Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 435
Filter
1.
Virology ; 597: 110152, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38968676

ABSTRACT

Cobalt-porphyrin phospholipid displays recombinant protein antigens on liposome surfaces via antigen polyhistidine-tag (His-tag), and when combined with monophosphorylated lipid A and QS-21 yields the "CPQ" vaccine adjuvant system. In this proof of principle study, CPQ was used to generate vaccine prototypes that elicited antibodies for two different alphaviruses (AV). Mice were immunized with computationally designed, His-tagged, physicochemical property consensus (PCPcon) protein antigens representing the variable B-domain of the envelope protein 2 (E2) from the serotype specific Venezuelan Equine Encephalitis Virus (VEEVcon) or a broad-spectrum AV-antigen termed EVCcon. The CPQ adjuvant enhanced the antigenicity of both proteins without eliciting detectable anti-His-tag antibodies. Antibodies elicited from mice immunized with antigens admixed with CPQ showed orders-of-magnitude higher levels of antigen-specific IgG compared to alternative control adjuvants. The ELISA results correlated with antiviral activity against VEEV strain TC83 and more weakly to Chikungunya virus 118/25. Thus, display of E.coli-produced His-tagged E2 protein segments on the surface of immunogenic liposomes elicits high levels of antigen-specific and AV neutralizing antibodies in mice with vaccination, while facilitating vaccine preparation and providing dose-sparing potential.


Subject(s)
Adjuvants, Immunologic , Alphavirus , Antibodies, Viral , Antigens, Viral , Liposomes , Viral Envelope Proteins , Viral Vaccines , Animals , Antibodies, Viral/immunology , Mice , Liposomes/immunology , Alphavirus/immunology , Antigens, Viral/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Encephalitis Virus, Venezuelan Equine/immunology , Female , Antibodies, Neutralizing/immunology , Chikungunya virus/immunology , Mice, Inbred BALB C , Immunoglobulin G/immunology , Immunoglobulin G/blood
2.
Sci Rep ; 12(1): 700, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027600

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a mosquito borne alphavirus which leads to high viremia in equines followed by lethal encephalitis and lateral spread to humans. In addition to naturally occurring outbreaks, VEEV is a potential biothreat agent with no approved human vaccine or therapeutic currently available. Single domain antibodies (sdAb), also known as nanobodies, have the potential to be effective therapeutic agents. Using an immune phage display library derived from a llama immunized with an equine vaccine that included inactivated VEEV, five sdAb sequence families were identified that showed varying ability to neutralize VEEV. One of the sequence families had been identified previously in selections against chikungunya virus, a related alphavirus of public health concern. A key advantage of sdAb is the ability to optimize properties such as neutralization capacity through protein engineering. Neutralization of VEEV was improved by two orders of magnitude by genetically linking sdAb. One of the bivalent constructs showed effective neutralization of both VEEV and chikungunya virus. Several of the bivalent constructs neutralized VEEV in cell-based assays with reductions in the number of plaques by 50% at protein concentrations of 1 ng/mL or lower, making future evaluation of their therapeutic potential compelling.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/virology , Horse Diseases/prevention & control , Horse Diseases/virology , Single-Domain Antibodies/therapeutic use , Animals , Antibodies, Neutralizing/pharmacology , Horses , Humans , Protein Engineering , Single-Domain Antibodies/pharmacology
3.
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
4.
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
5.
PLoS Negl Trop Dis ; 15(3): e0009306, 2021 03.
Article in English | MEDLINE | ID: mdl-33788849

ABSTRACT

Venezuelan Equine Encephalitis Virus (VEEV) is a major biothreat agent that naturally causes outbreaks in humans and horses particularly in tropical areas of the western hemisphere, for which no antiviral therapy is currently available. The host response to VEEV and the cellular factors this alphavirus hijacks to support its effective replication or evade cellular immune responses are largely uncharacterized. We have previously demonstrated tremendous cell-to-cell heterogeneity in viral RNA (vRNA) and cellular transcript levels during flaviviral infection using a novel virus-inclusive single-cell RNA-Seq approach. Here, we used this unbiased, genome-wide approach to simultaneously profile the host transcriptome and vRNA in thousands of single cells during infection of human astrocytes with the live-attenuated vaccine strain of VEEV (TC-83). Host transcription was profoundly suppressed, yet "superproducer cells" with extremely high vRNA abundance emerged during the first viral life cycle and demonstrated an altered transcriptome relative to both uninfected cells and cells with high vRNA abundance harvested at later time points. Additionally, cells with increased structural-to-nonstructural transcript ratio exhibited upregulation of intracellular membrane trafficking genes at later time points. Loss- and gain-of-function experiments confirmed pro- and antiviral activities in both vaccine and virulent VEEV infections among the products of transcripts that positively or negatively correlated with vRNA abundance, respectively. Lastly, comparison with single cell transcriptomic data from other viruses highlighted common and unique pathways perturbed by infection across evolutionary scales. This study provides a high-resolution characterization of the VEEV (TC-83)-host interplay, identifies candidate targets for antivirals, and establishes a comparative single-cell approach to study the evolution of virus-host interactions.


