Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
1.
Viruses ; 16(4)2024 Mar 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
2.
Microbiol Spectr ; 11(1): e0361322, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36511715

ABSTRACT

Most alphaviruses are transmitted by mosquito vectors and infect a wide range of vertebrate hosts, with a few exceptions. Eilat virus (EILV) in this genus is characterized by a host range restricted to mosquitoes. Its chimeric viruses have been developed as safe and effective vaccine candidates and diagnostic tools. Here, we investigated the interactions between these insect-specific viruses (ISVs) and mosquito cells, unveiling their potential roles in determining vector competence and arbovirus transmission. By RNA sequencing, we found that these ISVs profoundly modified host cell gene expression profiles. Two EILV-based chimeras, consisting of EILV's nonstructural genes and the structural genes of Chikungunya virus (CHIKV) or Venezuelan equine encephalitis virus (VEEV), namely, EILV/CHIKV (E/C) and EILV/VEEV (E/V), induced more intensive transcriptome regulation than parental EILV and activated different antiviral mechanisms in host cells. We demonstrated that E/C robustly promoted antimicrobial peptide production and E/V strongly upregulated the RNA interference pathway components. This also highlighted the intrinsic divergences between CHIKV and VEEV, representatives of the Old World and New World alphaviruses. In contrast, EILV triggered a limited antiviral response. We further showed that initial chimera infections efficiently inhibited subsequent pathogenic alphavirus replication, especially in the case of E/V infection, which almost prevented VEEV and Sindbis virus (SINV) superinfections. Altogether our study provided valuable information on developing ISVs as biological control agents. IMPORTANCE Mosquito-borne alphaviruses can cause emerging and reemerging infectious diseases, posing a considerable threat to human and animal health worldwide. However, no specific antivirals or commercial vaccines are currently available. Therefore, it is vital to develop biological control measures to contain virus transmission. Insect-specific EILV and its chimeras are supposed to induce superinfection exclusion owing to the close phylogenetical relationship with pathogenic alphaviruses. These viruses might also, like bacterial symbionts, modulate mosquito hosts' vector competence for arboviruses. However, little is known about the responses of mosquitoes or mosquito cells to ISV infections. Here, we found that EILV barely elicited antiviral defenses in host cells, while its chimeras, namely, E/C and E/V, potentiated the responses via different mechanisms. Furthermore, we showed that initial chimera infections could largely inhibit subsequent pathogenic alphavirus infections. Taken together, our study proposed insect-specific chimeras as a promising candidate for developing biological control measures against pathogenic alphaviruses.


Subject(s)
Alphavirus Infections , Alphavirus , Culicidae , Insect Viruses , Animals , Alphavirus/genetics , Alphavirus Infections/prevention & control
3.
Nat Rev Microbiol ; 21(6): 396-407, 2023 06.
Article in English | MEDLINE | ID: mdl-36474012

ABSTRACT

Alphaviruses are arthropod-transmitted RNA viruses that cause epidemics of human infection and disease on a global scale. These viruses are classified as either arthritogenic or encephalitic based on their genetic relatedness and the clinical syndromes they cause. Although there are currently no approved therapeutics or vaccines against alphaviruses, passive transfer of monoclonal antibodies confers protection in animal models. This Review highlights recent advances in our understanding of the host factors required for alphavirus entry, the mechanisms of action by which protective antibodies inhibit different steps in the alphavirus infection cycle and candidate alphavirus vaccines currently under clinical evaluation that focus on humoral immunity. A comprehensive understanding of alphavirus entry and antibody-mediated protection may inform the development of new classes of countermeasures for these emerging viruses.


Subject(s)
Alphavirus Infections , Alphavirus , Animals , Humans , Alphavirus/genetics , Alphavirus Infections/prevention & control , Antibodies, Monoclonal
4.
Int J Mol Sci ; 23(8)2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35456923

