Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1407: 191-208, 2023.
Article in English | MEDLINE | ID: mdl-36920698

ABSTRACT

Lyssaviruses, which belong to the family Rhabdoviridae, are enveloped and bullet-shaped ssRNA viruses with genetic diversity. All members of Lyssavirus genus are known to infect warm-blooded animals and cause the fatal disease rabies. The rabies virus (RABV) in lyssavirus is the major pathogen to cause fatal rabies. The pseudotyped RABV is constructed to study the biological functions of G protein and evaluation of anti-RABV products including vaccine-induced antisera, rabies immunoglobulins (RIG), neutralizing mAbs, and other antiviral inhibitors. In this chapter, we focus on RABV as a representative and describe the construction of RABV G protein bearing pseudotyped virus and its applications. Other non-RABV lyssaviruses are also included.


Subject(s)
Lyssavirus , Rabies Vaccines , Rabies virus , Rabies , Rhabdoviridae Infections , Animals , Lyssavirus/genetics , Viral Pseudotyping , Rabies virus/genetics , Rabies Vaccines/genetics , Rabies Vaccines/metabolism
2.
Biomolecules ; 12(10)2022 10 07.
Article in English | MEDLINE | ID: mdl-36291645

ABSTRACT

Rabies is a neurological disease that causes between 40,000 and 70,000 deaths every year. Once a rabies patient has become symptomatic, there is no effective treatment for the illness, and in unvaccinated individuals, the case-fatality rate of rabies is close to 100%. French scientists Louis Pasteur and Émile Roux developed the first vaccine for rabies in 1885. If administered before the virus reaches the brain, the modern rabies vaccine imparts long-lasting immunity to the virus and saves more than 250,000 people every year. However, the rabies virus can suppress the host's immune response once it has entered the cells of the brain, making death likely. This study aimed to make use of disorder-based proteomics and bioinformatics to determine the potential impact that intrinsically disordered protein regions (IDPRs) in the proteome of the rabies virus might have on the infectivity and lethality of the disease. This study used the proteome of the Rabies lyssavirus (RABV) strain Pasteur Vaccins (PV), one of the best-understood strains due to its use in the first rabies vaccine, as a model. The data reported in this study are in line with the hypothesis that high levels of intrinsic disorder in the phosphoprotein (P-protein) and nucleoprotein (N-protein) allow them to participate in the creation of Negri bodies and might help this virus to suppress the antiviral immune response in the host cells. Additionally, the study suggests that there could be a link between disorder in the matrix (M) protein and the modulation of viral transcription. The disordered regions in the M-protein might have a possible role in initiating viral budding within the cell. Furthermore, we checked the prevalence of functional disorder in a set of 37 host proteins directly involved in the interaction with the RABV proteins. The hope is that these new insights will aid in the development of treatments for rabies that are effective after infection.


Subject(s)
Intrinsically Disordered Proteins , Rabies Vaccines , Rabies virus , Rabies , Humans , Rabies virus/metabolism , Rabies/prevention & control , Rabies Vaccines/metabolism , Computational Biology , Proteome/metabolism , Intrinsically Disordered Proteins/metabolism , Phosphoproteins/metabolism , Antiviral Agents/metabolism , Nucleoproteins/metabolism
3.
J Virol ; 95(20): e0079021, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34346765