Subject(s)
Biological Transport/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/pathology , Host-Pathogen Interactions/genetics , Transcription, Genetic/genetics , Virus Internalization , Animals , Antibodies, Viral/immunology , Astrocytes/virology , Cell Line , Chlorocebus aethiops , Cricetinae , Encephalitis Virus, Venezuelan Equine/immunology , Gene Expression Regulation, Viral/genetics , Horses , Humans , RNA, Viral/genetics , Single-Cell Analysis , Vaccines, Attenuated/immunology , Vero Cells , Virus Replication/physiology
6.
Virulence ; 12(1): 430-443, 2021 12.
Article in English | MEDLINE | ID: mdl-33487119

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus that can cause debilitating, acute febrile illness and potentially result in encephalitis. Currently, there are no FDA-licensed vaccines or specific therapeutics for VEEV. Previous studies have demonstrated that VEEV infection results in increased blood-brain barrier (BBB) permeability that is mediated by matrix metalloproteinases (MMPs). Furthermore, after subarachnoid hemorrhage in mice, MMP-9 is upregulated in the brain and mediates BBB permeability in a toll-like receptor 4 (TLR4)-dependent manner. Here, we demonstrate that disease in C3H mice during VEEV TC-83 infection is dependent on TLR4 because intranasal infection of C3H/HeN (TLR4 WT ) mice with VEEV TC-83 resulted in mortality as opposed to survival of TLR4-defective C3H/HeJ (TLR4 mut ) mice. In addition, BBB permeability was induced to a lesser extent in TLR4 mut mice compared with TLR4 WT mice during VEEV TC-83 infection as determined by sodium fluorescein and fluorescently-conjugated dextran extravasation. Moreover, MMP-9, MMP-2, ICAM-1, CCL2 and IFN-γ were all induced to significantly lower levels in the brains of infected TLR4 mut mice compared with infected TLR4 WT mice despite the absence of significantly different viral titers or immune cell populations in the brains of infected TLR4 WT and TLR4 mut mice. These data demonstrate the critical role of TLR4 in mediating BBB permeability and disease in C3H mice during VEEV TC-83 infection, which suggests that TLR4 is a potential target for the development of therapeutics for VEEV.


Subject(s)
Blood-Brain Barrier/metabolism , Encephalitis Virus, Venezuelan Equine/pathogenicity , Toll-Like Receptor 4/genetics , Animals , Brain/virology , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/virology , Female , Mice , Mice, Inbred C3H , Permeability , Toll-Like Receptor 4/metabolism , Virus Replication
7.
Mol Ther ; 29(3): 1174-1185, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33352107

ABSTRACT

Self-amplifying RNA (saRNA) is a cutting-edge platform for both nucleic acid vaccines and therapeutics. saRNA is self-adjuvanting, as it activates types I and III interferon (IFN), which enhances the immunogenicity of RNA vaccines but can also lead to inhibition of translation. In this study, we screened a library of saRNA constructs with cis-encoded innate inhibiting proteins (IIPs) and determined the effect on protein expression and immunogenicity. We observed that the PIV-5 V and Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a proteins enhance protein expression 100- to 500-fold in vitro in IFN-competent HeLa and MRC5 cells. We found that the MERS-CoV ORF4a protein partially abates dose nonlinearity in vivo, and that ruxolitinib, a potent Janus kinase (JAK)/signal transducer and activator of transcription (STAT) inhibitor, but not the IIPs, enhances protein expression of saRNA in vivo. Both the PIV-5 V and MERS-CoV ORF4a proteins were found to enhance the percentage of resident cells in human skin explants expressing saRNA and completely rescued dose nonlinearity of saRNA. Finally, we observed that the MERS-CoV ORF4a increased the rabies virus (RABV)-specific immunoglobulin G (IgG) titer and neutralization half-maximal inhibitory concentration (IC50) by ∼10-fold in rabbits, but not in mice or rats. These experiments provide a proof of concept that IIPs can be directly encoded into saRNA vectors and effectively abate the nonlinear dose dependency and enhance immunogenicity.