ABSTRACT

Mayaro virus (MAYV) is an emerging alphavirus causing acute febrile illness associated with chronic polyarthralgia. Although MAYV is currently restricted to tropical regions in South America around the Amazon basin, it has the potential to spread globally by Aedes species mosquitoes. In addition, there are currently no specific therapeutics or licenced vaccines against MAYV infection. We have previously shown that an adenovirus based Mayaro vaccine (ChAdOx1 May) was able to provide full protection against MAYV challenge in vaccinated A129 mice and induced high neutralising antibody titres. In this study, we have constructed a replication deficient simian adenovirus (ChAdOx2) and a Modified Ankara Virus (MVA) based vaccine expressing the MAYV structural cassette (sMAYV) similar to ChAdOx1 May, and characterised recombinant MAYV E2 glycoprotein expressed in a mammalian system for immune monitoring. We demonstrate that ChAdOx2 May was able to induce high antibody titres similar to ChAdOx1 May, and MVA May was shown to be an effective boosting strategy following prime vaccination with ChAdOx1 or ChAdOx2 May. In order to measure MAYV neutralising ability, we have developed a virus replicon particle-based neutralisation assay which effectively detected neutralising antibodies against MAYV. In summary, our study indicates the potential for further clinical development of the viral vectored MAYV vaccines against MAYV infections.


Subject(s)
Alphavirus Infections , Chikungunya virus , Viral Vaccines , Alphavirus Infections/prevention & control , Animals , Antibodies, Viral , Mammals , Mice , Replicon , Viral Vaccines/genetics
5.
Methods Mol Biol ; 2411: 205-218, 2022.
Article in English | MEDLINE | ID: mdl-34816407

ABSTRACT

Despite vaccination, pancreas disease (PD) caused by salmonid alphavirus (SAV) has been the economically most important virus disease in salmon farming in Ireland, Scotland, and Norway. A vaccine based on DNA plasmid has been authorized to be used in Norwegian aquaculture since 2018. DNA vaccination of plasmids expressed subcellular viral proteins have been shown its particular protective effect against SAV3 that surface expression of the E2 protein with the whole viral protein construct, yielding a more effective vaccine. The chapter describes methods to design and test the sublocalization of expressed viral protein and the performance evaluation of vaccines against SAV3 infection in Atlantic salmon.


Subject(s)
Alphavirus Infections , Alphavirus , Fish Diseases , Salmonidae , Vaccines, DNA , Alphavirus/genetics , Alphavirus/immunology , Alphavirus Infections/prevention & control , Alphavirus Infections/veterinary , Animals , Fish Diseases/prevention & control , Viral Proteins
6.
Epidemics ; 37: 100502, 2021 12.
Article in English | MEDLINE | ID: mdl-34610550

ABSTRACT

Pancreas Disease (PD) is a viral disease caused by Salmonid Alphavirus (SAV). It affects farmed salmonids in the North Atlantic, and leads to reduced feed intake and increased mortality with reduced production and welfare as a consequence. In 2013, the estimated cost of an outbreak on an average salmon farm was about 6.6 mil €. In Norway, PD has been notifiable since 2008, and regulations to mitigate disease spread are in place. However, despite the regulations, 140-170 farms are affected by PD every year. The aquaculture industry is growing continuously, introducing farms in new geographical areas, and fish are moved between hydrographically separated zones for trade and slaughter. All such movements and relocations need to be approved by the competent authorities. Thus, there is a demand for support to farmers and competent authorities when making decisions on disease management and especially on the effect of moving infected fish. We have used a disease-transmission model for outbreak-simulation in real time for assessing the probability of disease transmission from a farm that gets infected with PD. We have also simulated the effects of three different control-regimes: no stamping-out, delayed stamping-out or immediate stamping-out, on the transmission of PD to surrounding farms. Simulations showed that the immediate stamping out of an infected farm led to effective containment of an outbreak. No stamping out led to up to 32.1% of farms within 100 km of the index farm to become effected. We have used real production data for the model building and the scenario simulations, and the results illustrate that a risk assessment of horizontal disease transmission must be undertaken on a case-by-case basis, because the time and place of the outbreak has a large influence on the risk of transmission.