ABSTRACT

Rabies is a fatal zoonosis that causes encephalitis in mammals, and vaccination is the most effective method to control and eliminate rabies. Virus-like vesicles (VLVs), which are characterized as infectious, self-propagating membrane-enveloped particles composed of only Semliki Forest virus (SFV) replicase and vesicular stomatitis virus glycoprotein (VSV-G), have been proven safe and efficient as vaccine candidates. However, previous studies showed that VLVs containing rabies virus glycoprotein (RABV-G) grew at relatively low titers in cells, impeding their potential use as a rabies vaccine. In this study, we constructed novel VLVs by transfection of a mutant SFV RNA replicon encoding RABV-G. We found that these VLVs could self-propagate efficiently in cell culture and could evolve to high titers (approximately 108 focus-forming units [FFU]/ml) by extensive passaging 25 times in BHK-21 cells. Furthermore, we found that the evolved amino acid changes in SFV nonstructural protein 1 (nsP1) at positions 470 and 482 was critical for this high-titer phenotype. Remarkably, VLVs could induce robust type I interferon (IFN) expression in BV2 cells and were highly sensitive to IFN-α. We found that direct inoculation of VLVs into the mouse brain caused reduced body weight loss, mortality, and neuroinflammation compared with the RABV vaccine strain. Finally, it could induce increased generation of germinal center (GC) B cells, plasma cells (PCs), and virus-neutralizing antibodies (VNAs), as well as provide protection against virulent RABV challenge in immunized mice. This study demonstrated that VLVs containing RABV-G could proliferate in cells and were highly evolvable, revealing the feasibility of developing an economic, safe, and efficacious rabies vaccine. IMPORTANCE VLVs have been shown to represent a more versatile and superior vaccine platform. In previous studies, VLVs containing the Semliki Forest virus replicase (SFV nsP1 to nsP4) and rabies virus glycoprotein (RABV-G) grew to relatively low titers in cells. In our study, we not only succeeded in generating VLVs that proliferate in cells and stably express RABV-G, but the VLVs that evolved grew to higher titers, reaching 108 FFU/ml. We also found that nucleic acid changes at positions 470 and 482 in nsP1 were vital for this high-titer phenotype. Moreover, the VLVs that evolved in our studies were highly attenuated in mice, induced potent immunity, and protected mice from lethal RABV infection. Collectively, our study showed that high titers of VLVs containing RABV-G were achieved, demonstrating that these VLVs could be an economical, safe, and efficacious rabies vaccine candidate.


Subject(s)
Rabies Vaccines/immunology , Rabies/immunology , Vaccination/methods , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , Disease Models, Animal , Female , Genetic Engineering/methods , Germinal Center/immunology , Glycoproteins/genetics , Immunization/methods , Male , Mice , Mice, Inbred ICR , Rabies/metabolism , Rabies Vaccines/metabolism , Rabies Vaccines/pharmacology , Rabies virus/immunology , Semliki forest virus/immunology , Vesiculovirus/genetics , Viral Proteins/genetics
4.
Int J Pharm ; 545(1-2): 318-328, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29746999

ABSTRACT

Plasmid DNA (pDNA) vaccines have the potential for protection against a wide range of diseases including rabies but are rapid in degradation and poor in uptake by antigen-presenting cells. To overcome the limitations, we fabricated a pDNA nanoparticulate vaccine. The negatively charged pDNA was adsorbed onto the surface of cationic PLGA (poly (d, l-lactide-co-glycolide))-chitosan nanoparticles and were used as a delivery vehicle. To create a hydrogel for sustainable vaccine release, we dispersed the pDNA nanoparticles in poloxamer 407 gel which is liquid at 4 °C and turns into soft gels at 37 °C, providing ease of administration and preventing burst release of pDNA. Complete immobilization of pDNA to cationic nanoparticles was achieved at a pDNA to nanoparticles ratio (P/N) of 1/50. Cellular uptake of nanoparticles was both time and concentration dependent and followed a saturation kinetics with Vmax of 11.389 µg/mL h and Km of 139.48 µg/mL. The in vitro release studies showed the nanoparticulate vaccine has a sustained release for up to 24 days. In summary, pDNA PLGA-chitosan nanoparticles were non-cytotoxic, their buffering capacity and cell uptake were enhanced, and sustained the release of pDNA. We expect our pDNA vaccine's potency will be greatly improved in the animal studies.


Subject(s)
Chitosan/chemistry , Drug Carriers , Lactic Acid/chemistry , Nanoparticles , Poloxamer/chemistry , Polyglycolic Acid/chemistry , Rabies Vaccines/chemistry , Animals , Cell Line , Chitosan/toxicity , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Compounding , Drug Liberation , Drug Stability , Electrophoretic Mobility Shift Assay , Hydrogels , Kinetics , Lactic Acid/toxicity , Mice, Inbred C57BL , Nanotechnology , Poloxamer/toxicity , Polyglycolic Acid/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , Rabies Vaccines/administration & dosage , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Solubility , Spectroscopy, Fourier Transform Infrared , Surface Properties , Technology, Pharmaceutical/methods , Vaccines, DNA/chemistry , Vaccines, DNA/immunology
5.
Clin. transl. oncol. (Print) ; 19(7): 785-792, jul. 2017.
Article in English | IBECS | ID: ibc-163433

