in the infected and inflamed musculoskeletal tissues of RRV- and CHIKV-infected mice, and specific deletion of Arg1 from myeloid cells resulted in enhanced viral control. Here, we show that Arg1, along with other genes associated with suppressive myeloid cells, is induced in PBMCs isolated from CHIKV-infected patients during the acute phase as well as the chronic phase, and that high Arg1 expression levels were associated with high viral loads and disease severity. Depletion of both CD4 and CD8 T cells from RRV-infected Arg1-deficient mice restored viral loads to levels detected in T cell-depleted wild-type mice. Moreover, Arg1-expressing myeloid cells inhibited virus-specific T cells in the inflamed and infected musculoskeletal tissues, but not lymphoid tissues, following RRV infection in mice, including suppression of interferon-γ and CD69 expression. Collectively, these data enhance our understanding of the immune response following arthritogenic alphavirus infection and suggest that immunosuppressive myeloid cells may contribute to the duration or severity of these debilitating infections.


Subject(s)
Alphavirus Infections/immunology , Arginase/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology , Viral Load/immunology , Adoptive Transfer , Animals , Blotting, Western , Chikungunya Fever/immunology , Chikungunya virus , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Ross River virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL