Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 811802, 2022.
Article in English | MEDLINE | ID: mdl-35250985

ABSTRACT

A mouse model of SARS-CoV-2 that can be developed in any molecular biology lab with standard facilities will be valuable in evaluating drugs and vaccines. Here we present a simplified SARS-CoV-2 mouse model exploiting the rapid adenoviral purification method. Mice that are sensitive to SARS-CoV-2 infection were generated by transducing human angiotensin-converting enzyme 2 (hACE2) by an adenovirus. The expression kinetics of the hACE2 in transduced mice were assessed by immunohistochemistry, RT-PCR, and qPCR. Further, the ability of the hACE2 to support viral replication was determined in vitro and in vivo. The hACE2 expression in the lungs of mice was observed for at least nine days after transduction. The murine macrophages expressing hACE2 supported viral replication with detection of high viral titers. Next, in vivo studies were carried out to determine viral replication and lung disease following SARS-CoV-2 challenge. The model supported viral replication, and the challenged mouse developed lung disease characteristic of moderate interstitial pneumonia. Further, we illustrated the utility of the system by demonstrating protection using an oral mRNA vaccine. The multicistronic vaccine design enabled by the viral self-cleaving peptides targets receptor binding domain (RBD), heptad repeat domain (HR), membrane glycoprotein (M) and epitopes of nsp13 of parental SARS-CoV-2. Further, Salmonella and Semliki Forest virus replicon were exploited, respectively, for gene delivery and mRNA expression. We recorded potent cross-protective neutralizing antibodies in immunized mice against the SARS-CoV-2 delta variant. The vaccine protected the mice against viral replication and SARS-CoV-2-induced weight loss and lung pathology. The findings support the suitability of the model for preclinical evaluation of anti-SARS-CoV-2 therapies and vaccines. In addition, the findings provide novel insights into mRNA vaccine design against infectious diseases not limiting to SARS-CoV-2.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Replicon/immunology , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , mRNA Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Cell Line , Disease Models, Animal , HEK293 Cells , Humans , Lung/virology , Male , Mice , Mice, Inbred BALB C , Spike Glycoprotein, Coronavirus/immunology , Virus Replication/immunology
2.
Antiviral Res ; 191: 105090, 2021 07.
Article in English | MEDLINE | ID: mdl-34044061

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) causes mild to severe and fatal disease in humans. Person-to-person transmission is common, necessitating the availability of rapidly deliverable therapeutic and prophylactic interventions to mitigate CCHFV spread. Previously, we showed complete protection using one dose of a viral replicon particle (VRP) vaccine administered 28 days before CCHFV challenge. In order to determine the utility of the VRP vaccine for rapid vaccination protocols, we assessed the efficacy of such vaccination administered at various intervals relative to challenge in IFNAR-/- mice. Unvaccinated mice uniformly succumbed to disease by 8 days post infection (dpi). All mice vaccinated 14, 7, or 3 days prior to CCHFV challenge survived infection. Mice vaccinated -14 or -7 dpi were fully protected from clinical disease, whereas mice inoculated -3 dpi developed signs of disease prior to recovering to baseline values 5-9 dpi. These data support the utility of the VRP vaccine for modified short course vaccination protocols to protect against disease and severe outcomes.


Subject(s)
Antibodies, Viral/blood , Hemorrhagic Fever, Crimean/prevention & control , Immunogenicity, Vaccine , Receptor, Interferon alpha-beta/genetics , Replicon/immunology , Viral Vaccines/immunology , Virion/immunology , Animals , Antibodies, Neutralizing/blood , Disease Models, Animal , Female , Hemorrhagic Fever, Crimean/immunology , Male , Mice , Mice, Knockout , Vaccination , Viral Vaccines/administration & dosage
3.
Mol Ther ; 29(6): 1970-1983, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33823303

ABSTRACT

A self-transcribing and replicating RNA (STARR)-based vaccine (LUNAR-COV19) has been developed to prevent SARS-CoV-2 infection. The vaccine encodes an alphavirus-based replicon and the SARS-CoV-2 full-length spike glycoprotein. Translation of the replicon produces a replicase complex that amplifies and prolongs SARS-CoV-2 spike glycoprotein expression. A single prime vaccination in mice led to robust antibody responses, with neutralizing antibody titers increasing up to day 60. Activation of cell-mediated immunity produced a strong viral antigen-specific CD8+ T lymphocyte response. Assaying for intracellular cytokine staining for interferon (IFN)γ and interleukin-4 (IL-4)-positive CD4+ T helper (Th) lymphocytes as well as anti-spike glycoprotein immunoglobulin G (IgG)2a/IgG1 ratios supported a strong Th1-dominant immune response. Finally, single LUNAR-COV19 vaccination at both 2 µg and 10 µg doses completely protected human ACE2 transgenic mice from both mortality and even measurable infection following wild-type SARS-CoV-2 challenge. Our findings collectively suggest the potential of LUNAR-COV19 as a single-dose vaccine.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Synthetic/administration & dosage , Alphavirus/genetics , Alphavirus/immunology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , COVID-19 Vaccines/biosynthesis , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Female , Gene Expression , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Transgenic , Replicon/immunology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/virology , Transgenes , Treatment Outcome , Vaccination/methods , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , mRNA Vaccines
4.
Sci Rep ; 10(1): 15290, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943718

ABSTRACT

Autophagy, a degradation system, works to maintain cellular homeostasis. However, as the impact of Hepatitis C virus (HCV) infection on hepatocyte autophagy and its effect on HCV replication remain unclear, we examined them. HCV infection suppressed late-stage autophagy and increased Rubicon. siRNA-mediated knockdown of Rubicon promoted autophagy in HCV-infected cells. In Huh-7 cells harbouring the HCV replicon, Rubicon knockdown downregulated the expression of type 1 interferon (IFN)-related genes and upregulated HCV replication. Rubicon overexpression or administration of bafilomycin A1 or chloroquine, an inhibitor of late-stage autophagy, suppressed autophagy and activated the type 1 IFN pathway. On the other hand, Atg7 knockout suppressed early-stage autophagy and did not activate the type 1 IFN pathway. In livers of humanized liver chimeric mice, HCV infection increased Rubicon and enhanced type 1 IFN signalling. Elimination of HCV in the mice reduced the increase in Rubicon due to HCV infection. The expression levels of Rubicon and IFN-stimulated genes in chronic hepatitis C patients were higher than those in non-B, non-C hepatitis patients. HCV infection increased Rubicon and suppressed hepatocyte autophagy, leading to activation of the intracellular immune response. Rubicon induction is involved in HCV replication via activation of the intracellular immune response.


Subject(s)
Autophagy-Related Proteins/immunology , Autophagy/immunology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Immunity, Innate/immunology , Animals , Cell Line, Tumor , Cytoplasm/immunology , Hepatocytes/immunology , Host-Pathogen Interactions/immunology , Humans , Interferon Type I/immunology , Mice , Replicon/immunology , Signal Transduction/immunology , Virus Replication/immunology
5.
Front Immunol ; 11: 622385, 2020.
Article in English | MEDLINE | ID: mdl-33584723

ABSTRACT

Self-amplifying replicon RNA (RepRNA) promotes expansion of mRNA templates encoding genes of interest through their replicative nature, thus providing increased antigen payloads. RepRNA derived from the non-cytopathogenic classical swine fever virus (CSFV) targets monocytes and dendritic cells (DCs), potentially promoting prolonged antigen expression in the DCs, contrasting with cytopathogenic RepRNA. We engineered pestivirus RepRNA constructs encoding influenza virus H5N1 (A/chicken/Yamaguchi/7/2004) nucleoprotein (Rep-NP) or hemagglutinin (Rep-HA). The inherent RNase-sensitivity of RepRNA had to be circumvented to ensure efficient delivery to DCs for intracellular release and RepRNA translation; we have reported how only particular synthetic delivery vehicle formulations are appropriate. The question remained concerning RepRNA packaged in virus replicon particles (VRPs); we have now compared an efficient polyethylenimine (PEI)-based formulation (polyplex) with VRP-delivery as well as naked RepRNA co-administered with the potent bis-(3',5')-cyclic dimeric adenosine monophosphate (c-di-AMP) adjuvant. All formulations contained a Rep-HA/Rep-NP mix, to assess the breadth of both humoral and cell-mediated defences against the influenza virus antigens. Assessment employed pigs for their close immunological relationship to humans, and as natural hosts for influenza virus. Animals receiving the VRPs, as well as PEI-delivered RepRNA, displayed strong humoral and cellular responses against both HA and NP, but with VRPs proving to be more efficacious. In contrast, naked RepRNA plus c-di-AMP could induce only low-level immune responses, in one out of five pigs. In conclusion, RepRNA encoding different influenza virus antigens are efficacious for inducing both humoral and cellular immune defences in pigs. Comparisons showed that packaging within VRP remains the most efficacious for delivery leading to induction of immune defences; however, this technology necessitates employment of expensive complementing cell cultures, and VRPs do not target human cells. Therefore, choosing the appropriate synthetic delivery vehicle still offers potential for rapid vaccine design, particularly in the context of the current coronavirus pandemic.


Subject(s)
Immunity, Cellular , Immunity, Humoral , Influenza Vaccines/immunology , RNA, Viral/immunology , Replicon/immunology , Animals , COVID-19 , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Pestivirus , RNA, Viral/administration & dosage , SARS-CoV-2 , Swine , Viral Core Proteins/immunology
6.
Mol Ther ; 27(12): 2080-2090, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31515132

ABSTRACT

RNA replicons are a promising platform technology for vaccines. To evaluate the potential of lipid nanoparticle-formulated replicons for delivery of HIV immunogens, we designed and tested an alphavirus replicon expressing a self-assembling protein nanoparticle immunogen, the glycoprotein 120 (gp120) germline-targeting engineered outer domain (eOD-GT8) 60-mer. The eOD-GT8 immunogen is a germline-targeting antigen designed to prime human B cells capable of evolving toward VRC01-class broadly neutralizing antibodies. Replicon RNA was encapsulated with high efficiency in 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)-based lipid nanoparticles, which provided effective delivery in the muscle and expression of luciferase lasting ∼30 days in normal mice, contrasting with very brief and low levels of expression obtained by delivery of equivalent modified mRNA (modRNA). eOD-GT8 60-mer-encoding replicons elicited high titers of gp120-specific antibodies following a single injection in mice, and increased levels of antigen-specific germinal center B cells compared with protein immunization. Immunization of transgenic mice expressing human inferred-germline VRC01 heavy chain B cell receptors that are the targets of the eOD antigen led to priming of B cells and somatic hypermutation consistent with VRC01-class antibody development. Altogether, these data suggest replicon delivery of Env immunogens may be a promising avenue for HIV vaccine development.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/immunology , HIV Infections/prevention & control , HIV-1/immunology , Nanoparticles/chemistry , Replicon/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Encephalitis Virus, Venezuelan Equine/genetics , Female , Gene Knock-In Techniques , HIV Antigens/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , Humans , Immunization , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles/administration & dosage , Replicon/genetics , env Gene Products, Human Immunodeficiency Virus/genetics
7.
Antiviral Res ; 170: 104573, 2019 10.
Article in English | MEDLINE | ID: mdl-31377243

ABSTRACT

No vaccines are currently licensed to prevent Crimean-Congo hemorrhagic fever virus (CCHFV) infection, which can cause mild self-limiting clinical signs or severe, often fatal hemorrhagic fever disease. Here we continued investigations into the utility of a single-dose virus replicon particle (VRP) vaccine regimen by assessing protection against Turkey or Oman strains of CCHFV. We found that all mice were completely protected from disease, supporting broad applicability of this platform for CCHFV prevention.


Subject(s)
Antibodies, Viral/blood , Hemorrhagic Fever, Crimean/prevention & control , Immunity, Heterologous , Replicon/immunology , Viral Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/blood , Disease Models, Animal , Female , Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean/immunology , Mice , Viral Vaccines/immunology , Virion/immunology
8.
J Infect Dis ; 220(8): 1281-1289, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31152662

ABSTRACT

Lassa fever is a frequently severe human disease that is endemic to several countries in West Africa. To date, no licensed vaccines are available to prevent Lassa virus (LASV) infection, even though Lassa fever is thought to be an important disease contributing to mortality and both acute and chronic morbidity. We have previously described a vaccine candidate composed of single-cycle LASV replicon particles (VRPs) and a stable cell line for their production. Here, we refine the genetic composition of the VRPs and demonstrate the ability to reproducibly purify them with high yields. Studies in the guinea pig model confirm efficacy of the vaccine candidate, demonstrate that single-cycle replication is necessary for complete protection by the VRP vaccine, and show that postexposure vaccination can confer protection from lethal outcome.


Subject(s)
Lassa Fever/prevention & control , Lassa virus/immunology , Post-Exposure Prophylaxis/methods , Vaccination/methods , Viral Vaccines/administration & dosage , A549 Cells , Africa, Western , Animals , Chlorocebus aethiops , Disease Models, Animal , Female , Guinea Pigs , Humans , Immunization Schedule , Lassa Fever/virology , Lassa virus/genetics , Lassa virus/isolation & purification , Male , Replicon/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vero Cells , Viral Vaccines/genetics , Viral Vaccines/immunology
9.
Fish Shellfish Immunol ; 89: 378-383, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30978448

ABSTRACT

Viral replicon particles are single-cycle viruses defective for function(s) needed for viral replication, which allow them to be recognized as a safer form for the vaccination of animals compared to attenuated live viruses. However, deletion of genes that are critical for the induction of protective immunity can diminish the vaccine potential of viral replicon particles. Therefore, the manipulation of viral replicon particles to produce a molecular adjuvant can be a way to increase immunogenicity of vaccines based on viral replicon particles. Chemokines are a class of chemotactic cytokines that control the migration of diverse cells of vertebrates. CXC chemokine ligand 12 (CXCL12) binds to a receptor CXCR4, and CXCL12-CXCR4 signaling plays an important role in the migration of hematopoietic cells during embryogenesis and the attraction of leukocytes. In the present study, to evaluate the possible use of CXCL12 as a molecular adjuvant for an rVHSV-ΔG vaccine and to know differences between CXCL12a and CXCL12b in the adjuvant ability, we rescued VHSV replicon particles that are expressing olive flounder CXCL12a, CXCL12b, or eGFP (rVHSV-ΔG-CXCL12a, rVHSV-ΔG-CXCL12b, or rVHSV-ΔG-eGFP), and compared the ability to attract olive flounder leucocytes and to induce protection against a VHSV challenge. In the leukocytes migration assay, supernatants collected from cells infected with rVHSV-ΔG-CXCL12a and rVHSV-ΔG-CXCL12b showed significantly higher ability to attract olive flounder leukocytes than the supernatant of cells infected with rVHSV-ΔG-eGFP. Moreover, the significantly higher number of leukocytes were attracted to rVHSV-CXCL12a supernatant compared to rVHSV-CXCL12b supernatant, suggesting that CXCL12a would be more appropriate for the induction of immunity than CXCL12b in olive flounder. In the immunization experiment, olive flounder immunized with rVHSV-ΔG-CXCL12a showed significantly higher survival rate than fish immunized with rVHSV-ΔG-CXCL12b or rVHSV-ΔG-eGFP. In addition, fish immunized with rVHSV-ΔG-CXCL12a showed the highest serum neutralization activity. These results suggest the availability of CXCL12a for a molecular adjuvant of vaccines based on VHSV replicon particles.


Subject(s)
Chemokine CXCL12/immunology , Fish Diseases/prevention & control , Fish Proteins/immunology , Flatfishes/immunology , Hemorrhagic Septicemia, Viral/prevention & control , Novirhabdovirus/immunology , Viral Vaccines/administration & dosage , Animals , Cell Movement , Fish Diseases/immunology , Fish Diseases/virology , Hemorrhagic Septicemia, Viral/immunology , Hemorrhagic Septicemia, Viral/virology , Leukocytes/immunology , Leukocytes/physiology , Random Allocation , Replicon/immunology , Viral Vaccines/immunology
10.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30674625

ABSTRACT

Hepatitis C is a liver disease caused by the hepatitis C virus (HCV) affecting 71 million people worldwide with no licensed vaccines that prevent infection. Here, we have generated four novel alphavirus-based DNA-launched self-amplifying RNA replicon (DREP) vaccines expressing either structural core-E1-E2 or nonstructural p7-NS2-NS3 HCV proteins of genotype 1a placed under the control of an alphavirus promoter, with or without an alphaviral translational enhancer (grouped as DREP-HCV or DREP-e-HCV, respectively). DREP vectors are known to induce cross-priming and further stimulation of immune responses through apoptosis, and here we demonstrate that they efficiently trigger apoptosis-related proteins in transfected cells. Immunization of mice with the DREP vaccines as the priming immunization followed by a heterologous boost with a recombinant modified vaccinia virus Ankara (MVA) vector expressing the nearly full-length genome of HCV (MVA-HCV) induced potent and long-lasting HCV-specific CD4+ and CD8+ T cell immune responses that were significantly stronger than those of a homologous MVA-HCV prime/boost immunization, with the DREP-e-HCV/MVA-HCV combination the most immunogenic regimen. HCV-specific CD4+ and CD8+ T cell responses were highly polyfunctional, had an effector memory phenotype, and were mainly directed against E1-E2 and NS2-NS3, respectively. Additionally, DREP/MVA-HCV immunization regimens induced higher antibody levels against HCV E2 protein than homologous MVA-HCV immunization. Collectively, these results provided an immunization protocol against HCV by inducing high levels of HCV-specific T cell responses as well as humoral responses. These findings reinforce the combined use of DREP-based vectors and MVA-HCV as promising prophylactic and therapeutic vaccines against HCV.IMPORTANCE HCV represents a global health problem as more than 71 million people are chronically infected worldwide. Direct-acting antiviral agents can cure HCV infection in most patients, but due to the high cost of these agents and the emergence of resistant mutants, they do not represent a feasible and affordable strategy to eradicate the virus. Therefore, a vaccine is an urgent goal that requires efforts to understand the correlates of protection for HCV clearance. Here, we describe for the first time the generation of novel vaccines against HCV based on alphavirus DNA replicons expressing HCV antigens. We demonstrate that potent T cell immune responses, as well as humoral immune responses, against HCV can be achieved in mice by using a combined heterologous prime/boost immunization protocol consisting of the administration of alphavirus replicon DNA vectors as the priming immunization followed by a boost with a recombinant modified vaccinia virus Ankara vector expressing HCV antigens.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Replicon/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Alphavirus/immunology , Animals , Antibodies, Viral/immunology , DNA/immunology , Genetic Vectors/immunology , Immunization/methods , Mice , RNA/immunology , Vaccination/methods , Vaccines, DNA/immunology , Viral Nonstructural Proteins/immunology
11.
Sci Rep ; 8(1): 12459, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127450

ABSTRACT

There are currently no licensed therapeutic treatment or preventive vaccines against Ebolavirus disease, and the 2013-2016 West African outbreak of Ebolavirus disease spread rapidly and resulted in almost 30,000 cases and more than 11,000 deaths. However, the devastating outbreak has spurred the development of novel Ebolavirus vaccines. Here, we demonstrate that alphavirus-based DNA-launched self-replicating RNA replicon vaccines (DREP) encoding either the glycoprotein (GP) gene or co-expressing the GP and VP40 genes of Sudan or Zaire Ebolavirus are immunogenic in mice inducing both binding and neutralizing antibodies as well as CD8 T cell responses. In addition, antibodies were cross-reactive against another Ebolavirus, although the specificity was higher for the vaccination antigen. DREP vaccines were more immunogenic than recombinant MVA vaccines expressing the same Ebolavirus antigens. However, a DREP prime followed by an MVA boost immunization regimen improved vaccine immunogenicity as compared to DREP and MVA homologous prime-boost immunizations. Moreover, we show that a bivalent approach targeting both Sudan and Zaire Ebolavirus can be employed without significant loss of immunity. This opens for further investigation of a pan-Ebolavirus or even a pan-filovirus vaccine.


Subject(s)
DNA/immunology , Ebola Vaccines/immunology , Ebolavirus/immunology , RNA/immunology , Replicon/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Alphavirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Chlorocebus aethiops , Female , Glycoproteins/immunology , Humans , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C , Sudan , Vaccination/methods , Vero Cells
12.
J Infect Dis ; 217(12): 1957-1966, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29800368

ABSTRACT

Lassa fever is a viral zoonosis that can be transmitted from person to person, especially in the hospital setting. The disease is endemic to several countries in West Africa and can be a major contributor to morbidity and mortality in affected areas. There are no approved vaccines to prevent Lassa virus infection. In this work, we present a vaccine candidate that combines the scalability and efficacy benefits of a live vaccine with the safety benefits of single-cycle replication. The system consists of Lassa virus replicon particles devoid of the virus essential glycoprotein gene, and a cell line that expresses the glycoprotein products, enabling efficient vaccine propagation. Guinea pigs vaccinated with these particles showed no clinical reaction to the inoculum and were protected against fever, weight loss, and lethality after infection with Lassa virus.