Subject(s)
Immunity, Innate/drug effects , Immunogenicity, Vaccine , Protein Biosynthesis/drug effects , Vaccines, Synthetic/pharmacology , Viral Envelope Proteins/administration & dosage , Animals , Cell Line , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/pathogenicity , Fibroblasts , Gene Expression Regulation , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin G/biosynthesis , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Janus Kinases/immunology , Mice , Middle East Respiratory Syndrome Coronavirus/drug effects , Middle East Respiratory Syndrome Coronavirus/immunology , Middle East Respiratory Syndrome Coronavirus/pathogenicity , NF-kappa B/genetics , NF-kappa B/immunology , Nitriles , Parainfluenza Virus 5/drug effects , Parainfluenza Virus 5/immunology , Parainfluenza Virus 5/pathogenicity , Pyrazoles/pharmacology , Pyrimidines , Rabbits , Rabies virus/drug effects , Rabies virus/immunology , Rabies virus/pathogenicity , Rats , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Signal Transduction , Vaccines, Synthetic/biosynthesis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , mRNA Vaccines
8.
Am J Trop Med Hyg ; 103(6): 2429-2437, 2020 12.
Article in English | MEDLINE | ID: mdl-33124532

ABSTRACT

Madariaga virus (MADV) has recently been associated with severe human disease in Panama, where the closely related Venezuelan equine encephalitis virus (VEEV) also circulates. In June 2017, a fatal MADV infection was confirmed in a community of Darien Province. We conducted a cross-sectional outbreak investigation with human and mosquito collections in July 2017, where sera were tested for alphavirus antibodies and viral RNA. In addition, by applying a catalytic, force-of-infection (FOI) statistical model to two serosurveys from Darien Province in 2012 and 2017, we investigated whether endemic or epidemic alphavirus transmission occurred historically. In 2017, MADV and VEEV IgM seroprevalences were 1.6% and 4.4%, respectively; IgG antibody prevalences were MADV: 13.2%, VEEV: 16.8%, Una virus (UNAV): 16.0%, and Mayaro virus: 1.1%. Active viral circulation was not detected. Evidence of MADV and UNAV infection was found near households, raising questions about its vectors and enzootic transmission cycles. Insomnia was associated with MADV and VEEV infections, depression symptoms were associated with MADV, and dizziness with VEEV and UNAV. Force-of-infection analyses suggest endemic alphavirus transmission historically, with recent increased human exposure to MADV and VEEV in Aruza and Mercadeo, respectively. The lack of additional neurological cases suggests that severe MADV and VEEV infections occur only rarely. Our results indicate that over the past five decades, alphavirus infections have occurred at low levels in eastern Panama, but that MADV and VEEV infections have recently increased-potentially during the past decade. Endemic infections and outbreaks of MADV and VEEV appear to differ spatially in some locations of eastern Panama.


Subject(s)
Encephalomyelitis, Eastern Equine/epidemiology , Encephalomyelitis, Venezuelan Equine/epidemiology , Farmers/statistics & numerical data , Adolescent , Adult , Age Distribution , Aged , Aged, 80 and over , Alphavirus/immunology , Alphavirus Infections/epidemiology , Alphavirus Infections/immunology , Alphavirus Infections/physiopathology , Animals , Antibodies, Viral/immunology , Chikungunya Fever/epidemiology , Chikungunya Fever/immunology , Chikungunya Fever/physiopathology , Chikungunya virus/immunology , Child , Child, Preschool , Cross-Sectional Studies , Depression/physiopathology , Dizziness/physiopathology , Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Eastern Equine/immunology , Encephalomyelitis, Eastern Equine/physiopathology , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/physiopathology , Endemic Diseases , Epidemics , Fatigue/physiopathology , Female , Housing/statistics & numerical data , Humans , Immunoglobulin G , Immunoglobulin M , Male , Middle Aged , Mosquito Vectors/virology , Panama/epidemiology , Semliki forest virus/immunology , Seroepidemiologic Studies , Sleep Initiation and Maintenance Disorders/physiopathology , Young Adult
9.
Antiviral Res ; 182: 104905, 2020 10.
Article in English | MEDLINE | ID: mdl-32800880