Subject(s)
Alphavirus Infections , Fish Diseases , Pancreatic Diseases , Salmonidae , Alphavirus Infections/epidemiology , Alphavirus Infections/prevention & control , Alphavirus Infections/veterinary , Animals , Aquaculture , Fish Diseases/epidemiology , Norway/epidemiology , Pancreas , Pancreatic Diseases/epidemiology , Pancreatic Diseases/veterinary
7.
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
8.
J Gen Virol ; 102(8)2021 08.
Article in English | MEDLINE | ID: mdl-34435944

ABSTRACT

Human pathogens belonging to the Alphavirus genus, in the Togaviridae family, are transmitted primarily by mosquitoes. The signs and symptoms associated with these viruses include fever and polyarthralgia, defined as joint pain and inflammation, as well as encephalitis. In the last decade, our understanding of the interactions between members of the alphavirus genus and the human host has increased due to the re-appearance of the chikungunya virus (CHIKV) in Asia and Europe, as well as its emergence in the Americas. Alphaviruses affect host immunity through cytokines and the interferon response. Understanding alphavirus interactions with both the innate immune system as well as the various cells in the adaptive immune systems is critical to developing effective therapeutics. In this review, we summarize the latest research on alphavirus-host cell interactions, underlying infection mechanisms, and possible treatments.


Subject(s)
Alphavirus Infections , Alphavirus , Alphavirus/immunology , Alphavirus/pathogenicity , Alphavirus Infections/epidemiology , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Humans , Viral Vaccines/immunology
9.
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
10.
Fish Shellfish Immunol ; 118: 188-196, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34252544

ABSTRACT

Before seawater transfer, farmed Atlantic salmon are subjected to treatments that may affect the immune system and susceptibility to pathogens. E.g., exposure to constant light (CL) stimulates smoltification, which prepares salmon to life in sea water, but endocrine changes in this period are associated with suppression of immune genes. Salmon are vaccinated towards end of the freshwater period to safeguard that adequate vaccine efficacy is achieved by the time the fish is transferred to sea. In the present study, we investigated how the responses to vaccination and viral infection varied depending on the time of CL onset relative to vaccination. The salmon were either exposed to CL two weeks prior to vaccination (2-PRI) or exposed to CL at the time of vaccination (0-PRI). A cohabitant challenge with salmonid alphavirus, the causative agent of pancreatic disease, was performed 9 weeks post vaccination. The immunological effects of the different light manipulation were examined at 0- and 6-weeks post vaccination, and 6 weeks post challenge. Antibody levels in serum were measured using a serological bead-based multiplex panel as well as ELISA, and 92 immune genes in heart and spleen were measured using an integrated fluidic circuit-based qPCR array for multiple gene expression. The 2-PRI group showed a moderate transcript down-regulation of genes in the heart at the time of vaccination, which were restored 6 weeks after vaccination (WPV). Conversely, at 6WPV a down-regulation was seen for the 0-PRI fish. Moreover, the 2-PRI group had significantly higher levels of antibodies binding to three of the vaccine components at 6WPV, compared to 0-PRI. In response to SAV challenge, transcription of immune genes between 2-PRI and 0-PRI was markedly dissimilar in the heart and spleen of control fish, but no difference was found between vaccinated salmon from the two CL regimens. Thus, by using labor-saving high throughput detection methods, we demonstrated that light regimens affected antibody production and transcription of immune genes in non-vaccinated and virus challenged salmon, but the differences between the light treatment groups appeared eliminated by vaccination.


Subject(s)
Alphavirus Infections , Alphavirus , Fish Diseases , Salmo salar , Alphavirus Infections/prevention & control , Alphavirus Infections/veterinary , Animals , Fish Diseases/virology , Gene Expression , Salmo salar/virology , Vaccination/veterinary , Vaccine Efficacy
11.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34078739

ABSTRACT

Interferon-induced transmembrane (IFITM) proteins restrict membrane fusion and virion internalization of several enveloped viruses. The role of IFITM proteins during alphaviral infection of human cells and viral counteraction strategies are insufficiently understood. Here, we characterized the impact of human IFITMs on the entry and spread of chikungunya virus and Mayaro virus and provide first evidence for a CHIKV-mediated antagonism of IFITMs. IFITM1, 2, and 3 restricted infection at the level of alphavirus glycoprotein-mediated entry, both in the context of direct infection and cell-to-cell transmission. Relocalization of normally endosomal IFITM3 to the plasma membrane resulted in loss of antiviral activity. rs12252-C, a naturally occurring variant of IFITM3 that may associate with severe influenza in humans, restricted CHIKV, MAYV, and influenza A virus infection as efficiently as wild-type IFITM3 Antivirally active IFITM variants displayed reduced cell surface levels in CHIKV-infected cells involving a posttranscriptional process mediated by one or several nonstructural protein(s) of CHIKV. Finally, IFITM3-imposed reduction of specific infectivity of nascent particles provides a rationale for the necessity of a virus-encoded counteraction strategy against this restriction factor.