Subject(s)
Lassa Fever/immunology , Lassa virus/immunology , Replicon/immunology , Viral Vaccines/immunology , Africa, Western , Animals , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Guinea Pigs , Vaccines, Attenuated/immunology , Vero Cells
13.
Methods Mol Biol ; 1499: 15-35, 2017.
Article in English | MEDLINE | ID: mdl-27987141

ABSTRACT

Self-replicating RNA derived from the genomes of positive strand RNA viruses represents a powerful tool for both molecular studies on virus biology and approaches to novel safe and effective vaccines. The following chapter summarizes the principles how such RNAs can be established and used for design of vaccines. Due to the large variety of strategies needed to circumvent specific pitfalls in the design of such constructs the technical details of the experiments are not described here but can be found in the cited literature.


Subject(s)
RNA Viruses/genetics , RNA Viruses/immunology , RNA, Viral/genetics , RNA, Viral/immunology , Vaccines/immunology , Animals , Humans , Replicon/genetics , Replicon/immunology , Vaccines/genetics
14.
Methods Mol Biol ; 1499: 37-75, 2017.
Article in English | MEDLINE | ID: mdl-27987142

ABSTRACT

Most current vaccines are either inactivated pathogen-derived or protein/peptide-based, although attenuated and vector vaccines have also been developed. The former induce at best moderate protection, even as multimeric antigen, due to limitations in antigen loads and therefore capacity for inducing robust immune defense. While attenuated and vector vaccines offer advantages through their replicative nature, drawbacks and risks remain with potential reversion to virulence and interference from preexisting immunity. New advances averting these problems are combining self-amplifying replicon RNA (RepRNA) technology with nanotechnology. RepRNA are large self-replicating RNA molecules (12-15 kb) derived from viral genomes defective in at least one structural protein gene. They provide sustained antigen production, effectively increasing vaccine antigen payloads over time, without the risk of producing infectious progeny. The major limitation with RepRNA is RNase-sensitivity and inefficient uptake by dendritic cells (DCs)-absolute requirements for efficacious vaccine design. We employed biodegradable delivery vehicles to protect the RepRNA and promote DC delivery. Encapsulating RepRNA into chitosan nanoparticles, as well as condensing RepRNA with polyethylenimine (PEI), cationic lipids, or chitosans, has proven effective for delivery to DCs and induction of immune responses in vivo.


Subject(s)
Dendritic Cells/immunology , RNA/immunology , Replicon/immunology , Vaccines/immunology , Animals , Drug Delivery Systems/methods , Gene Transfer Techniques , Humans , RNA/genetics , Replicon/genetics , Vaccines/genetics
15.
Vet Res ; 47: 33, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26895704

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is the causative agent of one of the most devastating and economically significant viral disease of pigs worldwide. The vaccines currently available on the market elicit only limited protection. Recombinant vesicular stomatitis virus (VSV) replicon particles (VRP) have been used successfully to induce protection against influenza A virus (IAV) in chickens and bluetongue virus in sheep. In this study, VSV VRP expressing the PRRSV envelope proteins GP5, M, GP4, GP3, GP2 and the nucleocapsid protein N, individually or in combination, were generated and evaluated as a potential vector vaccine against PRRSV infection. High level expression of the recombinant PRRSV proteins was demonstrated in cell culture. However, none of the PRRSV antigens expressed from VRP, with the exception of the N protein, did induce any detectable antibody response in pigs before challenge infection with PRRSV. After challenge however, the antibody responses against GP5, GP4 and GP3 appeared in average 2 weeks earlier than in pigs vaccinated with the empty control VRP. No reduction of viremia was observed in the vaccinated group compared with the control group. When pigs were co-vaccinated with VRP expressing IAV antigens and VRP expressing PRRSV glycoproteins, only antibody responses to the IAV antigens were detectable. These data show that the VSV replicon vector can induce immune responses to heterologous proteins in pigs, but that the PRRSV envelope proteins expressed from VSV VRP are poorly immunogenic. Nevertheless, they prime the immune system for significantly earlier B-cell responses following PRRSV challenge infection.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Replicon/immunology , Viral Vaccines/immunology , Viremia/veterinary , Animals , Glycoproteins/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Swine , Vaccines, Synthetic/immunology , Vesiculovirus/genetics , Vesiculovirus/immunology , Viral Proteins/metabolism , Viremia/immunology , Viremia/prevention & control , Virion/immunology
16.
J Biomed Sci ; 22: 74, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26362772