ABSTRACT

There is a pressing need for new vaccines against alphaviruses, which can cause fatal encephalitis (Venezuelan equine encephalitis virus (VEEV) and others) and severe arthralgia (e.g. Chikungunya virus, CHIKV). These positive-strand RNA viruses are diverse and evolve rapidly, meaning that the sequence of any vaccine should cover multiple strains that may be quite different from any previous isolate. Here, consensus proteins were produced to represent the common physicochemical properties (PCPs) of the epitope rich, B domain of the E2 envelope protein. PCP-consensus proteins were based on multiple strains of VEEV (VEEVcon) and CHIKV (CHIKVcon) or the conserved PCPs of 24 different alphaviruses (AllAVcon). The AllAVcon was altered to include binding sites for neutralizing antibodies of both VEEV and CHIKV strains (Mosaikcon). All four designed proteins were produced solubly in E. coli and purified. They formed the ß-strand core expected from experimental structures of this region of the wild type E2 proteins as indicated by circular dichroism (CD) spectra. Furthermore, the CHIKVcon protein bound to a structure dependent, CHIKV neutralizing monoclonal antibody. The AllAVcon and Mosaikcon proteins bound to polyclonal antibodies generated during natural infection with either VEEV or CHIKV, indicating they contained epitopes of both serotypes. The Mosaikcon antigen induced antibodies in rabbit sera that recognized both the VEEVcon and CHIKVcon spike proteins. These PCP-consensus antigens are promising starting points for novel, broad-spectrum alphavirus vaccines.


Subject(s)
Alphavirus/chemistry , Alphavirus/immunology , Antibodies, Viral/blood , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/chemistry , Antigens, Viral/immunology , Chikungunya virus/chemistry , Chikungunya virus/immunology , Circular Dichroism , Consensus , Drug Design , Encephalitis Virus, Venezuelan Equine/chemistry , Encephalitis Virus, Venezuelan Equine/immunology , Epitopes/immunology , Freund's Adjuvant/administration & dosage , Male , Mass Spectrometry , Rabbits , Viral Vaccines/administration & dosage
10.
EBioMedicine ; 56: 102819, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32512518

ABSTRACT

BACKGROUND: Live attenuated vaccines (LAVs) can mimic natural infection and have advantages to stimulate a robust and sustained immune response as well as to confer long-term protection. However, safety concerns is one of the major obstacles for LAVs development. In an effort to achieve the optimal balance between immunogenicity and safety, researchers currently have taken different strategies for the development of LAVs. METHODS: We constructed a novel infectious self-propagating hybrid replicon particle (PRP), VEEV-RABV-G, through replacing the entire structural proteins of the Venezuelan equine encephalitis virus (VEEV) with the glycoprotein of rabies virus (RABV-G) as the single structural protein. We evaluated the potential of VEEV-RABV-G as a safe live attenuated vaccine in mice model. FINDINGS: We found that VEEV-RABV-G could self-propagate efficiently in cell culture and induce a robust humoral immunity and provide protection against virulent RABV challenge in immunized mice. Remarkably, VEEV-RABV-G is highly attenuated in both adult and sucking mice, causing much weaker inflammatory and apoptotic effects in the brains of infected adult mice and significantly lower weight loss and morbidity compared with the commonly used RABV-derived LAVs. INTERPRETATION: This study reveals the feasibility of developing novel rabies vaccines based on the self-replicating PRPs. FUNDING: This work was supported by the National Key Research and Development Program of China (2016YFD0500400).


Subject(s)
Encephalitis Virus, Venezuelan Equine/physiology , Rabies Vaccines/administration & dosage , Rabies virus/metabolism , Rabies/prevention & control , Viral Structural Proteins/immunology , Animals , Animals, Newborn , Cell Line , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/immunology , Female , Immunity, Humoral , Mice , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies virus/genetics , Rabies virus/immunology , Recombinant Proteins/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virus Replication
11.
Viruses ; 12(3)2020 02 27.
Article in English | MEDLINE | ID: mdl-32120905

ABSTRACT

Chikungunya virus (CHIKV) is an alphavirus that causes febrile illness punctuated by severe polyarthralgia. After the emergence of CHIKV in the Western Hemisphere, multiple reports of congenital infections were published that documented neurological complications, cardiac defects, respiratory distress, and miscarriage. The Western Hemisphere is endemic to several alphaviruses, and whether antigenic cross-reactivity can impact the course of infection has not been explored. Recent advances in biomedical engineering have produced cell co-culture models that replicate the cellular interface at the maternal fetal axis. We employed a trans-well assay to determine if cross-reactive antibodies affected the movement and replication of CHIKV across placental cells and into an embryoid body. The data showed that antibodies to Venezuelan equine encephalitis virus significantly reduced CHIKV viral load in embryoid bodies. The data highlighted the fact that viral pathogenesis can be cell-specific and that exploiting antigenic cross-reactivity could be an avenue for reducing the impact of congenital CHIKV infections.


