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











Publication year range
1.
BMC Vet Res ; 17(1): 184, 2021 May 04.
Article in English | MEDLINE | ID: mdl-33947419

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea (PED) is a contagious intestinal disease caused by porcine epidemic diarrhea virus (PEDV) characterized by vomiting, diarrhea, anorexia, and dehydration, which have caused huge economic losses around the world. At present, vaccine immunity is still the most effective method to control the spread of PED. In this study, we have constructed a novel recombinant L. casei-OMP16-PEDVS strain expressing PEDVS protein of PEDV and OMP16 protein of Brucella abortus strain. To know the immunogenicity of the recombinant L. casei-OMP16-PEDVS candidate vaccine, it was compared with BL21-OMP16-PEDVS-F, BL21-OMP16-PEDVS, and BL21-PEDVS recombinant protein. RESULTS: The results showed that we could detect higher levels of IgG, neutralizing antibody, IL-4, IL-10, and INF-γ in serum and IgA in feces of L. casei-OMP16-PEDVS immunized mice, which indicated that L. casei-OMP16-PEDVS candidate vaccine could induce higher levels of humoral immunity, cellular immunity, and mucosal immunity. CONCLUSION: Therefore, L. casei-OMP16-PEDVS is a promising candidate vaccine for prophylaxis of PEDV infection.


Subject(s)
Brucella abortus/genetics , Coronavirus Infections/prevention & control , Lacticaseibacillus casei/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology , Animals , Antibodies, Neutralizing , Antibodies, Viral/immunology , Brucella abortus/metabolism , Coronavirus Infections/immunology , Female , Immunity, Cellular , Immunity, Humoral , Immunity, Mucosal , Immunization , Lacticaseibacillus casei/metabolism , Mice, Inbred BALB C , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
2.
BMC Vet Res ; 17(1): 164, 2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33853597

ABSTRACT

BACKGROUND: Since 2011, numerous highly virulent and antigenic variant viral strains have been reported in pigs that were vaccinated against the swine pseudorabies virus. These infections have led to substantial economic losses in the Chinese swine industry. RESULTS: This study, constructed a novel recombinant vaccine strain with gI/gE deletion (PRV-GD2013-ΔgI/gE) by overlapping PCR and homologous recombination technology. The growth curves and plaque morphology of the recombinant virus were similar to those of the parental strain. However, PRV-GD2013-ΔgI/gE infection was significantly attenuated in mice compared with that of PRV-GD2013. Two-week-old piglets had normal rectal temperatures and displayed no clinical symptoms after being inoculated with 105 TCID50 PRV-GD2013-ΔgI/gE, indicating that the recombinant virus was avirulent in piglets. Piglets were immunized with different doses of PRV-GD2013-ΔgI/gE, or a single dose of Bartha-K61 or DMEM, and infected with PRV-GD2013 at 14 days post-vaccination. Piglets given high doses of PRV-GD2013-ΔgI/gE showed no obvious clinical symptoms, and their antibody levels were higher than those of other groups, indicating that the piglets were completely protected from PRV-GD2013. CONCLUSIONS: The PRV-GD2013-ΔgI/gE vaccine strain could be effective for immunizing Chinese swine herds against the pseudorabies virus (PRV) strain.


Subject(s)
Pseudorabies Vaccines/immunology , Pseudorabies/prevention & control , Swine Diseases/virology , Animals , Cell Line , Cricetinae , Female , Gene Deletion , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/immunology , Homologous Recombination , Mice, Inbred BALB C , Polymerase Chain Reaction , Pseudorabies/immunology , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control , Vaccines, Synthetic/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
3.
Microbiol Immunol ; 64(2): 153-161, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31710119