ABSTRACT

To review the literature about the use of Rabies Virus-Vaccine (RV-V) as an anticancer immunotherapeutic modality in the light of recent findings. The literature search in relevant databases with the following key words: Rabies virus, cancer, remission. Remissions occured following RV-V injections in patients with cervical cancer and melanoma. Pilot clinical studies showed that RV-V injections enhanced survival in glioblastoma patients, which is supported by findings in GL261 mouse glioma model. If public health studies demonstrate protective role of RV-V against certain types of cancers, it can be benefitted as a novel immune adjuvant in clinic (AU)


No disponible


Subject(s)
Humans , Rabies Vaccines/administration & dosage , Rabies Vaccines/metabolism , Glioblastoma/drug therapy , Glioblastoma/prevention & control , Immunotherapy/methods , Neoplasm Regression, Spontaneous , Rabies/prevention & control , Rabies virus/isolation & purification , Immunity, Cellular
6.
J Control Release ; 239: 19-26, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27524283

ABSTRACT

Because humans get rabies primarily through dog bites, stray dog population control and mass or mandatory vaccination of domestic dogs and other animals has virtually eliminated human rabies in industrialized countries. However, thousands of people in developing countries die of rabies each year due to the inability to control dog populations and implement mass vaccination because of financial, logistical and other challenges. The availability of an easier-to-administer and more cost-effective vaccine may help to address some of these issues. Here, we propose the use of dissolving microneedle patches for simple and potentially cost-effective rabies vaccination, and assess the safety and immunogenicity of microneedle patch vaccination using a rabies DNA vaccine in dogs. The vaccine was stable upon formulation and storage for at least 3weeks at 4°C in a microneedle patch. For vaccination, the patches were applied to the inner ear by hand without an applicator. Microneedle patches were well tolerated in the skin, with mild erythema, minimal wheal formation and complete resolution of skin reactions within 7days, and generated no systemic adverse events. Microneedle patches were at least as immunogenic as intramuscular injection at the same dose, as demonstrated by similar serum neutralizing antibody titers. A ten-fold lower vaccine dose administered by microneedle patch generated a weaker immune response compared to full-dose intramuscular vaccination. We conclude that dissolving microneedle patches may provide an innovative approach to mass vaccination of dogs.


Subject(s)
Microinjections/methods , Needles , Rabies Vaccines/administration & dosage , Transdermal Patch , Vaccination/methods , Animals , Antibodies, Viral/blood , CHO Cells , Cricetinae , Cricetulus , Dogs , Female , Male , Microinjections/instrumentation , Rabies/metabolism , Rabies/prevention & control , Rabies Vaccines/metabolism , Skin Absorption/drug effects , Skin Absorption/physiology , Vaccines, DNA/administration & dosage , Vaccines, DNA/metabolism
7.
J Immunol ; 195(9): 4358-68, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26408670

ABSTRACT

Much of our understanding of CNS immunity has been gained from models involving pathological inflammation. Attenuated rabies viruses (RABV) are unique tools to study CNS immunity in the absence of conventional inflammatory mechanisms, as they spread from the site of inoculation to the CNS transaxonally, thereby bypassing the blood-brain barrier (BBB), and are cleared without neutrophil or monocyte infiltration. To better understand the role of CD4 T cell subsets in the clearance of the virus from CNS tissues, we examined the development of antiviral immunity in wild-type (WT) and T-bet knockout mice (T-bet(-/-)), which lack Th1 cells. Early control of RABV replication in the CNS tissues of WT mice is associated with the production of IFN-γ, with antiviral effects likely mediated through the enhanced expression of type I IFNs. Of interest, IFN-α and -γ are overexpressed in the infected T-bet(-/-) by comparison with WT CNS tissues, and the initial control of RABV infection is similar. Ultimately, attenuated RABV are cleared from the CNS tissues of WT mice by Ab locally produced by the activities of infiltrating T and B cells. Although T and B cell infiltration into the CNS of infected T-bet(-/-) mice is comparable, their activities are not, the consequence being delayed, low-level Ab production and prolonged RABV replication. More importantly, neither T-bet(-/-) mice immunized with an attenuated virus, nor WT mice with Th2 RABV-specific immunity induced by immunization with inactivated virus, are protected in the long term against challenge with a pathogenic RABV.