Subject(s)
Antibodies, Viral/immunology , Chikungunya Fever/prevention & control , Chikungunya virus/immunology , Cross Reactions/immunology , Embryoid Bodies/immunology , Embryoid Bodies/virology , Encephalitis Virus, Venezuelan Equine/immunology , Animals , Cells, Cultured , Chikungunya Fever/virology , Coculture Techniques , Female , Horses , Human Umbilical Vein Endothelial Cells , Humans , Neutralization Tests , Pregnancy
12.
Vaccine ; 38(17): 3378-3386, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32085953

ABSTRACT

Live-attenuated V4020 vaccine for Venezuelan equine encephalitis virus (VEEV) containing attenuating rearrangement of the virus structural genes was evaluated in a non-human primate model for immunogenicity and protective efficacy against aerosol challenge with wild-type VEEV. The genomic RNA of V4020 vaccine virus was encoded in the pMG4020 plasmid under control of the CMV promoter and contained the capsid gene downstream from the glycoprotein genes. It also included attenuating mutations from the VEE TC83 vaccine, with E2-120Arg substitution genetically engineered to prevent reversion mutations. The population of V4020 vaccine virus derived from pMG4020-transfected Vero cells was characterized by next generation sequencing (NGS) and indicated no detectable genetic reversions. Cynomolgus macaques were vaccinated with V4020 vaccine virus. After one or two vaccinations including by intramuscular route, high levels of virus-neutralizing antibodies were confirmed with no viremia or apparent adverse reactions to vaccinations. The protective effect of vaccination was evaluated using an aerosol challenge with VEEV. After challenge, macaques had no detectable viremia, demonstrating a protective effect of vaccination with live V4020 VEEV vaccine.


Subject(s)
Encephalomyelitis, Venezuelan Equine , Viral Vaccines/immunology , Aerosols , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chlorocebus aethiops , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Macaca , Vero Cells , Viral Vaccines/genetics , Viremia/prevention & control
13.
Front Immunol ; 11: 598847, 2020.
Article in English | MEDLINE | ID: mdl-33542715

ABSTRACT

Venezuelan, eastern and western equine encephalitis viruses (EEV) can cause severe disease of the central nervous system in humans, potentially leading to permanent damage or death. Yet, no licensed vaccine for human use is available to protect against these mosquito-borne pathogens, which can be aerosolized and therefore pose a bioterror threat in addition to the risk of natural outbreaks. Using the mouse aerosol challenge model, we evaluated the immunogenicity and efficacy of EEV vaccines that are based on the modified vaccinia Ankara-Bavarian Nordic (MVA-BN®) vaccine platform: three monovalent vaccines expressing the envelope polyproteins E3-E2-6K-E1 of the respective EEV virus, a mixture of these three monovalent EEV vaccines (Triple-Mix) as a first approach to generate a multivalent vaccine, and a true multivalent alphavirus vaccine (MVA-WEV, Trivalent) encoding the polyproteins of all three EEVs in a single non-replicating MVA viral vector. BALB/c mice were vaccinated twice in a four-week interval and samples were assessed for humoral and cellular immunogenicity. Two weeks after the second immunization, animals were exposed to aerosolized EEV. The majority of vaccinated animals exhibited VEEV, WEEV, and EEEV neutralizing antibodies two weeks post-second administration, whereby the average VEEV neutralizing antibodies induced by the monovalent and Trivalent vaccine were significantly higher compared to the Triple-Mix vaccine. The same statistical difference was observed for VEEV E1 specific T cell responses. However, all vaccinated mice developed comparable interferon gamma T cell responses to the VEEV E2 peptide pools. Complete protective efficacy as evaluated by the prevention of mortality and morbidity, lack of clinical signs and viremia, was demonstrated for the respective monovalent MVA-EEV vaccines, the Triple-Mix and the Trivalent single vector vaccine not only in the homologous VEEV Trinidad Donkey challenge model, but also against heterologous VEEV INH-9813, WEEV Fleming, and EEEV V105-00210 inhalational exposures. These EEV vaccines, based on the safe MVA vector platform, therefore represent promising human vaccine candidates. The trivalent MVA-WEV construct, which encodes antigens of all three EEVs in a single vector and can potentially protect against all three encephalitic viruses, is currently being evaluated in a human Phase 1 trial.