ABSTRACT

Development of a serotyping-capable dengue detection test is hampered by the absence of an identified unique marker that can detect specific dengue virus (DENV) serotype. In the current commercially available antibody-capture diagnostic methods, immobilized nonstructural 1 (NS1) antigen indiscriminately binds and detects immunoglobulin M or immunoglobulin G against any serotype, thus limiting its capability to distinguish existing serotypes of dengue. Identification of dengue serotype is important because certain serotypes are associated with severe forms of dengue as well as dengue hemorrhagic fever. In this study, we aimed to identify an immunogenic epitope unique to DENV2 NS1 antigen and determine the binding specificity of its synthetic peptide mimotope to antibodies raised in animal models. Selection of a putative B-cell epitope from the reported DENV2 NS1 antigen was done using Kolaskar and Tongaonkar Antigenicity prediction, Emini surface accessibility prediction, and Parker hydrophilicity prediction available at the immune epitope database and analysis resource. Uniqueness of the B-cell epitope to DENV2 was analyzed by BLASTp. Immunogenicity of the synthetic peptide analog of the predicted immunogenic epitope was tested in rabbits. The binding specificity of the antibodies raised in animals and the synthetic peptide mimotope was tested by indirect ELISA. A synthetic peptide analog comprising the unique epitope of DENV2 located at the 170th-183rd position of DENV2 NS1 was found to be immunogenic in animal models. The antipeptide antibody produced in rabbits showed specific binding to the synthetic peptide mimotope of the predicted unique DENV2 NS1 immunogenic epitope.


Subject(s)
Dengue Virus/immunology , Dengue/diagnosis , Vaccines, Synthetic/virology , Viral Nonstructural Proteins , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Formation , Antigens, Viral/immunology , Computer Simulation , Dengue/immunology , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Humans , Immunoglobulin G , Rabbits , Serogroup , Vaccines, Synthetic/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology
4.
Viruses ; 11(9)2019 08 25.
Article in English | MEDLINE | ID: mdl-31450681

ABSTRACT

Newcastle disease (ND) is responsible for significant economic losses in the poultry industry. The disease is caused by virulent strains of Avian avulavirus 1 (AAvV-1), a species within the family Paramyxoviridae. We developed a recombinant construct based on the herpesvirus of turkeys (HVT) as a vector expressing two genes: F and HN (HVT-NDV-F-HN) derived from the AAvV-1 genotype VI ("pigeon variant" of AAvV-1). This recombinant viral vaccine candidate was used to subcutaneously immunize one group of specific pathogen-free (SPF) chickens and two groups of broiler chickens (20 one-day-old birds/group). Humoral immune response was evaluated by hemagglutination-inhibition test and enzyme-linked immunosorbent assay (ELISA). The efficacy of the immunization was assessed in two separate challenge studies performed at 6 weeks of age with the use of virulent AAvV-1 strains representing heterologous genotypes IV and VII. The developed vaccine candidate elicited complete protection in SPF chickens since none of the birds became sick or died during the 2-week observation period. In the broiler groups, 90% and 100% clinical protection were achieved after challenges with AAvV-1 of IV and VII genotypes, respectively. We found no obvious relationship between antibody levels and protection assessed in broilers in the challenge study. The developed recombinant HVT-NDV-F-HN construct containing genes from a genotype VI AAvV-1 offers promising results as a potential vaccine candidate against ND in chickens.


Subject(s)
HN Protein/immunology , Immunization/veterinary , Newcastle disease virus , Vaccines, Synthetic/immunology , Viral Fusion Proteins/immunology , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Chickens/virology , Cross Protection , Genes, Viral , HN Protein/biosynthesis , HN Protein/genetics , Hemagglutination Inhibition Tests , Herpesvirus 1, Meleagrid/genetics , Herpesvirus 1, Meleagrid/immunology , Herpesvirus 1, Meleagrid/metabolism , Immunity, Heterologous , Newcastle Disease/immunology , Newcastle Disease/prevention & control , Newcastle disease virus/genetics , Newcastle disease virus/immunology , Poultry Diseases/virology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, Synthetic/virology , Viral Fusion Proteins/biosynthesis , Viral Fusion Proteins/genetics , Viral Vaccines/biosynthesis , Viral Vaccines/immunology
5.
J Vet Med Sci ; 81(2): 314-320, 2019 Feb 28.
Article in English | MEDLINE | ID: mdl-30584200