Subject(s)
Central Nervous System/immunology , Rabies virus/immunology , Rabies/immunology , T-Box Domain Proteins/immunology , Animals , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Central Nervous System/metabolism , Central Nervous System/virology , Flow Cytometry , Gene Expression/immunology , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Rabies/metabolism , Rabies/virology , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Rabies virus/metabolism , Rabies virus/physiology , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/virology , Time Factors , Vaccines, Attenuated/immunology , Vaccines, Attenuated/metabolism
8.
Virus Genes ; 50(2): 299-302, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724175

ABSTRACT

Rabies virus (RABV) can cause a fatal neurological disease in human and animals, and vaccines were generally applied for the immunoprophylaxis of rabies. Here, a recombinant viral vector carrying the exogenous gene expression component between phosphoprotein (P) and matrix protein (M) genes of RABV was constructed based on the vaccine strain SRV9 used in China. To develop a reverse genetic system, the full-length cDNA plasmids of SRV9 were constructed using the eukaryotic expression vector pCI or pcDNA3.1(+). However, recovery efficiency based on the pcDNA3.1 vector was significantly higher than that of the pCI vector. The exogenous gene expression component PE-PS-BsiWI-PmeI or PS-BsiWI-PmeI-PE was introduced in different locations between the P and M genes of SRV9. When the enhanced green fluorescent protein (eGFP) was used as a reporter gene, both locations could rescue recombinant RABV (rRABV) expressing eGFP with high efficiency. Characterization of rRABV expressing eGFP in vitro revealed that its growth was similar to that of the parental virus. Animal experiments showed that rRABV expressing eGFP could replicate and express eGFP in the brains of suckling mice. Furthermore, rRABV of SRV9 was nonpathogenic for 3-week-old mice and could be cleared from the central nervous system at 5 days post-inoculation. Our results showed that the recombinant SRV9 virus could be used as a useful viral vector for exogenous gene expression.


Subject(s)
Gene Expression , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Rabies Vaccines/genetics , Rabies virus/genetics , Animals , Female , Genetic Vectors/metabolism , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Plasmids/genetics , Plasmids/metabolism , Rabies Vaccines/metabolism , Rabies virus/metabolism , Reverse Genetics
9.
J Proteome Res ; 12(1): 481-90, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23163834

ABSTRACT

Rabies is a rapidly progressive lyssavirus encephalitis that is statistically 100% fatal. There are no clinically effective antiviral drugs for rabies. An immunologically naïve teenager survived rabies in 2004 through improvised supportive care; since then, 5 additional survivors have been associated with use of the so-called Milwaukee Protocol (MP). The MP applies critical care focused on the altered metabolic and physiologic states associated with rabies. The aim of this study was to examine the metabolic profile of cerebrospinal fluid (CSF) from rabies patients during clinical progression of rabies encephalitis in survivors and nonsurvivors and to compare these samples with control CSF samples. Unsupervised clustering algorithms distinguished three stages of rabies disease and identified several metabolites that differentiated rabies survivors from those who subsequently died, in particular, metabolites related to energy metabolism and cell volume control. Moreover, for those patients who survived, the trajectory of their metabolic profile tracked toward the control profile and away from the rabies profile. NMR metabolomics of human rabies CSF provide new insights into the mechanisms of rabies pathogenesis, which may guide future therapy of this disease.


Subject(s)
Metabolomics , Rabies virus , Rabies , Adolescent , Adult , Antiviral Agents/cerebrospinal fluid , Antiviral Agents/immunology , Female , Gene Expression Profiling , Humans , Male , Rabies/cerebrospinal fluid , Rabies/drug therapy , Rabies/immunology , Rabies/metabolism , Rabies/pathology , Rabies Vaccines/cerebrospinal fluid , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Rabies Vaccines/therapeutic use , Rabies virus/drug effects , Rabies virus/immunology , Rabies virus/pathogenicity
10.
Virol J ; 9: 319, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23270301