Subject(s)
Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Western Equine/immunology , Encephalomyelitis, Equine/prevention & control , Viral Vaccines/immunology , Aerosols , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cross Protection/immunology , Disease Models, Animal , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Equine/mortality , Female , Immunization , Mice , Mortality , Neutralization Tests , Vaccines, DNA , Viral Vaccines/administration & dosage
14.
Viruses ; 11(9)2019 08 31.
Article in English | MEDLINE | ID: mdl-31480472

ABSTRACT

Licensure of a vaccine to protect against aerosolized Venezuelan equine encephalitis virus (VEEV) requires use of the U.S. Food and Drug Administration (FDA) Animal Rule to assess vaccine efficacy as human studies are not feasible or ethical. An approach to selecting VEEV challenge strains for use under the Animal Rule was developed, taking into account Department of Defense (DOD) vaccine requirements, FDA Animal Rule guidelines, strain availability, and lessons learned from the generation of filovirus challenge agents within the Filovirus Animal Nonclinical Group (FANG). Initial down-selection to VEEV IAB and IC epizootic varieties was based on the DOD objective for vaccine protection in a bioterrorism event. The subsequent down-selection of VEEV IAB and IC isolates was based on isolate availability, origin, virulence, culture and animal passage history, known disease progression in animal models, relevancy to human disease, and ability to generate sufficient challenge material. Methods for the propagation of viral stocks (use of uncloned (wild-type), plaque-cloned, versus cDNA-cloned virus) to minimize variability in the potency of the resulting challenge materials were also reviewed. The presented processes for VEEV strain selection and the propagation of viral stocks may serve as a template for animal model development product testing under the Animal Rule to other viral vaccine programs. This manuscript is based on the culmination of work presented at the "Alphavirus Workshop" organized and hosted by the Joint Vaccine Acquisition Program (JVAP) on 15 December 2014 at Fort Detrick, Maryland, USA.


Subject(s)
Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Viral Vaccines/therapeutic use , Animals , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/virology , Guidelines as Topic , Humans , Immunization Programs/methods , Immunization Programs/standards , Virology/methods
15.
Vaccine ; 37(25): 3317-3325, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31072736

ABSTRACT

Novel live-attenuated V4020 vaccine was prepared for Venezuelan equine encephalitis virus (VEEV), an alphavirus from the Togaviridae family. The genome of V4020 virus was rearranged, with the capsid gene expressed using a duplicate subgenomic promoter downstream from the glycoprotein genes. V4020 also included both attenuating mutations from the TC83 VEEV vaccine secured by mutagenesis to prevent reversion mutations. The full-length infectious RNA of V4020 vaccine virus was expressed from pMG4020 plasmid downstream from the CMV promoter and launched replication of live-attenuated V4020 in vitro or in vivo. BALB/c mice vaccinated with a single dose of V4020 virus or with pMG4020 plasmid had no adverse reactions to vaccinations and developed high titers of neutralizing antibodies. After challenge with the wild type VEEV, vaccinated mice survived with no morbidity, while all unvaccinated controls succumbed to lethal infection. Intracranial injections in mice showed attenuated replication of V4020 vaccine virus as compared to the TC83. We conclude that V4020 vaccine has safety advantage over TC83, while provides equivalent protection in a mouse VEEV challenge model.


Subject(s)
Antibodies, Viral/blood , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/prevention & control , Genome, Viral , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , DNA, Viral/genetics , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/immunology , Horses , Mice , Mice, Inbred BALB C , Mutation , Plasmids/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/genetics , Virus Replication
16.
Mol Ther ; 27(4): 850-865, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30770173

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a known biological defense threat. A live-attenuated investigational vaccine, TC-83, is available, but it has a high non-response rate and can also cause severe reactogenicity. We generated two novel VEE vaccine candidates using self-amplifying mRNA (SAM). LAV-CNE is a live-attenuated VEE SAM vaccine formulated with synthetic cationic nanoemulsion (CNE) and carrying the RNA genome of TC-83. IAV-CNE is an irreversibly-attenuated VEE SAM vaccine formulated with CNE, delivering a TC-83 genome lacking the capsid gene. LAV-CNE launches a TC-83 infection cycle in vaccinated subjects but eliminates the need for live-attenuated vaccine production and potentially reduces manufacturing time and complexity. IAV-CNE produces a single cycle of RNA amplification and antigen expression without generating infectious viruses in subjects, thereby creating a potentially safer alternative to live-attenuated vaccine. Here, we demonstrated that mice vaccinated with LAV-CNE elicited immune responses similar to those of TC-83, providing 100% protection against aerosol VEEV challenge. IAV-CNE was also immunogenic, resulting in significant protection against VEEV challenge. These studies demonstrate the proof of concept for using the SAM platform to streamline the development of effective attenuated vaccines against VEEV and closely related alphavirus pathogens such as western and eastern equine encephalitis and Chikungunya viruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/drug therapy , Gene Amplification , Immunogenicity, Vaccine , RNA, Messenger/genetics , Vaccines, Attenuated/therapeutic use , Viral Vaccines/therapeutic use , A549 Cells , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Emulsions/chemistry , Encephalomyelitis, Venezuelan Equine/virology , Female , Humans , Mice , Mice, Inbred BALB C , Transfection , Viral Vaccines/pharmacology , Virus Replication
17.
Comp Med ; 68(5): 380-395, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30282570