Subject(s)
Alphavirus Infections/prevention & control , Chikungunya Fever/prevention & control , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Alphavirus/pathogenicity , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Chikungunya Fever/metabolism , Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Endosomes/metabolism , Humans , Membrane Proteins/physiology , RNA-Binding Proteins/physiology , Virus Internalization
12.
Virology ; 561: 117-124, 2021 09.
Article in English | MEDLINE | ID: mdl-33823988

ABSTRACT

There is a pressing need for vaccines against mosquito-borne alphaviruses such as Venezualen and eastern equine encephalitis viruses (VEEV, EEEV). We demonstrate an approach to vaccine development based on physicochemical properties (PCP) of amino acids to design a PCP-consensus sequence of the epitope-rich B domain of the VEEV major antigenic E2 protein. The consensus "spike" domain was incorporated into a live-attenuated VEEV vaccine candidate (ZPC/IRESv1). Mice inoculated with either ZPC/IRESv1 or the same virus containing the consensus E2 protein fragment (VEEVconE2) were protected against lethal challenge with VEEV strains ZPC-738 and 3908, and Mucambo virus (MUCV, related to VEEV), and had comparable neutralizing antibody titers against each virus. Both vaccines induced partial protection against Madariaga virus (MADV), a close relative of EEEV, lowering mortality from 60% to 20%. Thus PCP-consensus sequences can be integrated into a replicating virus that could, with further optimization, provide a broad-spectrum vaccine against encephalitic alphaviruses.


Subject(s)
Alphavirus Infections/prevention & control , Alphavirus/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Vaccine Development , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Alphavirus Infections/immunology , Amino Acids/chemistry , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Eastern Equine/immunology , Encephalomyelitis, Eastern Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/immunology , Female , Immunogenicity, Vaccine , Mice , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
13.
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
14.
J Fish Dis ; 44(7): 923-937, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33591590

ABSTRACT

Pancreas disease (PD) is a serious challenge in European salmonid aquaculture caused by salmonid alphavirus (SAV). In this study, we report the effect of immunization of Atlantic salmon with three attenuated infectious SAV3 strains with targeted mutations in a glycosylation site of the envelope E2 protein and/or in a nuclear localization signal in the capsid protein. In a pilot experiment, it was shown that the mutated viral strains replicated in fish, transmitted to naïve cohabitants and that the transmission had not altered the sequences. In the main experiment, the fish were immunized with the strains and challenged with SAV3 eight weeks after immunization. Immunization resulted in infection both in injected fish and 2 weeks later in the cohabitant fish, followed by a persistent but declining load of the mutated virus variants in the hearts. The immunized fish developed clinical signs and pathology consistent with PD prior to challenge. However, fish injected with the virus mutated in both E2 and capsid showed little clinical signs and had higher average weight gain than the groups immunized with the single mutated variants. The SAV strain used for challenge was not detected in the immunized fish indicating that these fish were protected against superinfection with SAV during the 12 weeks of the experiment.


Subject(s)
Alphavirus Infections/veterinary , Alphavirus/classification , Fish Diseases/prevention & control , Pancreatic Diseases/veterinary , Viral Vaccines/immunology , Alphavirus/genetics , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Fish Diseases/virology , Immunization/veterinary , Pancreatic Diseases/prevention & control , Salmo salar , Vaccines, Attenuated
15.
Fish Shellfish Immunol ; 108: 116-126, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33285168