ABSTRACT

BACKGROUND: During the vaccination campaigns, puppies younger than 3 months old are not targeted and remain unvaccinated for at least the first year of their lives. Almost half of the reported rabid dogs are 6 months or younger. Hence, we should recommend the vaccination against rabies of young puppies. Unfortunately, owing to the exposure of puppies to infections with either canine parvovirus (CPV) or distemper virus (CDV) after the intervention of the vaccinators, owners are reluctant to vaccinate puppies against rabies. Therefore, it is necessary to include the CPV and CDV valences in the vaccine against rabies. Multivalent DNA-based vaccination in dogs, including rabies and distemper valences, could help in raising vaccine coverage. METHODS: We have designed monovalent and multivalent DNA-based vaccine candidates for in vitro and in vivo assays. These plasmids encode to the rabies virus glycoprotein and/or the canine distemper virus hemagglutinin. The first strategy of multivalent DNA-based vaccination is by mixing plasmids encoding to a single antigen each. The second is by simply fusing the genes of the antigens together. The third is by adding the foot and mouth disease virus (FMDV) 2A oligopeptide gene into the antigen genes. The last strategy is by the design and use of a bicistronic plasmid with an "Internal Ribosome Entry Site" (IRES) domain. RESULTS: The monovalent construct against canine distemper was efficiently validated by inducing higher humoral immune responses compared to cell-culture-derived vaccine both in mice and dogs. All multivalent plasmids efficiently expressed both valences after in vitro transfection of BHK-21 cells. In BALB/c mice, the bicistronic IRES-dependant construct was the most efficient inducer of virus-neutralizing antibodies against both valences. It was able to induce better humoral immune responses compared to the administration of either cell-culture-derived vaccines or monovalent plasmids. The FMDV 2A was also efficient in the design of multivalent plasmids. CONCLUSIONS: In a single shot, the design of efficient multivalent plasmids will be very beneficial for DNA-based vaccination against numerous diseases.


Subject(s)
Distemper Virus, Canine/immunology , Distemper/prevention & control , Rabies/prevention & control , Rabies/veterinary , Vaccination/methods , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Chlorocebus aethiops , Distemper/genetics , Distemper/immunology , Distemper/virology , Dogs/immunology , Dogs/virology , Female , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Gene Fusion , Genes, Viral , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Hemagglutinins, Viral/metabolism , Immunity, Humoral , Mice , Mice, Inbred BALB C , Plasmids/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Rabies/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Rabies virus/immunology , Transfection , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vero Cells , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
11.
Vopr Virusol ; 57(3): 44-7, 2012.
Article in Russian | MEDLINE | ID: mdl-22905428

ABSTRACT

The paper describes a new principle of the selection of the rabies virus vaccine strain ERA-CB20M based on quantitation of the expression of glycoprotein, a major immunogen of rabies virus. There is a correlation between the level of glycoprotein expression and that of vaccine virus attenuation. The application of the new principle for vaccine virus selection will permit a safer and more immunogenic rabies vaccine to be prepared.


Subject(s)
Glycoproteins/immunology , Rabies Vaccines , Rabies virus , Rabies , Animals , Cattle , Gene Expression Regulation, Viral/immunology , Glycoproteins/metabolism , Humans , Mice , Rabies/prevention & control , Rabies/virology , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Rabies virus/immunology , Rabies virus/metabolism , Rabies virus/pathogenicity , Rats
12.
Protein J ; 31(6): 447-56, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22592449

ABSTRACT

Rabies virus surface glycoprotein (rabies G-protein) with (G+RS) and without (G-RS) endoplasmic reticulum retrieval signal was expressed and characterized in tobacco plants. Transgenically expressed rabies G-protein was estimated at 0.015-0.38 % of total leaf protein. The relative migration of the rabies G-protein on SDS-PAGE was at the position, as anticipated for the viral coat protein (~66 kDa). Immunolocalization by confocal microscopy established that immunoprotective G+RS expressed in tobacco was primarily confined to ER. G+RS showed binding to Con A lectin and was susceptible to N-glycosidase F activity similar to native rabies G-protein. However, the G-RS transgenically expressed in tobacco leaves was glycosylated differently and was resitant to N-glycosidase F. Immunological studies and Rapid Fluorescent Foci Inhibition Test (RFFIT) showed that G+RS was immunogenic and immunoprotective, whereas G-RS was moderately immunogenic but non-protective against live virus challenge. Hence, plants can express the antigenic component of rabies virus with suitable glycosylation, which is important to give protection against rabies virus infection.


