Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 382
Filter
1.
Appl Microbiol Biotechnol ; 108(1): 355, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822832

ABSTRACT

Getah virus (GETV) is a re-emerging mosquito-borne alphavirus that is highly pathogenic, mainly to pigs and horses. There are no vaccines or treatments available for GETV in swine in China. Therefore, the development of a simple, rapid, specific, and sensitive serological assay for GETV antibodies is essential for the prevention and control of GETV. Current antibody monitoring methods are time-consuming, expensive, and dependent on specialized instrumentation, and these features are not conducive to rapid detection in clinical samples. To address these problem, we developed immunochromatographic test strips (ICTS) using eukaryotically expressed soluble recombinant p62-E1 protein of GETV as a labelled antigen, which has good detection sensitivity and no cross-reactivity with other common porcine virus-positive sera. The ICTS is highly compatible with IFA and ELISA and can be stored for 1 month at 37 °C and for at least 3 months at room temperature. Hence, p62-E1-based ICTS is a rapid, accurate, and convenient method for rapid on-site detection of GETV antibodies. KEY POINTS: • We established a rapid antibody detection method that can monitor GETV infection • We developed colloidal gold test strips with high sensitivity and specificity • The development of colloidal gold test strips will aid in the field serologic detection of GETV.


Subject(s)
Alphavirus , Antibodies, Viral , Gold Colloid , Sensitivity and Specificity , Animals , Gold Colloid/chemistry , Antibodies, Viral/blood , Antibodies, Viral/immunology , Alphavirus/immunology , Swine , Chromatography, Affinity/methods , Alphavirus Infections/diagnosis , Alphavirus Infections/immunology , Swine Diseases/diagnosis , Swine Diseases/virology , Reagent Strips , China , Enzyme-Linked Immunosorbent Assay/methods
2.
Dev Comp Immunol ; 157: 105193, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38729458

ABSTRACT

The development and persistence of antibody secreting cells (ASC) after antigenic challenge remain inadequately understood in teleosts. In this study, intraperitoneal (ip) injection of Atlantic salmon (Salmo salar) with salmonid alphavirus (WtSAV3) increased the total ASC response, peaking 3-6 weeks post injection (wpi) locally in the peritoneal cavity (PerC) and in systemic lymphoid tissues, while at 13 wpi the response was only elevated in PerC. At the same time point a specific ASC response was induced by WtSAV3 in PerC and systemic tissues, with the highest frequency in PerC, suggesting a local role. Inactivated SAV (InSAV1) induced comparatively lower ASC responses in all sites, and specific serum antibodies were only induced by WtSAV3 and not by InSAV1. An InSAV1 boost did not increase these responses. Expression of immune marker genes implies a role for PerC adipose tissue in the PerC immune response. Overall, the study suggests the Atlantic salmon PerC as a secondary immune site and an ASC survival niche.


Subject(s)
Alphavirus Infections , Alphavirus , Antibodies, Viral , Antibody-Producing Cells , Fish Diseases , Peritoneal Cavity , Salmo salar , Animals , Salmo salar/immunology , Salmo salar/virology , Alphavirus/immunology , Alphavirus Infections/immunology , Alphavirus Infections/veterinary , Alphavirus Infections/virology , Peritoneal Cavity/cytology , Fish Diseases/immunology , Fish Diseases/virology , Antibody-Producing Cells/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Injections, Intraperitoneal/veterinary
3.
Viruses ; 16(4)2024 03 25.
Article in English | MEDLINE | ID: mdl-38675846

ABSTRACT

Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the presence of RNA-amplifying enzymes known as replicases. We recently demonstrated that this property could enhance immune responses with minimal injected RNA amounts. In saRNA-based vaccines, replicase and antigens are encoded on the same mRNA molecule, resulting in very long RNA sequences, which poses significant challenges in production, delivery, and stability. In taRNA-based vaccines, these challenges can be overcome by splitting the replication system into two parts: one that encodes replicase and the other that encodes a short antigen-encoding RNA called transreplicon. Here, we review the identification and use of transreplicon RNA in alphavirus research, with a focus on the development of novel taRNA technology as a state-of-the art vaccine platform. Additionally, we discuss remaining challenges essential to the clinical application and highlight the potential benefits related to the unique properties of this future vaccine platform.