ABSTRACT

Pancreas disease (PD) caused by salmonid alphavirus subtype 3 (SAV3) is a serious disease with large economic impact on farmed Norwegian Atlantic salmon production despite years of use of oil-adjuvanted vaccines against PD (OAVs). In this study, two commercially available PD vaccines, a DNA vaccine (DNAV) and an OAV, were compared in an experimental setting. At approximately 1040° days (dd) at 12 °C post immunization, the fish were challenged with SAV3 by cohabitation 9 days after transfer to sea water. Sampling was done prior to challenge and at 19, 54, and 83 days post-challenge (dpc). When compared to the OAV and control (Saline) groups, the DNAV group had significantly higher SAV3 neutralizing antibody titers after the immunization period, significantly lower SAV3 viremia levels at 19 dpc, significantly reduced transmission of SAV3 to naïve fish in the latter part of the viremic phase, significantly higher weight gain post-challenge, and significantly reduced prevalence and/or severity of SAV-induced morphologic changes in target organs. The DNAV group had also significantly higher post-challenge survival compared to the Saline group, but not to the OAV group. The data suggest that use of DNAV may reduce the economic impact of PD by protecting against destruction of the pancreas tissue and subsequent growth impairment which is the most common and costly clinical outcome of this disease.


Subject(s)
Alphavirus Infections/virology , Alphavirus/immunology , Fish Diseases/prevention & control , Pancreatic Diseases/veterinary , Salmo salar , Viral Vaccines/immunology , Alphavirus Infections/prevention & control , Animals , Fish Diseases/virology , Pancreatic Diseases/prevention & control , Pancreatic Diseases/virology , Vaccines, DNA/immunology
16.
Front Immunol ; 11: 591885, 2020.
Article in English | MEDLINE | ID: mdl-33224148

ABSTRACT

Mayaro (MAYV) and chikungunya viruses (CHIKV) are vector-borne arthritogenic alphaviruses that cause acute febrile illnesses. CHIKV is widespread and has recently caused large urban outbreaks, whereas the distribution of MAYV is restricted to tropical areas in South America with small and sporadic outbreaks. Because MAYV and CHIKV are closely related and have high amino acid similarity, we investigated whether vaccination against one could provide cross-protection against the other. We vaccinated A129 mice (IFNAR -/-) with vaccines based on chimpanzee adenoviral vectors encoding the structural proteins of either MAYV or CHIKV. ChAdOx1 May is a novel vaccine against MAYV, whereas ChAdOx1 Chik is a vaccine against CHIKV already undergoing early phase I clinical trials. We demonstrate that ChAdOx1 May was able to afford full protection against MAYV challenge in mice, with most samples yielding neutralizing PRNT80 antibody titers of 1:258. ChAdOx1 May also provided partial cross-protection against CHIKV, with protection being assessed using the following parameters: survival, weight loss, foot swelling and viremia. Reciprocally, ChAdOx1 Chik vaccination reduced MAYV viral load, as well as morbidity and lethality caused by this virus, but did not protect against foot swelling. The cross-protection observed is likely to be, at least in part, secondary to cross-neutralizing antibodies induced by both vaccines. In summary, our findings suggest that ChAdOx1 Chik and ChAdOx1 May vaccines are not only efficacious against CHIKV and MAYV, respectively, but also afford partial heterologous cross-protection.


Subject(s)
Adenoviridae , Alphavirus Infections/prevention & control , Alphavirus/immunology , Chikungunya Fever/prevention & control , Chikungunya virus/immunology , Genetic Vectors , Viral Vaccines , Adenoviridae/genetics , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Cross Protection/immunology , Disease Models, Animal , Genetic Engineering/methods , Genetic Vectors/genetics , Immunization , Mice , Mice, Inbred BALB C , Pan troglodytes , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
17.
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
18.
Sci Rep ; 10(1): 5306, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210270

ABSTRACT

Mayaro virus (MAYV) is endemic in South American countries where it is responsible for sporadic outbreaks of acute febrile illness. The hallmark of MAYV infection is a highly debilitating and chronic arthralgia. Although MAYV emergence is a potential threat, there are no specific therapies or licensed vaccine. In this study, we developed a murine model of MAYV infection that emulates many of the most relevant clinical features of the infection in humans and tested a live-attenuated MAYV vaccine candidate (MAYV/IRES). Intraplantar inoculation of a WT strain of MAYV into immunocompetent mice induced persistent hypernociception, transient viral replication in target organs, systemic production of inflammatory cytokines, chemokines and specific humoral IgM and IgG responses. Inoculation of MAYV/IRES in BALB/c mice induced strong specific cellular and humoral responses. Moreover, MAYV/IRES vaccination of immunocompetent and interferon receptor-defective mice resulted in protection from disease induced by the virulent wt MAYV strain. Thus, this study describes a novel model of MAYV infection in immunocompetent mice and highlights the potential role of a live-attenuated MAYV vaccine candidate in host's protection from disease induced by a virulent MAYV strain.