Subject(s)
Antigens, Viral/immunology , Endoplasmic Reticulum/metabolism , Glycoproteins/immunology , Nicotiana/genetics , Plant Leaves/genetics , Rabies Vaccines/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/genetics , Antigens, Viral/metabolism , Escherichia coli , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Mice , Mice, Inbred BALB C , Plant Leaves/metabolism , Plants, Genetically Modified , Protein Processing, Post-Translational , Protein Sorting Signals , Protein Transport , Rabies/prevention & control , Rabies Vaccines/genetics , Rabies Vaccines/metabolism , Rabies virus/immunology , Nicotiana/metabolism , Vaccination , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
13.
Biologicals ; 38(5): 586-93, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20682459

ABSTRACT

Rabies virus glycoprotein (RVG) is a major structural protein and antigen of rabies virus that induces a highly immunogenic response. In the present study, we have used 2A self-cleaving sequence of the foot-and-mouth disease virus (FMDV) to express double or triple copies of the RVG from a single open reading frame derived from human adenovirus 5 (AdHu5). The recombinant adenoviruses produce similar virus titers, indicating that the insertion of double or triple copies of the RVG gene linked with the FMDV 2A sequence does not affect virus replication. The RVG was efficiently expressed by constructs containing the 2A sequence and retained its antigenic property. The 2A self-cleaving peptide mediated efficient generation of individual glycoprotein in transient expression assay and did not lead to an altered surface distribution of RVG. Flow cytometry demonstrated that the expression levels of RVG were improved in recombinant Ads carrying multiple RVG gene copies. We conclude that ribosome skipping induced by the FMDV 2A sequence is an effective strategy to express multiple glycoprotein genes of rabies virus in adenoviruses and 2A-containing recombinant Ads may represent an attractive alternative to other coexpression strategies for multiple gene expression.


Subject(s)
Gene Dosage/genetics , Rabies Vaccines/genetics , Rabies virus/genetics , Viral Fusion Proteins/genetics , Adenoviridae/genetics , Animals , Cells, Cultured , Cloning, Molecular , Cricetinae , DNA Replication/genetics , Gene Expression Regulation , Genetic Vectors/genetics , Humans , Protein Processing, Post-Translational/genetics , Protein Sorting Signals/genetics , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Viral Proteins/genetics
14.
J Virol ; 81(13): 7041-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17459937

ABSTRACT

The nonpathogenic phenotype of the live rabies virus (RV) vaccine SPBNGAN is determined by an Arg-->Glu exchange at position 333 in the glycoprotein, designated GAN. We recently showed that after several passages of SPBNGAN in mice, an Asn-->Lys mutation arose at position 194 of GAN, resulting in GAK, which was associated with a reversion to the pathogenic phenotype. Because an RV vaccine candidate containing two GAN genes (SPBNGAN-GAN) exhibits increased immunogenicity in vivo compared to the single-GAN construct, we tested whether the presence of two GAN genes might also enhance the probability of reversion to pathogenicity. Comparison of SPBNGAN-GAN with RVs constructed to contain either both GAN and GAK genes (SPBNGAN-GAK and SPBNGAK-GAN) or two GAK genes (SPBNGAK-GAK) showed that while SPBNGAK-GAK was pathogenic, SPBNGAN-GAN and SPBNGAN-GAK were completely nonpathogenic and SPBNGAK-GAN showed strongly reduced pathogenicity. Analysis of genomic RV RNA in mouse brain tissue revealed significantly lower virus loads in SPBNGAN-GAK- and SPBNGAK-GAN-infected brains than those detected in SPBNGAK-GAK-infected brains, indicating the dominance of the nonpathogenic phenotype determined by GAN over the GAK-associated pathogenic phenotype. Virus production and viral RNA synthesis were markedly higher in SPBNGAN-, SPBNGAK-GAN-, and SPBNGAN-GAK-infected neuroblastoma cells than in the SPBNGAK- and SPBNGAK-GAK-infected counterparts, suggesting control of GAN dominance at the level of viral RNA synthesis. These data point to the lower risk of reversion to pathogenicity of a recombinant RV carrying two identical GAN genes compared to that of an RV carrying only a single GAN gene.