Subject(s)
Alphavirus , RNA, Viral , Alphavirus/genetics , Alphavirus/immunology , RNA, Viral/genetics , Animals , Humans , Viral Vaccines/immunology , Viral Vaccines/genetics , Virus Replication , Alphavirus Infections/virology , Alphavirus Infections/prevention & control , Alphavirus Infections/immunology , Vaccine Development
4.
J Virol ; 98(1): e0110223, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38169294

ABSTRACT

Mayaro virus (MAYV) is an emerging arbovirus member of the Togaviridae family and Alphavirus genus. MAYV infection causes an acute febrile illness accompanied by persistent polyarthralgia and myalgia. Understanding the mechanisms involved in arthritis caused by alphaviruses is necessary to develop specific therapies. In this work, we investigated the role of the CCL2/CCR2 axis in the pathogenesis of MAYV-induced disease. For this, wild-type (WT) C57BL/6J and CCR2-/- mice were infected with MAYV subcutaneously and evaluated for disease development. MAYV infection induced an acute inflammatory disease in WT mice. The immune response profile was characterized by an increase in the production of inflammatory mediators, such as IL-6, TNF, and CCL2. Higher levels of CCL2 at the local and systemic levels were followed by the significant recruitment of CCR2+ macrophages and a cellular response orchestrated by these cells. CCR2-/- mice showed an increase in CXCL-1 levels, followed by a replacement of the macrophage inflammatory infiltrate by neutrophils. Additionally, the absence of the CCR2 receptor protected mice from bone loss induced by MAYV. Accordingly, the silencing of CCL2 chemokine expression in vivo and the pharmacological blockade of CCR2 promoted a partial improvement in disease. Cell culture data support the mechanism underlying the bone pathology of MAYV, in which MAYV infection promotes a pro-osteoclastogenic microenvironment mediated by CCL2, IL-6, and TNF, which induces the migration and differentiation of osteoclast precursor cells. Overall, these data contribute to the understanding of the pathophysiology of MAYV infection and the identification future of specific therapeutic targets in MAYV-induced disease.IMPORTANCEThis work demonstrates the role of the CCL2/CCR2 axis in MAYV-induced disease. The infection of wild-type (WT) C57BL/6J and CCR2-/- mice was associated with high levels of CCL2, an important chemoattractant involved in the recruitment of macrophages, the main precursor of osteoclasts. In the absence of the CCR2 receptor, there is a mitigation of macrophage migration to the target organs of infection and protection of these mice against bone loss induced by MAYV infection. Much evidence has shown that host immune response factors contribute significantly to the tissue damage associated with alphavirus infections. Thus, this work highlights molecular and cellular targets involved in the pathogenesis of arthritis triggered by MAYV and identifies novel therapeutic possibilities directed to the host inflammatory response unleashed by MAYV.


Subject(s)
Alphavirus Infections , Arthritis , Chemokine CCL2 , Receptors, CCR2 , Animals , Mice , Alphavirus , Alphavirus Infections/immunology , Arthritis/immunology , Arthritis/virology , Chemokine CCL2/immunology , Interleukin-6/immunology , Mice, Inbred C57BL , Receptors, CCR2/immunology , Mice, Knockout , Male , Bone Diseases/virology
5.
J Virol ; 97(10): e0095923, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37772825

ABSTRACT

IMPORTANCE: Viral encephalomyelitis outcome is dependent on host responses to neuronal infection. Interferon (IFN) is an important component of the innate response, and IFN regulatory factor (IRF) 7 is an inducible transcription factor for the synthesis of IFN-α. IRF7-deficient mice develop fatal paralysis after CNS infection with Sindbis virus, while wild-type mice recover. Irf7 -/- mice produce low levels of IFN-α but high levels of IFN-ß with induction of IFN-stimulated genes, so the reason for this difference is not understood. The current study shows that Irf7 -/- mice developed inflammation earlier but failed to clear virus from motor neuron-rich regions of the brainstem and spinal cord. Levels of IFN-γ and virus-specific antibody were comparable, indicating that IRF7 deficiency does not impair expression of these known viral clearance factors. Therefore, IRF7 is either necessary for the neuronal response to currently identified mediators of clearance or enables the production of additional antiviral factor(s) needed for clearance.