ABSTRACT

BACKGROUND: To construct safer recombinant flavivirus vaccine, we exploited Japanese encephalitis virus (JEV) replicon-based platform to generate single-round infectious particles (SRIPs) that expressed heterologous neutralizing epitope SP70 derived from enterovirus-71 (EV71). Such pseudo-infectious virus particles, named SRIP-SP70, although are not genuine viable viruses, closely mimic live virus infection to elicit immune responses within one round of viral life cycle. RESULTS: We found that, besides gaining of full protection to thwart JEV lethal challenge, female outbred ICR mice, when were immunized with SRIP-SP70 by prime-boost protocol, could not only induce SP70-specific and IgG2a predominant antibodies but also provide their newborns certain degree of protection against EV71 lethal challenge. CONCLUSIONS: Our results therefore exemplify that this vaccination strategy could indeed confer an immunized host a dual protective immunity against subsequent lethal challenge from JEV or EV71.


Subject(s)
Encephalitis Virus, Japanese/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/prevention & control , Epitopes/immunology , Replicon/immunology , Viral Vaccines/immunology , Animals , Cricetinae , Encephalitis Virus, Japanese/genetics , Enterovirus A, Human/genetics , Enterovirus Infections/genetics , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Epitopes/genetics , Female , Gene Expression Regulation, Viral/immunology , Humans , Mice , Mice, Inbred ICR , Viral Vaccines/genetics , Viral Vaccines/pharmacology
17.
PLoS One ; 10(8): e0136635, 2015.
Article in English | MEDLINE | ID: mdl-26317509

ABSTRACT

There is an urgent need for effective prophylactic measures against Mycobacterium tuberculosis (Mtb) infection, particularly given the highly variable efficacy of Bacille Calmette-Guerin (BCG), the only licensed vaccine against tuberculosis (TB). Most studies indicate that cell-mediated immune responses involving both CD4+ and CD8+ T cells are necessary for effective immunity against Mtb. Genetic vaccination induces humoral and cellular immune responses, including CD4+ and CD8+ T-cell responses, against a variety of bacterial, viral, parasitic and tumor antigens, and this strategy may therefore hold promise for the development of more effective TB vaccines. Novel formulations and delivery strategies to improve the immunogenicity of DNA-based vaccines have recently been evaluated, and have shown varying degrees of success. In the present study, we evaluated DNA-launched Venezuelan equine encephalitis replicons (Vrep) encoding a novel fusion of the mycobacterial antigens α-crystallin (Acr) and antigen 85B (Ag85B), termed Vrep-Acr/Ag85B, for their immunogenicity and protective efficacy in a murine model of pulmonary TB. Vrep-Acr/Ag85B generated antigen-specific CD4+ and CD8+ T cell responses that persisted for at least 10 wk post-immunization. Interestingly, parenterally administered Vrep-Acr/Ag85B also induced T cell responses in the lung tissues, the primary site of infection, and inhibited bacterial growth in both the lungs and spleens following aerosol challenge with Mtb. DNA-launched Vrep may, therefore, represent an effective approach to the development of gene-based vaccines against TB, particularly as components of heterologous prime-boost strategies or as BCG boosters.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Mycobacterium tuberculosis/immunology , Replicon/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , alpha-Crystallins/immunology , Acyltransferases/genetics , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/genetics , Immunity, Cellular , Immunity, Humoral , Mice , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/genetics , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/pathology , Tuberculosis, Pulmonary/prevention & control , Vaccination , alpha-Crystallins/genetics
18.
J Clin Invest ; 125(8): 3241-55, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26168222