Subject(s)
Genes, Dominant , Genes, Viral , Glycoproteins/metabolism , Rabies Vaccines/metabolism , Rabies virus/metabolism , Rabies/metabolism , Viral Proteins/metabolism , Amino Acid Substitution , Animals , Brain/metabolism , Brain/virology , Cell Line , Glycoproteins/genetics , Male , Mice , Mutation, Missense , RNA, Viral/biosynthesis , RNA, Viral/genetics , Rabies/genetics , Rabies Vaccines/genetics , Rabies virus/genetics , Rabies virus/pathogenicity , Viral Load , Viral Proteins/genetics
15.
Hum Vaccin ; 1(5): 187-90, 2005.
Article in English | MEDLINE | ID: mdl-17033270

ABSTRACT

A meta-analysis was done to study the relationship between antigenecity and immunogenecity of human rabies vaccines. The data of ten cell culture human rabies vaccine studies conducted at a single centre during 1993-2004 were used in the study. The vaccines studied included Purified Chick Embryo Cell Vaccine (Kaketsuken, Japan and Rabipur, India), Purified Vero cell Rabies Vaccine (Verorab, France), Human Diploid Cell Vaccine (MIRV, France and Rabivax, Adsorbed and Lyophilized, India) and Rhesus Diploid Rabies Vaccine (adsorbed, USA). Interestingly, it was revealed that an higher antigenecity of rabies vaccines viz. potency of > or = 5 IU per single intramuscular dose did not result in significantly higher immunogenecity, as measured by rabies virus neutralizing antibody (RVNA) titers in the vaccinees, both on day 14 (t = 0.42, p > 0.66, GMR = 1.06, 95% CI of GMR = 0.82, 1.37) and day 90 (t = 0.80, p > 0.43, GMR = 1.15, 95% CI of GMR = 0.74, 1.14). However, as there are no reports of meta-analysis of cell culture human rabies vaccine trials, to confirm this observation the authors recommend further studies in this regard.


Subject(s)
Antigens, Viral/immunology , Immunocompetence/immunology , Rabies Vaccines/immunology , Adolescent , Adult , Animals , Antibodies, Viral/immunology , Cells, Cultured , Chick Embryo , Child , Clinical Trials, Phase III as Topic , Clinical Trials, Phase IV as Topic , Female , Humans , Immunization Schedule , Injections, Intramuscular , Male , Middle Aged , Neutralization Tests/methods , Rabies Vaccines/administration & dosage , Rabies Vaccines/metabolism , Time Factors
16.
Biologicals ; 30(1): 1-6, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11846424

ABSTRACT

In vaccines produced in eukaryote cells as well as in commercial medical preparations of leukocyte interferon a number of cytokines such as IL-1beta, IL-6 and TNF-alpha have been detected. Among the vaccines examined in this study the highest level of IL-1beta was demonstrated in inactivated hepatitis A vaccine prepared in the green monkey kidney cell line 4647, that of IL-6 in inactivated rabies vaccine produced in Syrian hamster kidney (SHK) cell culture, and that of TNF-alpha in live poliomyelitis vaccine manufactured in VERO cells. A spontaneous and poliovirus-induced capacity of cell cultures to produce cytokines was detected. The level of cytokines produced depend on the kind of cell culture and the type of virus, a more pronounced effect being generated by types 1 and 2 poliovirus as compared with type 3. The presence of highly active cytokines in virus vaccines and interferon preparations points to the necessity of investigating the influence of the presence of cytokines on the biological activity of these preparations and to the advisability of standardizing and controlling the cytokine content.


Subject(s)
Cytokines/biosynthesis , Interferons/metabolism , Leukocytes/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Hepatitis A Vaccines/metabolism , Humans , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Kidney/metabolism , Mesocricetus , Poliovirus/metabolism , Poliovirus Vaccines/metabolism , Rabies Vaccines/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Vero Cells
17.
Clin Infect Dis ; 34(3): 404-6, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11774089

ABSTRACT

Storing freshly reconstituted purified chick embryo rabies vaccine at 4 degrees C for 1 week allowed use of <1 ampoule of 1.0 mL for 1 patient for day 0, 3, and 7 immunizations, representing considerable savings in vaccine and also possibly allowing use of this cost-saving regimen in centers that see <1 rabies-exposed patient daily. The 90-day booster dose mandated in the current intradermal regimen may not be necessary if the day 28 dose is doubled, eliminating 1 clinic visit at no additional cost in vaccine.