ABSTRACT

Duck Tembusu virus disease, caused by the duck Tembusu virus (DTMUV), can lead to a severe reduction in egg production and growth retardation in laying ducks and ducklings, respectively. In this study, we engineered a novel recombinant adenovirus expressing the E protein of DTMUV (rAd-E) in AAV-293 cells (analyzed by western blot and indirect immunofluorescence assays). Intramuscular immunization of Cherry Valley ducks with rAd-E was performed to evaluate host cellular and humoral immune responses. Compared to the phosphate-buffered saline administered group and the negative control wild-type adenovirus (wtAd) group, the rAd-E vaccinated group showed increased cellular and humoral responses. The results from the cytokine release and lymphocyte proliferation assays showed that rAd-E induced a stronger cellular immune response than the control group (P<0.01), 4 weeks after primary immunization. The results of enzyme-linked immunosorbent and virus neutralization assays showed that rAd-E induced higher titers of specific neutralizing antibodies, 2 weeks after primary immunization. The DTMUV challenge experiment showed a higher survival rate (80%) of ducks in the rAd-E group, when challenged with 0.5 ml (ELD50=10-2.67/0.2 ml) of the DTMUV strain AH-F10. These results indicate that rAd-E effectively protects ducks against DTMUV infection. Therefore, rAd-E could be a vaccine candidate to provide an effective and safe method for prevention and control of DTMUV infection.


Subject(s)
Adenoviridae/immunology , Ducks/virology , Flavivirus Infections/veterinary , Flavivirus/immunology , Poultry Diseases/virology , Vaccines, Synthetic/genetics , Viral Vaccines/genetics , Adenoviridae/genetics , Animals , Blotting, Western/veterinary , Ducks/immunology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Fluorescent Antibody Technique/veterinary , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Neutralization Tests/veterinary , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology , Viral Vaccines/immunology
6.
Avian Dis ; 60(1): 22-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26953940

ABSTRACT

Domestic ducks are the second most abundant poultry species in many Asian countries and have played a critical role in the epizootiology of H5N1 highly pathogenic avian influenza (HPAI).In this study, the protective efficacy of a live recombinant vector vaccine based on a turkey herpesvirus (HVT) expressing the H5 gene from a clade 2.2 H5N1 HPAI strain (A/Swan/Hungary/4999/ 2006) (rHVT-H5/2.2), given at 3 days of age, was examined in Pekin ducks (Anas platyrhynchos domesticus). The vaccine was given alone or in combination with an inactivated H5N1 clade 2.3.2.1 reverse genetic (rgGD/2.3.2.1) vaccine given at 16 days of age, either as a single vaccination or in a prime-boost regime. At 30 days of age, ducks were challenged with one of two H5N1 HPAI viruses: A/duck/Vietnam/NCVD-2721/2013 (clade 1.1.2) or A/duck/Vietnam/NCVD-1584/2012 (clade 2.3.2.1.C). These viruses produced 100% mortality in less than 5 days in nonvaccinated control ducks. Ducks vaccinated with the rgGD/2.3.2.1 vaccine, with or without the rHVT-H5/2.2 vaccine, were 90%-100% protected against mortality after challenge with either of the two H5N1 HPAI viruses. The rHVT-H5/2.2 vaccine alone, however, conferred only 30% protection against mortality after challenge with either H5N1 HPAI virus; the surviving ducks from these groups shed higher amount of virus and for longer than the single-vaccinated rgGD/2.3.2.1 group. Despite low protection, ducks vaccinated with the rHVT-H5/2.2 vaccine and challenged with the clade 1.1.2 Vietnam virus had a longer mean death time than nonvaccinated controls (P = 0.02). A booster effect was found on reduction of virus shedding when using both vaccines, with lower oropharyngeal viral titers at 4 days after challenge with either HPAI virus (P < 0.05). Neither rHVT-H5/2.2 nor standard HVT vaccine could be detected in samples collected from multiple tissues at different time points, indicting minimal levels of viral replication. In conclusion, although a minor effect on survival was observed, this study demonstrates the suboptimal protection with the rHVT-H5/2.2 vaccine given alone in Pekin ducks against H5N1 HPAI viruses and only a minor additive effect on virus shedding reduction when used with an inactivated vaccine in a prime-boost regime.