ABSTRACT

Direct delivery of aerosolized vaccines to the respiratory mucosa elicits both systemic and mucosal responses. This vaccine strategy has not been tested for Ebola virus (EBOV) or other hemorrhagic fever viruses. Here, we examined the immunogenicity and protective efficacy of an aerosolized human parainfluenza virus type 3-vectored vaccine that expresses the glycoprotein (GP) of EBOV (HPIV3/EboGP) delivered to the respiratory tract. Rhesus macaques were vaccinated with aerosolized HPIV3/EboGP, liquid HPIV3/EboGP, or an unrelated, intramuscular, Venezuelan equine encephalitis replicon vaccine expressing EBOV GP. Serum and mucosal samples from aerosolized HPIV3/EboGP recipients exhibited high EBOV-specific IgG, IgA, and neutralizing antibody titers, which exceeded or equaled titers observed in liquid recipients. The HPIV3/EboGP vaccine induced an EBOV-specific cellular response that was greatest in the lungs and yielded polyfunctional CD8+ T cells, including a subset that expressed CD103 (αE integrin), and CD4+ T helper cells that were predominately type 1. The magnitude of the CD4+ T cell response was greater in aerosol vaccinees. The HPIV3/EboGP vaccine produced a more robust cell-mediated and humoral immune response than the systemic replicon vaccine. Moreover, 1 aerosol HPIV3/EboGP dose conferred 100% protection to macaques exposed to EBOV. Aerosol vaccination represents a useful and feasible vaccination mode that can be implemented with ease in a filovirus disease outbreak situation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Ebola Vaccines , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Lung/immunology , Administration, Inhalation , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebola Vaccines/immunology , Ebola Vaccines/pharmacology , Ebolavirus/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Macaca mulatta , Male , Replicon/immunology
19.
J Infect Dis ; 212 Suppl 2: S282-94, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25943199

ABSTRACT

A major obstacle in ebolavirus research is the lack of a small-animal model for Sudan virus (SUDV), as well as other wild-type (WT) ebolaviruses. Here, we expand on research by Bray and by Lever et al suggesting that WT ebolaviruses are pathogenic in mice deficient for the type 1 interferon (IFN) α/ß receptor (IFNα/ßR-/-). We examined the disease course of several WT ebolaviruses: Boneface (SUDV/Bon) and Gulu variants of SUDV, Ebola virus (EBOV), Bundibugyo virus (BDBV), Taï Forest virus, and Reston virus (RESTV). We determined that exposure to WT SUDV or EBOV results in reproducible signs of disease in IFNα/ßR-/- mice, as measured by weight loss and partial lethality. Vaccination with the SUDV or EBOV glycoprotein (GP)-expressing Venezuelan equine encephalitis viral replicon particle vaccine protected these mice from SUDV/Bon and EBOV challenge, respectively. Treatment with SUDV- or EBOV-specific anti-GP antibodies protected mice from challenge when delivered 1-3 days after infection. Serial sampling experiments revealed evidence of disseminated intravascular coagulation in the livers of mice infected with the Boneface variant of SUDV, EBOV, and BDBV. Taken together, these data solidify the IFNα/ßR-/- mouse as an important and useful model for the study of WT EBOV disease.


Subject(s)
Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Receptor, Interferon alpha-beta/deficiency , Virulence/physiology , Animals , Antibodies, Viral/immunology , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Ebola Vaccines/immunology , Ebolavirus/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Replicon/immunology , Vaccination/methods , Vero Cells/virology , Viral Proteins/immunology , Viral Proteins/metabolism , Virulence/immunology
20.
J Infect Dis ; 212 Suppl 2: S368-71, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25732811

ABSTRACT

The current unprecedented outbreak of Ebola virus (EBOV) disease in West Africa has demonstrated the urgent need for a vaccine. Here, we describe the evaluation of an EBOV vaccine candidate based on Kunjin replicon virus-like particles (KUN VLPs) encoding EBOV glycoprotein with a D637L mutation (GP/D637L) in nonhuman primates. Four African green monkeys (Cercopithecus aethiops) were injected subcutaneously with a dose of 10(9) KUN VLPs per animal twice with an interval of 4 weeks, and animals were challenged 3 weeks later intramuscularly with 600 plaque-forming units of Zaire EBOV. Three animals were completely protected against EBOV challenge, while one vaccinated animal and the control animal died from infection. We suggest that KUN VLPs encoding GP/D637L represent a viable EBOV vaccine candidate.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Replicon/immunology , Vaccines, Virus-Like Particle/immunology , West Nile virus/immunology , Africa, Western , Animals , Chlorocebus aethiops , Glycoproteins/immunology , Immunization/methods , Primates , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...