Subject(s)
Alphavirus Infections , Encephalomyelitis , Interferon Regulatory Factor-7 , Sindbis Virus , Animals , Mice , Alphavirus Infections/immunology , Alphavirus Infections/virology , Brain Stem/virology , Encephalomyelitis/immunology , Encephalomyelitis/virology , Inflammation/virology , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Motor Neurons/virology , Sindbis Virus/immunology , Spinal Cord/virology
6.
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
7.
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
8.
PLoS Pathog ; 18(1): e1010202, 2022 01.
Article in English | MEDLINE | ID: mdl-34990484

ABSTRACT

The exogenous small interfering RNA (exo-siRNA) pathway is a key antiviral mechanism in the Aedes aegypti mosquito, a widely distributed vector of human-pathogenic arboviruses. This pathway is induced by virus-derived double-stranded RNAs (dsRNA) that are cleaved by the ribonuclease Dicer 2 (Dcr2) into predominantly 21 nucleotide (nt) virus-derived small interfering RNAs (vsiRNAs). These vsiRNAs are used by the effector protein Argonaute 2 within the RNA-induced silencing complex to cleave target viral RNA. Dcr2 contains several domains crucial for its activities, including helicase and RNase III domains. In Drosophila melanogaster Dcr2, the helicase domain has been associated with binding to dsRNA with blunt-ended termini and a processive siRNA production mechanism, while the platform-PAZ domains bind dsRNA with 3' overhangs and subsequent distributive siRNA production. Here we analyzed the contributions of the helicase and RNase III domains in Ae. aegypti Dcr2 to antiviral activity and to the exo-siRNA pathway. Conserved amino acids in the helicase and RNase III domains were identified to investigate Dcr2 antiviral activity in an Ae. aegypti-derived Dcr2 knockout cell line by reporter assays and infection with mosquito-borne Semliki Forest virus (Togaviridae, Alphavirus). Functionally relevant amino acids were found to be conserved in haplotype Dcr2 sequences from field-derived Ae. aegypti across different continents. The helicase and RNase III domains were critical for silencing activity and 21 nt vsiRNA production, with RNase III domain activity alone determined to be insufficient for antiviral activity. Analysis of 21 nt vsiRNA sequences (produced by functional Dcr2) to assess the distribution and phasing along the viral genome revealed diverse yet highly consistent vsiRNA pools, with predominantly short or long sequence overlaps including 19 nt overlaps (the latter representing most likely true Dcr2 cleavage products). Combined with the importance of the Dcr2 helicase domain, this suggests that the majority of 21 nt vsiRNAs originate by processive cleavage. This study sheds new light on Ae. aegypti Dcr2 functions and properties in this important arbovirus vector species.


Subject(s)
Aedes/immunology , Aedes/virology , Alphavirus Infections/immunology , Ribonuclease III/immunology , Aedes/genetics , Animals , DNA Mutational Analysis , Mosquito Vectors/virology , RNA, Small Interfering/immunology , RNA, Viral/immunology , Ribonuclease III/genetics , Semliki forest virus
9.
Virology ; 565: 13-21, 2022 01 02.
Article in English | MEDLINE | ID: mdl-34626907

ABSTRACT

Eastern equine encephalitis virus (EEEV), western equine encephalitis virus (WEEV) and Venezuelan equine encephalitis virus (VEEV) can cause fatal encephalitis in humans and equids. Some MAbs to the E1 glycoprotein are known to be cross-reactive, weakly neutralizing in vitro but can protect from disease in animal models. We investigated the mechanism of neutralization of VEEV infection by the broadly cross-reactive E1-specific MAb 1A4B-6. 1A4B-6 protected 3-week-old Swiss Webster mice prophylactically from lethal VEEV challenge. Likewise, 1A4B-6 inhibited virus growth in vitro at a pre-attachment step after virions were incubated at 37 °C and inhibited virus-mediated cell fusion. Amino acid residue N100 in the fusion loop of E1 protein was identified as critical for binding. The potential to elicit broadly cross-reactive MAbs with limited virus neutralizing activity in vitro but that can inhibit virus entry and protect animals from infection merits further exploration for vaccine and therapeutic developmental research.