Subject(s)
Alphavirus Infections/prevention & control , Alphavirus/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Immunocompromised Host/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage , Alphavirus Infections/immunology , Alphavirus Infections/virology , Animals , Cytokines , Male , Mice , Mice, Inbred BALB C , South America , Viral Vaccines/immunology , Virus Replication
19.
Front Immunol ; 11: 237, 2020.
Article in English | MEDLINE | ID: mdl-32133008

ABSTRACT

Arboviruses including alphavirus are responsible for most emerging infectious diseases worldwide. Recent outbreaks of chikungunya virus serve as a stark reminder to their pathogenic potential. There are no vaccines or therapeutics currently available to contain alphavirus outbreaks. In this study we evaluated the effect of immunomodulatory CpG ODN on the clinical progression of neurotropic Sindbis virus infection. Neonatal C57Bl-6 mice challenged with Sindbis virus AR339 (25 PFU Subcutaneous) infect neurons in the CNS leading to the development of ataxia, seizures, paralysis, and death. We show that systemic administration of CpG ODN modulates the cytokine and chemokine gene expression levels in the CNS and ultimately protects neonatal mice from lethal neurotropic infection. The protection conferred by CpG ODN is controlled by innate immune response and T and B cells were dispensable. Further, protection required Type I, Type II interferons, and TNF as well as functional NK cells, but did not involve iNOS. This study confirms that administration of innate immune modulators can be used as a strategy to boost host innate immune responses and protect against neurotropic viruses reducing their pathogenic footprint.


Subject(s)
Alphavirus Infections/prevention & control , Encephalitis, Viral/prevention & control , Interferons/physiology , Killer Cells, Natural/physiology , Oligodeoxyribonucleotides/therapeutic use , Sindbis Virus , Tumor Necrosis Factor-alpha/physiology , Animals , Chlorocebus aethiops , Immunity, Innate , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/physiology , Vero Cells
20.
Parasit Vectors ; 13(1): 54, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32041638

ABSTRACT

BACKGROUND: Wolbachia pipientis are bacterial endosymbionts of arthropods currently being implemented as biocontrol agents to reduce the global burden of arboviral diseases. Some strains of Wolbachia, when introduced into Aedes aegypti mosquitoes, reduce or block the replication of RNA viruses pathogenic to humans. The wAlbB strain of Wolbachia was originally isolated from Aedes albopictus, and when transinfected into Ae. aegypti, persists in mosquitoes under high temperature conditions longer than other strains. The utility of wAlbB to block a broad spectrum of RNA viruses has received limited attention. Here we test the ability of wAlbB to reduce or block the replication of a range of Flavivirus and Alphavirus species in cell culture. METHODS: The C6/36 mosquito cell line was stably infected with the wAlbB strain using the shell-vial technique. The replication of dengue, West Nile and three strains of Zika (genus Flavivirus), and Ross River, Barmah Forest and Sindbis (genus Alphavirus) viruses was compared in wAlbB-infected cells with Wolbachia-free controls. Infectious virus titres were determined using either immunofocus or plaque assays. A general linear model was used to test for significant differences in replication between flaviviruses and alphaviruses. RESULTS: Titres of all viruses were significantly reduced in cell cultures infected with wAlbB versus Wolbachia-free controls. The magnitude of reduction in virus yields varied among virus species and, within species, also among the strains utilized. CONCLUSION: Our results suggest that wAlbB infection of arthropods could be used to reduce transmission of a wide range of pathogenic RNA viruses.


Subject(s)
Alphavirus/growth & development , Flavivirus/growth & development , Microbial Interactions , Virus Replication , Wolbachia , Aedes/microbiology , Aedes/virology , Alphavirus Infections/prevention & control , Animals , Cell Line/microbiology , Cell Line/virology , Dengue/prevention & control , Humans , Insect Vectors/microbiology , Insect Vectors/virology , Pest Control, Biological , Virus Diseases/prevention & control , Virus Diseases/transmission , West Nile Fever/prevention & control , Zika Virus Infection/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...