Subject(s)
Ducks , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Herpesvirus 1, Meleagrid/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/immunology , Influenza in Birds/immunology , Poultry Diseases/immunology , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Herpesvirus 1, Meleagrid/metabolism , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/virology , Phylogeny , Poultry Diseases/virology , Sequence Analysis, DNA/veterinary , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology
7.
J Virol ; 89(23): 12070-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26401031

ABSTRACT

UNLABELLED: Current vaccines do not provide sufficient levels of protection against divergent porcine reproductive and respiratory syndrome virus (PRRSV) strains circulating in the field, mainly due to the substantial variation of the viral genome. We describe here a novel approach to generate a PRRSV vaccine candidate that could confer unprecedented levels of heterologous protection against divergent PRRSV isolates. By using a set of 59 nonredundant, full-genome sequences of type 2 PRRSVs, a consensus genome (designated PRRSV-CON) was generated by aligning these 59 PRRSV full-genome sequences, followed by selecting the most common nucleotide found at each position of the alignment. Next, the synthetic PRRSV-CON strain was generated through the use of reverse genetics. PRRSV-CON replicates as efficiently as our prototype PRRSV strain FL12, both in vitro and in vivo. Importantly, when inoculated into pigs, PRRSV-CON confers significantly broader levels of heterologous protection than does wild-type PRRSV. Collectively, our data demonstrate that PRRSV-CON can serve as an excellent candidate for the development of a broadly protective PRRSV vaccine. IMPORTANCE: The extraordinary genetic variation of RNA viruses poses a monumental challenge for the development of broadly protective vaccines against these viruses. To minimize the genetic dissimilarity between vaccine immunogens and contemporary circulating viruses, computational strategies have been developed for the generation of artificial immunogen sequences (so-called "centralized" sequences) that have equal genetic distances to the circulating viruses. Thus far, the generation of centralized vaccine immunogens has been carried out at the level of individual viral proteins. We expand this concept to PRRSV, a highly variable RNA virus, by creating a synthetic PRRSV strain based on a centralized PRRSV genome sequence. This study provides the first example of centralizing the whole genome of an RNA virus to improve vaccine coverage. This concept may be significant for the development of vaccines against genetically variable viruses that require active viral replication in order to achieve complete immune protection.


Subject(s)
Genetic Variation , Immunity, Heterologous/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , Viral Vaccines/genetics , Animals , Base Sequence , Fluorescent Antibody Technique, Indirect , Molecular Sequence Data , Neutralization Tests , Sequence Alignment , Sequence Analysis, DNA , Swine , Vaccines, Synthetic/virology , Viral Plaque Assay , Viral Vaccines/immunology
8.
Mol Ther ; 23(6): 1022-1033, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25881000

ABSTRACT

The immunogenic properties of recombinant adeno-associated virus (rAAV) gene transfer vectors remain incompletely characterized in spite of their usage as gene therapy vectors or as vaccines. Molecular interactions between rAAV and various types of antigen-presenting cells (APCs), as well as the impact of these interactions on transgene or capsid-specific immunization remain unclear. We herein show that binding motifs recognized by the capsid and which determine the vector tissue tropism are also critical for key immune activation processes. Using rAAV capsid serotype 1 (rAAV1) vectors which primary receptors on target cells are α2,3 and α2,6 N-linked sialic acids, we show that sialic acid-dependent binding of rAAV1 on APCs is essential to trigger CD4(+) T-cell responses by increasing rAAV1 uptake and contributing to antigenic presentation of both the capsid and transgene product although this involves different APCs. In addition, the nanoparticulate structure of the vector in itself appears to be sufficient to trigger mobilization and activation of some APCs. Therefore, combinations of structural and of serotype-specific cell-targeting properties of rAAV1 determine its complex immunogenicity. These findings may be useful to guide a selection of rAAV variants depending on the intended level of immunogenicity for either gene therapy or vaccination applications.


Subject(s)
Dependovirus/genetics , Nanoparticles/chemistry , Serogroup , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Capsid/immunology , Dependovirus/immunology , Female , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Injections, Intramuscular , Lectins/immunology , Mice , Mice, Inbred C57BL , Nanospheres/chemistry , Nanospheres/virology , Principal Component Analysis , Transgenes , Vaccines, Synthetic/virology
9.
J Virol ; 88(4): 2056-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24307589