Subject(s)
Antibodies, Viral/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/virology , Viral Envelope Proteins/immunology , Virus Replication/drug effects , Alphavirus/immunology , Alphavirus Infections/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cell Line , Chlorocebus aethiops , Cross Reactions , Encephalomyelitis, Venezuelan Equine/therapy , Glycoproteins/immunology , Immunotherapy , Mice , Protein Binding , Vero Cells , Viral Envelope Proteins/metabolism , Virion/immunology , Virion/metabolism
10.
Cell Rep ; 37(13): 110150, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965418

ABSTRACT

Enteric pathogens overcome barrier immunity within the intestinal environment that includes the endogenous flora. The microbiota produces diverse ligands, and the full spectrum of microbial products that are sensed by the epithelium and prime protective immunity is unknown. Using Drosophila, we find that the gut presents a high barrier to infection, which is partially due to signals from the microbiota, as loss of the microbiota enhances oral viral infection. We report cyclic dinucleotide (CDN) feeding is sufficient to protect microbiota-deficient flies from enhanced oral infection, suggesting that bacterial-derived CDNs induce immunity. Mechanistically, we find CDN protection is dSTING- and dTBK1-dependent, leading to NF-kB-dependent gene expression. Furthermore, we identify the apical nucleoside transporter, CNT2, as required for oral CDN protection. Altogether, our studies define a role for bacterial products in priming immune defenses in the gut.


Subject(s)
Alphavirus Infections/immunology , Antiviral Agents/pharmacology , Drosophila melanogaster/immunology , Enterocytes/immunology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Nucleotides, Cyclic/administration & dosage , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Animals , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enterocytes/drug effects , Enterocytes/virology , Female , Immunity, Innate , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sindbis Virus/immunology
11.
Am J Trop Med Hyg ; 106(2): 607-609, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34844213

ABSTRACT

Mayaro virus (MAYV) is an alphavirus endemic to both Latin America and the Caribbean. Recent reports have questioned the ability of MAYV and its close relative, Chikungunya virus (CHIKV), to generate cross-reactive, neutralizing antibodies to one another. Since CHIKV was introduced to South America in 2013, discerning whether individuals have cross-reactive antibodies or whether they have had exposures to both viruses previously has been difficult. Using samples obtained from people infected with MAYV prior to the introduction of CHIKV in the Americas, we performed neutralizing assays and observed no discernable neutralization of CHIKV by sera from patients previously infected with MAYV. These data suggest that a positive CHIKV neutralization test cannot be attributed to prior exposure to MAYV and that previous exposure to MAYV may not be protective against a subsequent CHIKV infection.


Subject(s)
Alphavirus Infections/diagnosis , Alphavirus Infections/epidemiology , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chikungunya Fever/diagnosis , Chikungunya Fever/epidemiology , Alphavirus/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Cross Reactions , Humans , Immune Sera/chemistry , Neutralization Tests , Peru/epidemiology
12.
J Virol ; 95(23): e0112221, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34549980

ABSTRACT

Chikungunya virus (CHIKV) and Mayaro virus (MAYV) are closely related members of the Semliki Forest virus antigenic complex classified as belonging to the genus Alphavirus of the family Togaviridae. These viruses cause human disease, with sudden fever and joint inflammation that can persist for long periods. CHIKV is the causative agent of large outbreaks worldwide, and MAYV infection represents a growing public health concern in Latin America, causing sporadic cases and geographically limited outbreaks. Considering the relationship between CHIKV and MAYV, the present study aimed to evaluate if preexisting CHIKV immunity protects against MAYV infection. Immunocompetent C57BL/6 mice were intraperitoneally infected with CHIKV and, 4 weeks later, they were infected with MAYV in their hind paw. We observed that the preexistence of CHIKV immunity conferred partial cross-protection against secondary MAYV infection, reducing disease severity, tissue viral load, and histopathological scores. Interestingly, CHIKV antibodies from humans and mice showed low cross-neutralization to MAYV, but neutralizing activity significantly increased after secondary infection. Furthermore, depletion of adaptive immune cells (CD4+ T, CD8+ T, and CD19+ B cells) did not alter the cross-protection phenotype, suggesting that distinct cell subsets or a combination of adaptive immune cells stimulated by CHIKV are responsible for the partial cross-protection against MAYV. The reduction of proinflammatory cytokines, such as interferon gamma (IFN-γ), in animals secondarily infected by MAYV, suggests a role for innate immunity in cross-protection. Our findings shed light on how preexisting immunity to arthritogenic alphaviruses may affect secondary infection, which may further develop relevant influence in disease outcome and viral transmission. IMPORTANCE Mosquito-borne viruses have a worldwide impact, especially in tropical climates. Chikungunya virus has been present mostly in developing countries, causing millions of infections, while Mayaro virus, a close relative, has been limited to the Caribbean and tropical regions of Latin America. The potential emergence and spread of Mayaro virus to other high-risk areas have increased the scientific community's attention to an imminent worldwide epidemic. Here, we designed an experimental protocol of chikungunya and Mayaro virus mouse infection, which develops a measurable and quantifiable disease that allows us to make inferences about potential immunological effects during secondary virus infection. Our results demonstrate that previous chikungunya virus infection is able to reduce the severity of clinical outcomes during secondary Mayaro infection. We provide scientific understanding of immunological features during secondary infection with the closely related virus, thus assisting in better comprehending viral transmission and the pathological outcome of these diseases.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Chikungunya virus/immunology , Cross Protection/immunology , Alphavirus/immunology , Alphavirus Infections/pathology , Animals , Antibodies, Viral/immunology , Chikungunya Fever/virology , Disease Models, Animal , Epidemics , Female , Inflammation , Mice , Mice, Inbred C57BL , Viral Load
13.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507983