Subject(s)
Health Care Costs , Rabies Vaccines/administration & dosage , Drug Stability , Drug Storage , Humans , Immunization, Secondary , Rabies Vaccines/metabolism , Temperature , World Health Organization
18.
Adv Enzyme Regul ; 34: 145-58, 1994.
Article in English | MEDLINE | ID: mdl-7942272

ABSTRACT

Cellular and humoral immune responses to vaccines of hepatitis B and rabies as antigens were suppressed by specific inhibitors of cathepsin B, anti-cathepsin B antibody and the specific substrate of cathepsin B. The antigenic peptides of these vaccines are processed by cathepsin B and the fragments are capable of binding with the desetope of MHC class II, beta-chain, because one of the active sites of cathepsin B (14, 15) VN217-222 shares high homology with a part of the desetope, VN57-62, of MHC class II, beta-chain. Rechallenge of the synthesized antigenic peptides of these vaccine molecules shows a strong proliferative response to the splenocyte primed by these vaccines. However, the response to these antigenic peptides was not inhibited by cathepsin B inhibitors. These findings suggest that cathepsin B inhibitors do not inhibit any other processes of immune responses than the proteolytic processing of antigens. Some investigators reported recently that the Ii-chain is degraded by purified cathepsin B in vitro (23-25). However, we showed that the suppression of these immune responses by cathepsin B inhibitors is not due to the inhibition of invariant chain degradation. We found that the invariant chain shares about 40% homology with the cystatin family which are the endogenous inhibitors of cysteine proteases (23, 24). Therefore, the Ii-chain is one of the members of the cystatin superfamily and may participate in the regulation of presentation of antigenic peptides and also antigen processing by cathepsin B.


Subject(s)
Antigen Presentation , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Histocompatibility Antigens Class II/metabolism , Peptide Fragments/metabolism , T-Lymphocytes/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Antigen Presentation/drug effects , Cathepsin C , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , HLA-DR7 Antigen/genetics , Hepatitis B Vaccines/immunology , Hepatitis B Vaccines/metabolism , Immunosuppression Therapy , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Models, Biological , Molecular Sequence Data , Protein Binding , Rabies Vaccines/immunology , Rabies Vaccines/metabolism , Sequence Homology, Amino Acid , Viral Vaccines/metabolism
19.
Vaccine ; 8(5): 425-37, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2251869

ABSTRACT

The use of vaccinia virus vector for the delivery of antigens was first described by Moss and Paoletti and their colleagues in 1982. Such vaccines could be of particular value in developing countries because they would be cheap, stable, easy to administer and provide long-lasting immunity. WHO recognized the potential value of such a delivery system by convening two meetings, one at the National Institutes of Health, USA in November 1984 and the second at WHO headquarters, Geneva, Switzerland in September 1985, to discuss the possibility of using such products, particularly with regard to their safety. Since that time, other vehicles which could be useful for the delivery of antigens have been described. These include Escherichia coli, Salmonella typhimurium, BCG, all other poxviruses, adenovirus, herpesvirus and poliovirus. At its meeting in July 1988, the Scientific Advisory Group of Experts of the Programme for Vaccine Development (SAGE) concluded that it was appropriate to discuss the general topic of live vectors and proceeded to arrange a meeting to discuss the present position and to prepare a report on the following key issues: requirements for safety and efficacy; immunological factors which may influence efficacy; medical constraints on use. The present report results from a meeting held in Geneva from 19 to 22 June 1989.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Viral/immunology , Vaccines, Synthetic/therapeutic use , Adenoviridae/immunology , Animals , BCG Vaccine/genetics , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , HIV-1/immunology , Humans , Poliovirus Vaccine, Inactivated/genetics , Poliovirus Vaccine, Inactivated/immunology , Rabies Vaccines/adverse effects , Rabies Vaccines/metabolism , Rinderpest virus/immunology , Salmonella/immunology , Salmonella/pathogenicity , Vaccination/adverse effects , Vaccinia virus/immunology , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...