ABSTRACT

Licensure of medical countermeasure vaccines to protect against aerosolized Venezuelan equine encephalitis virus (VEEV) requires the use of the Animal Rule to assess vaccine efficacy, because human studies are not feasible or ethical. We therefore performed a retrospective study of VEE cases that occurred in at-risk laboratory workers and support personnel during the United States Biowarfare Program (1943-1969) to better define percutaneous- and aerosol-acquired VEE in humans and to compare these results with those described for the NHP model (in which high-dose aerosol VEEV challenge led to more severe encephalitis than parenteral challenge). Record review and analysis of 17 aerosol- and 23 percutaneous-acquired human cases of VEE included incubation period, symptoms, physical examination findings, and markers of infection. Human VEE disease by both exposure routes presented as acute febrile illness, typically with fever, chills, headache, back pain, malaise, myalgia, anorexia, and nausea. Aerosol exposure more commonly led to upper respiratory tract-associated findings of sore throat (59% compared with 26%), pharyngeal erythema (76% compared with 52%), neck pain (29% compared with 4%), and cervical lymphadenopathy (29% compared with 4%). Other disease manifestations, including encephalitis, were similar between the 2 exposure groups. The increase in upper respiratory tract findings in aerosol-acquired VEE in humans has not previously been reported but is supported by the mouse model, which showed nasal mucosal necrosis, necrotizing rhinitis, and an increase in upper respiratory tract viral burden associated with aerosol VEEV challenge. Fever, viremia, and lymphopenia were common markers of VEE disease in both humans and NHP, regardless of the exposure route. Taken collectively, our findings provide support for use of the nonlethal NHP model for advanced development of medical countermeasures against aerosol- or percutaneous-acquired VEE.


Subject(s)
Encephalomyelitis, Venezuelan Equine/prevention & control , Primates/virology , Viral Vaccines/therapeutic use , Aerosols , Animals , Antibodies, Viral/blood , Biological Warfare Agents , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/transmission , Humans , Infectious Disease Incubation Period , Neutralization Tests , Primates/immunology , Retrospective Studies , Treatment Outcome
18.
Virulence ; 9(1): 1403-1421, 2018.
Article in English | MEDLINE | ID: mdl-30101649

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a neurotropic arbovirus that is highly infectious as an aerosol and can result in an encephalitic phenotype in infected individuals. VEEV infections are known to be associated with robust inflammation that eventually contributes to neurodegenerative phenotypes. In this study, we utilize the TC-83 strain of VEEV, which is known to induce the expression of IL-6, IL-8, and other pro-inflammatory cytokines. We had previously demonstrated that TC-83 infection resulted in changes in mitochondrial function, eventually resulting in mitophagy. In this manuscript, we provide data that links upstream mitochondrial dysfunction with downstream pro-inflammatory cytokine production in the context of microglia and astrocytoma cells. We also provide data on the role of bystander cells, which significantly contribute to the overall inflammatory load. Use of a mitochondrial-targeted antioxidant, mitoquinone mesylate, greatly reduced the inflammatory cytokine load and ameliorated bystander cell inflammatory responses more significantly than a broad-spectrum anti-inflammatory compound (BAY 11-7082). Our data suggest that the inflammatory mediators, especially IL-1ß, may prime naïve cells to infection and lead to increased infection rates in microglial and astrocytoma cells. Cumulatively, our data suggest that the interplay between mitochondrial dysfunction and inflammatory events elicited in a neuronal microenvironment during a TC-83 infection may contribute to the spread of infection.