ABSTRACT

Infectious clone technologies allow the rational design of live attenuated viral vaccines with the possibility of vaccine-driven coexpression of immunomodulatory molecules for additional vaccine safety and efficacy. The latter could lead to novel strategies for vaccine protection against infectious diseases where traditional approaches have failed. Here we show for the flavivirus Murray Valley encephalitis virus (MVEV) that incorporation of the internal ribosome entry site (IRES) of Encephalomyocarditis virus between the capsid and prM genes strongly attenuated virulence and that the resulting bicistronic virus was both genetically stable and potently immunogenic. Furthermore, the novel bicistronic genome organization facilitated the generation of a recombinant virus carrying an beta interferon (IFN-ß) gene. Given the importance of IFNs in limiting virus dissemination and in efficient induction of memory B and T cell antiviral immunity, we hypothesized that coexpression of the cytokine with the live vaccine might further increase virulence attenuation without loss of immunogenicity. We found that bicistronic mouse IFN-ß coexpressing MVEV yielded high virus and IFN titers in cultured cells that do not respond to the coexpressed IFN. However, in IFN response-sufficient cell cultures and mice, the virus produced a self-limiting infection. Nevertheless, the attenuated virus triggered robust innate and adaptive immune responses evidenced by the induced expression of Mx proteins (used as a sensitive biomarker for measuring the type I IFN response) and the generation of neutralizing antibodies, respectively. IMPORTANCE The family Flaviviridae includes a number of important human pathogens, such as Dengue virus, Yellow fever virus, Japanese encephalitis virus, West Nile virus, and Hepatitis C virus. Flaviviruses infect large numbers of individuals on all continents. For example, as many as 100 million people are infected annually with Dengue virus, and 150 million people suffer a chronic infection with Hepatitis C virus. However, protective vaccines against dengue and hepatitis C are still missing, and improved vaccines against other flaviviral diseases are needed. The present study investigated the effects of a redesigned flaviviral genome and the coexpression of an antiviral protein (interferon) on virus replication, pathogenicity, and immunogenicity. Our findings may aid in the rational design of a new class of well-tolerated and safe vaccines.


Subject(s)
Cloning, Molecular/methods , Encephalitis Virus, Murray Valley/genetics , Encephalomyocarditis virus/genetics , Immunity, Cellular/immunology , Ribosomes/genetics , Vaccines, Synthetic/genetics , Viral Vaccines/biosynthesis , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , DNA Primers/genetics , Encephalitis Virus, Murray Valley/pathogenicity , Genetic Engineering/methods , Immunohistochemistry , Interferon-beta/metabolism , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Myxovirus Resistance Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, Synthetic/virology , Vero Cells , Viral Vaccines/genetics
10.
Avian Pathol ; 42(3): 215-20, 2013.
Article in English | MEDLINE | ID: mdl-23607580

ABSTRACT

We developed a transgenic potato (TrP/R7) expressing the recombinant R7 (rR7) antigen for use as an oral vaccine to protect against a chicken protozoan disease, chicken leucocytozoonosis. The TrP/R7 potato was produced by Agrobacterium tumefaciens-mediated transformation and regeneration, and the R7 gene insertion into potato chromosomes was confirmed by genomic polymerase chain reaction and Southern hybridization. rR7 antigen expression in TrP/R7 potato was also confirmed by sandwich enzyme-linked immunosorbent assay and western blotting using an antibody against the second-generation schizont of Leucocytozoon caulleryi. A transgenic potato clone with the highest rR7 antigen expression (3 µg rR7 antigen per gram of fresh-weight potato leaves) was selected, cultivated, and used in oral administration experiments to examine its ability to boost immunity. Chickens were immunized with chicken leucocytozoonosis vaccine "Hokken" by injection, and chickens that developed moderate levels of antibody titres were fed with TrP/R7 leaves. Chickens fed with TrP/R7 leaves showed increased antibody responses. In contrast, chickens fed with non-transgenic potato leaves showed a continuous decrease in antibody titres. Furthermore, chickens fed with TrP/R7 potato leaves showed strong resistance against experimental challenge with L. caulleryi infection. This study demonstrates the use of a plant-based oral vaccine to boost immunity against a protozoan disease.