ABSTRACT

Arthritogenic alphaviruses are globally distributed, mosquito-transmitted viruses that cause rheumatological disease in humans and include Chikungunya virus (CHIKV), Mayaro virus (MAYV), and others. Although serological evidence suggests that some antibody-mediated heterologous immunity may be afforded by alphavirus infection, the extent to which broadly neutralizing antibodies that protect against multiple arthritogenic alphaviruses are elicited during natural infection remains unknown. Here, we describe the isolation and characterization of MAYV-reactive alphavirus monoclonal antibodies (mAbs) from a CHIKV-convalescent donor. We characterized 33 human mAbs that cross-reacted with CHIKV and MAYV and engaged multiple epitopes on the E1 and E2 glycoproteins. We identified five mAbs that target distinct regions of the B domain of E2 and potently neutralize multiple alphaviruses with differential breadth of inhibition. These broadly neutralizing mAbs (bNAbs) contain few somatic mutations and inferred germline-revertants retained neutralizing capacity. Two bNAbs, DC2.M16 and DC2.M357, protected against both CHIKV- and MAYV-induced musculoskeletal disease in mice. These findings enhance our understanding of the cross-reactive and cross-protective antibody response to human alphavirus infections.


Subject(s)
Alphavirus Infections/immunology , Antibodies, Monoclonal/isolation & purification , Broadly Neutralizing Antibodies/immunology , Alphavirus/immunology , Alphavirus/pathogenicity , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Arthritis/etiology , Arthritis/immunology , Arthritis/virology , Broadly Neutralizing Antibodies/isolation & purification , Broadly Neutralizing Antibodies/pharmacology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Cross Reactions , Epitopes/immunology , Germ Cells/immunology , Glycoproteins/immunology , Humans , Male , Mice , Mice, Inbred C57BL
14.
Cell ; 184(17): 4414-4429.e19, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34416146

ABSTRACT

Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.


Subject(s)
Alphavirus/immunology , Antibodies, Viral/immunology , Conserved Sequence/immunology , Epitopes/immunology , Viral Proteins/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chlorocebus aethiops , Epitope Mapping , Epitopes/chemistry , Humans , Male , Mice, Inbred C57BL , Models, Biological , Monocytes/metabolism , Vero Cells , Viral Proteins/chemistry , Virus Release
15.
J Fish Dis ; 44(12): 1911-1924, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34402092

ABSTRACT

Pancreas disease (PD) caused by salmonid alphavirus (SAV) continues to negatively impact salmon farming. To assess the effect on growth and mortality of three vaccines against PD, two controlled field designs were employed: one controlled field study with individual marked fish (PIT tag) assessing three PD vaccines and three controls groups, and a second controlled field study with group marked fish (Maxilla) comparing two PD vaccines against controls. In addition, a descriptive study using whole cages compared fish immunized with two different PD vaccines against controls. The target populations experienced a natural PD outbreak where both SAV 2 and SAV 3 were identified. Only one of the PD vaccines provided statistically significant improvements in harvest weight of 0.43 kg (CI: 0.29-0.57) and 0.51 kg (CI: 0.36-0.65) compared with the control in the PIT tag and the Maxilla study, respectively. In the latter, a significant reduction in mortality of 1.31 (CI:0.8-1.8) per cent points was registered for the same vaccine compared with controls. These results aligned with the growth and PD-specific mortality registered in the descriptive Cage study. The data in this study show a difference in the efficacy of PD vaccines in farmed Atlantic salmon.