Subject(s)
Cytokines/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , Neuroglia/immunology , Animals , Astrocytes/immunology , Astrocytes/virology , Astrocytoma/immunology , Astrocytoma/virology , Cell Line, Tumor , Humans , Inflammation , Membrane Potential, Mitochondrial , Microglia/immunology , Microglia/virology , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/pathology , Organophosphorus Compounds/pharmacology , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
19.
Vector Borne Zoonotic Dis ; 18(5): 266-272, 2018 05.
Article in English | MEDLINE | ID: mdl-29652644

ABSTRACT

INTRODUCTION: Alphaviruses can produce febrile illness and encephalitis in dead-end hosts such as horses and humans. Within this genus, the Venezuelan Equine Encephalitis virus (VEEV) complex includes pathogenic epizootic subtypes and enzootic subtypes that are not pathogenic in horses (except subtype IE, Mexican strains), although they can cause febrile symptoms in humans. The Rio Negro virus (RNV-VEEV subtype VI) circulates in Argentina, where it was associated with undifferentiated febrile illness. Mayaro (MAYV) and Una (UNAV) viruses belong to a different group, the Semliki Forest virus complex, with confirmed circulation. OBJECTIVE: The present study aimed to determine RNV, MAYV, and UNAV seroprevalences by plaque reduction neutralization test in 652 samples of Paraguayan individuals mainly from the Central Department, between years 2012 and 2013. METHODS: Samples with antibodies titer >1:20 against RNV were also tested for Mosso das Pedras-subtype IF, subtype IAB, and Pixuna (PIXV)-subtype IV viruses that belongs to VEEV antigenic complex. RESULTS: The overall seroprevalence of RNV was 3.83%, and for UNAV it was 0.46%, and no neutralizing antibodies were detected against MAYV in the studied population. Two of the twenty-seven heterotypic samples were positive for PIXV. The 50.1% of neutralizing antibody titers against RNV were high (equal to or greater than 1/640), suggesting recent infections. The effect of age on the prevalence of RNV was negligible. CONCLUSIONS: These results bring new information about neglected alphaviruses in South America, and these data will serve as the basis for future studies of seroprevalence of other VEEV, and studies to search potential hosts and vectors of these viruses in the region.


Subject(s)
Alphavirus Infections/epidemiology , Alphavirus/immunology , Antibodies, Viral/blood , Adolescent , Adult , Aged , Aged, 80 and over , Alphavirus/genetics , Alphavirus/isolation & purification , Alphavirus Infections/virology , Child , Child, Preschool , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/isolation & purification , Female , Humans , Infant , Male , Middle Aged , Paraguay/epidemiology , Seroepidemiologic Studies , Young Adult
20.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29540599

ABSTRACT

Zoonotic viruses circulate as swarms in animal reservoirs and can emerge into human populations, causing epidemics that adversely affect public health. Portable, safe, and effective vaccine platforms are needed in the context of these outbreak and emergence situations. In this work, we report the generation and characterization of an alphavirus replicon vaccine platform based on a non-select agent, attenuated Venezuelan equine encephalitis (VEE) virus vaccine, strain 3526 (VRP 3526). Using both noroviruses and coronaviruses as model systems, we demonstrate the utility of the VRP 3526 platform in the generation of recombinant proteins, production of virus-like particles, and in vivo efficacy as a vaccine against emergent viruses. Importantly, packaging under biosafety level 2 (BSL2) conditions distinguishes VRP 3526 from previously reported alphavirus platforms and makes this approach accessible to the majority of laboratories around the world. In addition, improved outcomes in the vulnerable aged models as well as against heterologous challenge suggest improved efficacy compared to that of previously attenuated VRP approaches. Taking these results together, the VRP 3526 platform represents a safe and highly portable system that can be rapidly deployed under BSL2 conditions for generation of candidate vaccines against emerging microbial pathogens.IMPORTANCE While VEE virus replicon particles provide a robust, established platform for antigen expression and vaccination, its utility has been limited by the requirement for high-containment-level facilities for production and packaging. In this work, we utilize an attenuated vaccine strain capable of use at lower biocontainment level but retaining the capacity of the wild-type replicon particle. Importantly, the new replicon platform provides equal protection for aged mice and following heterologous challenge, which distinguishes it from other attenuated replicon platforms. Together, the new system represents a highly portable, safe system for use in the context of disease emergence.


Subject(s)
Antibodies, Viral/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Aging/immunology , Animals , Antibodies, Viral/blood , Cell Line , Chlorocebus aethiops , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/virology , Female , Humans , Mice , Mice, Inbred BALB C , Severe Acute Respiratory Syndrome/prevention & control , Severe Acute Respiratory Syndrome/virology , Vero Cells , Zoonoses/prevention & control , Zoonoses/virology
SELECTION OF CITATIONS
SEARCH DETAIL