Subject(s)
Haemosporida , Immunization, Secondary/veterinary , Plants, Genetically Modified/chemistry , Poultry Diseases/prevention & control , Poultry Diseases/parasitology , Protozoan Infections, Animal/prevention & control , Vaccines, Synthetic/virology , Administration, Oral , Animals , Antigens, Protozoan/immunology , Blotting, Southern/veterinary , Blotting, Western/veterinary , Chickens , DNA Primers/genetics , Plant Leaves/immunology , Polymerase Chain Reaction/veterinary , Solanum tuberosum/genetics , Vaccines, Synthetic/administration & dosage
11.
Bioengineered ; 4(6): 374-8, 2013.
Article in English | MEDLINE | ID: mdl-23337287

ABSTRACT

In pursuit of better influenza vaccines, many strategies are being studied worldwide. An attractive alternative is the generation of a broadly cross-reactive vaccine based on the induction of cytotoxic T-lymphocytes (CTL) directed against conserved internal antigens of influenza A virus. The feasibility of this approach using recombinant viral vectors has recently been demonstrated in mice and humans by several research groups. However, similar results might also be achieved through immunization with viral proteins expressed in a prokaryotic system formulated with the appropriate adjuvants and delivery systems. This approach would be much simpler and less expensive. Recent results from several laboratories seem to confirm this is as a valid option to be considered.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/pharmacology , Animals , Cross Reactions , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors , Humans , Influenza A virus/immunology , Vaccination , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology , Viral Proteins/immunology
12.
J Med Primatol ; 36(2): 80-94, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17493138

ABSTRACT

BACKGROUND: The immunogenicity and protective efficacy of recombinant modified vaccinia virus Ankara (rMVA) vectors expressing structural (gag/pol, env) and regulatory (tat, rev, nef) genes of SIVmac251/32H-J5 (rMVA-J5) were assessed. METHODS: Immunization with rMVA constructs (2.5 x 10(7) IU) 32, 20 and 8 weeks pre-challenge was compared with 32 and 20 weeks but with a final boost 8 weeks pre-challenge with 2 x 10(6) fixed-inactivated HSC-F4 cells infected with SIVmac32H. Controls received rMVA vectors expressing an irrelevant transgene or were naïve challenge controls. All received 10 MID(50) SIVmac32H/J5 intravenously. RESULTS: Vaccinates immunized with rMVA-J5 exhibited significant, albeit transient, control of peak primary viraemia despite inconsistent and variable immune responses elicted by vaccination. Humoral and cellular responses to Env were most consistent, with lower responses to Nef, Rev and Tat. Increasing titres of anti-vaccinia neutralizing antibodies reflected the number and dose of rMVA inoculations. CONCLUSIONS: Improved combinations of viral vectors are required to elicit appropriate immune responses to control viral replication.


Subject(s)
Acquired Immunodeficiency Syndrome/prevention & control , Macaca fascicularis/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Vaccination/veterinary , Vaccinia virus/immunology , Animals , Antibodies, Viral/immunology , Flow Cytometry , Genetic Vectors , In Situ Hybridization , RNA, Viral/blood , T-Lymphocytes, Cytotoxic/immunology , Transgenes/genetics , Vaccines, Synthetic/virology , Viral Proteins/metabolism
13.
AIDS Res Hum Retroviruses ; 23(2): 278-86, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17331034

ABSTRACT

Oral immunization is a safe and easily applicable route to induce mucosal immunity to HIV infection. We examined the ability of oral attenuated Salmonella typhimurium (ST) vaccine expressing Gag for the efficiency of generating Gag-specific mucosal IgA and CD8+ T cell responses in intestinal lymphoid tissues. By optimizing the codon of HIV-1 gag to the preferred codon bias of Salmonella, the expression of Gag in Salmonella was dramatically improved. The oral ST-Gag vaccine by itself was not so powerful and induces little Gag-specific CD8+ T cell responses in the intestine. Nevertheless, we found that it potentiates otherwise weak intestinal CD8+ T cell responses in nasally primed mice with Gag p24 and cholera toxin adjuvant. Thus, the oral delivery of Salmonella expressing Gag would be utilized in combination with other parenteral vaccine to direct and strengthen intestinal HIV-specific CTL responses.