Subject(s)
Alphavirus Infections/veterinary , Fish Diseases/virology , Pancreatic Diseases/veterinary , Viral Vaccines/pharmacology , Alphavirus/drug effects , Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Animals , Aquaculture , Fish Diseases/immunology , Fish Diseases/prevention & control , Pancreatic Diseases/prevention & control , Pancreatic Diseases/virology , Salmo salar , Vaccines, Inactivated/pharmacology
16.
Viruses ; 13(7)2021 06 22.
Article in English | MEDLINE | ID: mdl-34206519

ABSTRACT

Alphaviruses have a single-stranded, positive-sense RNA genome that contains two open reading frames encoding either the non-structural or the structural genes. Upon infection, the genomic RNA is translated into the non-structural proteins (nsPs). NsPs are required for viral RNA replication and transcription driven from the subgenomic promoter (sgP). Transfection of an RNA encoding the luciferase gene under the control of the sgP into cells enabled the detection of replication-competent chikungunya virus (CHIKV) or Mayaro virus (MAYV) with high sensitivity as a function of the induced luciferase activity. This assay principle was additionally used to analyze virus-neutralizing antibodies in sera and might be an alternative to standard virus neutralization assays based on virus titration or the use of genetically modified tagged viruses.


Subject(s)
Alphavirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , RNA, Viral/genetics , Serologic Tests/methods , Alphavirus/classification , Alphavirus Infections/blood , Alphavirus Infections/diagnosis , Alphavirus Infections/immunology , Animals , Cell Line , Chikungunya virus/genetics , Cross Reactions , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Luciferases/genetics , Mice , Mice, Inbred BALB C , Sensitivity and Specificity , Serologic Tests/standards
17.
Sci Rep ; 11(1): 15374, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321560

ABSTRACT

Mayaro virus (MAYV), which causes mayaro fever, is endemic to limited regions of South America that may expand due to the possible involvement of Aedes spp. mosquitoes in its transmission. Its effective control will require the accurate identification of infected individuals, which has been restricted to nucleic acid-based tests due to similarities with other emerging members of the Alphavirus genus of the Togaviridae family; both in structure and clinical symptoms. Serological tests have a more significant potential to expand testing at a reasonable cost, and their performance primarily reflects that of the antigen utilized to capture pathogen-specific antibodies. Here, we describe the assembly of a synthetic gene encoding multiple copies of antigenic determinants mapped from the nsP1, nsP2, E1, and E2 proteins of MAYV that readily expressed as a stable chimeric protein in bacteria. Its serological performance as the target in ELISAs revealed a high accuracy for detecting anti-MAYV IgM antibodies. No cross-reactivity was observed with serum from seropositive individuals for dengue, chikungunya, yellow fever, Zika, and other infectious diseases as well as healthy individuals. Our data suggest that this bioengineered antigen could be used to develop high-performance serological tests for MAYV infections.


Subject(s)
Alphavirus Infections/diagnosis , Alphavirus/immunology , Epitopes/immunology , Togaviridae Infections/diagnosis , Aedes/virology , Alphavirus/pathogenicity , Alphavirus Infections/immunology , Alphavirus Infections/transmission , Alphavirus Infections/virology , Animals , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Epitopes/ultrastructure , Female , Genes, Synthetic/genetics , Genes, Synthetic/immunology , Humans , Immunoglobulin M/immunology , Male , Serologic Tests , South America/epidemiology , Togaviridae/isolation & purification , Togaviridae/pathogenicity , Togaviridae Infections/immunology , Togaviridae Infections/transmission , Togaviridae Infections/virology
18.
Nat Commun ; 12(1): 3038, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031424

ABSTRACT

Mayaro virus (MAYV) is an emerging arbovirus of the Americas that may cause a debilitating arthritogenic disease. The biology of MAYV is not fully understood and largely inferred from related arthritogenic alphaviruses. Here, we present the structure of MAYV at 4.4 Å resolution, obtained from a preparation of mature, infective virions. MAYV presents typical alphavirus features and organization. Interactions between viral proteins that lead to particle formation are described together with a hydrophobic pocket formed between E1 and E2 spike proteins and conformational epitopes specific of MAYV. We also describe MAYV glycosylation residues in E1 and E2 that may affect MXRA8 host receptor binding, and a molecular "handshake" between MAYV spikes formed by N262 glycosylation in adjacent E2 proteins. The structure of MAYV is suggestive of structural and functional complexity among alphaviruses, which may be targeted for specificity or antiviral activity.


Subject(s)
Alphavirus Infections/virology , Alphavirus/ultrastructure , Cryoelectron Microscopy , Mass Spectrometry , Alphavirus/immunology , Alphavirus Infections/immunology , Animals , Antibodies, Neutralizing , Chlorocebus aethiops , Glycosylation , Humans , Immunoglobulins , Membrane Proteins , Vero Cells
19.
Viruses ; 13(3)2021 03 11.
Article in English | MEDLINE | ID: mdl-33799906

ABSTRACT

Mayaro virus (MAYV) and chikungunya virus (CHIKV) are known for their arthrotropism, but accumulating evidence shows that CHIKV infections are occasionally associated with serious neurological complications. However, little is known about the capacity of MAYV to invade the central nervous system (CNS). We show that human neural progenitors (hNPCs), pericytes and astrocytes are susceptible to MAYV infection, resulting in the production of infectious viral particles. In primary astrocytes, MAYV, and to a lesser extent CHIKV, elicited a strong antiviral response, as demonstrated by an increased expression of several interferon-stimulated genes, including ISG15, MX1 and OAS2. Infection with either virus led to an enhanced expression of inflammatory chemokines, such as CCL5, CXCL10 and CXCL11, whereas MAYV induced higher levels of IL-6, IL-12 and IL-15 in these cells. Moreover, MAYV was more susceptible than CHIKV to the antiviral effects of both type I and type II interferons. Taken together, this study shows that although MAYV and CHIKV are phylogenetically related, they induce different types of antiviral responses in astrocytes. This work is the first to evaluate the potential neurotropism of MAYV and shows that brain cells and particularly astrocytes and hNPCs are permissive to MAYV, which, consequently, could lead to MAYV-induced neuropathology.


Subject(s)
Alphavirus Infections/immunology , Alphavirus/immunology , Astrocytes/immunology , Astrocytes/virology , Brain/immunology , 2',5'-Oligoadenylate Synthetase/metabolism , Alphavirus Infections/pathology , Animals , Brain/virology , Cell Line , Chemokine CCL5/metabolism , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chikungunya Fever/immunology , Chikungunya virus/immunology , Chlorocebus aethiops , Cytokines/metabolism , Humans , Interferon Type I/immunology , Interferon-gamma/immunology , Myxovirus Resistance Proteins/metabolism , Neural Stem Cells/virology , Pericytes/virology , Ubiquitins/metabolism , Vero Cells
20.
Cell Rep ; 35(1): 108962, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33826892

ABSTRACT

Although neutralizing monoclonal antibodies (mAbs) against epitopes within the alphavirus E2 protein can protect against infection, the functional significance of non-neutralizing mAbs is poorly understood. Here, we evaluate the activity of 13 non-neutralizing mAbs against Mayaro virus (MAYV), an emerging arthritogenic alphavirus. These mAbs bind to the MAYV virion and surface of infected cells but fail to neutralize infection in cell culture. Mapping studies identify six mAb binding groups that localize to discrete epitopes within or adjacent to the A domain of the E2 glycoprotein. Remarkably, passive transfer of non-neutralizing mAbs protects against MAYV infection and disease in mice, and their efficacy requires Fc effector functions. Monocytes mediate the protection of non-neutralizing mAbs in vivo, as Fcγ-receptor-expressing myeloid cells facilitate the binding, uptake, and clearance of MAYV without antibody-dependent enhancement of infection. Humoral protection against alphaviruses likely reflects contributions from non-neutralizing antibodies through Fc-dependent mechanisms that accelerate viral clearance.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Alphavirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Animals , Antibodies, Monoclonal/immunology , Epitope Mapping , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Monocytes/metabolism , Musculoskeletal Diseases/immunology , Musculoskeletal Diseases/virology , Myeloid Cells/metabolism , Receptors, IgG/metabolism , Recombinant Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...