Subject(s)
AIDS Vaccines/immunology , Gene Products, gag/immunology , HIV Infections/prevention & control , HIV-1/immunology , Intestinal Mucosa/immunology , Viral Vaccines/pharmacology , AIDS Vaccines/biosynthesis , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Gene Products, gag/biosynthesis , HIV Infections/immunology , Immunization/methods , Mice , Mice, Inbred BALB C , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Salmonella typhimurium/metabolism , Salmonella typhimurium/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/biosynthesis , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/virology , Viral Vaccines/biosynthesis
14.
Vaccine ; 23(29): 3808-13, 2005 May 31.
Article in English | MEDLINE | ID: mdl-15893618

ABSTRACT

The safety and efficacy of a canarypox vector expressing PrM and E genes of West Nile virus (WNV) (ALVAC-WNV) was evaluated in dogs and cats. One group of 17 dogs (vaccinated with 10(5.6) TCID(50)) and two groups of cats (groups 1 [n=14] vaccinated with 10(7.5) TCID(50) and 2 [n=8] 10(5.6) TCID(50)) were vaccinated twice at 28-day intervals. Fifteen dogs and eleven cats served as negative controls. The cats and dogs were challenged 120 and 135 days after the second immunization, respectively via the bites of Aedes albopictus mosquitoes infected with WNV. The first dose of vaccine induced a detectable antibody response in four dogs and five cats (one immunized with low and four with high doses). After the second dose, all the vaccinated dogs and all of the cats, immunized with high dose had detectable antibody titers, whereas only four of eight cats in the low dose group were seropositive. None of the vaccinated dogs and one vaccinated cat developed viremia following the WNV mosquito-challenge. In contrast, 14 of the 15 control dogs and 9 of the 11 control cats developed viremia. The experimental vaccine described in this study may be of value in the prevention of WNV infection in dogs and cats.


Subject(s)
Cat Diseases/prevention & control , Dog Diseases/prevention & control , Viral Vaccines/immunology , West Nile Fever/veterinary , West Nile virus/immunology , Animals , Antibodies, Viral/blood , Canarypox virus/genetics , Cats , Dogs , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/adverse effects , Viral Vaccines/genetics , West Nile Fever/prevention & control , West Nile virus/genetics
15.
J Infect Dis ; 191(7): 1093-104, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15747245

ABSTRACT

BACKGROUND: Recombination technology can be used to create live attenuated respiratory syncytial virus (RSV) vaccines that contain combinations of known attenuating mutations. METHODS: Two live attenuated, recombinantly derived RSV vaccine candidates, rA2cp248/404 Delta SH and rA2cp248/404/1030 Delta SH, were evaluated in 31 adults and in 95 children >/=6 months old. rA2cp248/404/1030 Delta SH was subsequently evaluated in 44 infants 1-2 months old. These vaccine candidates share 4 attenuating genetic elements and differ only in a missense mutation (1030) in the polymerase gene. RESULTS: Both vaccines were highly attenuated in adults and RSV-seropositive children and were well tolerated and immunogenic in RSV-seronegative children. Compared with that of rA2cp248/404 Delta SH, replication of rA2cp248/404/1030 Delta SH was restricted in RSV-seronegative children (mean peak titer, 10(4.3) vs. 10(2.5) plaque-forming units [pfu]/mL), indicating that the 1030 mutation had a potent attenuating effect. Although rA2cp248/404/1030 Delta SH was well tolerated in infants, only 44% of infants who received two 10(5.3)-pfu doses of vaccine had detectable antibody responses. However, replication after administration of the second dose was highly restricted, indicating that protective immunity was induced. At least 4 of 5 attenuating genetic elements were retained in recovered vaccine viruses. CONCLUSIONS: rA2cp248/404/1030 Delta SH is the first RSV vaccine candidate to be sufficiently attenuated in young infants. Additional studies are needed to determine whether rA2cp248/404/1030 Delta SH can induce protective immunity against wild-type RSV.


Subject(s)
Antibodies, Viral/blood , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Adult , Amino Acid Substitution , Child, Preschool , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/immunology , Double-Blind Method , Genes, Viral , Humans , Infant , Mutation, Missense , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/physiology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Virus Replication , Virus Shedding
16.
J Virol Methods ; 116(1): 35-43, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14715305

ABSTRACT

Poliomyelitis outbreaks in areas that were free for a long time of wild-type polioviruses have been reported. Characterization at nucleotide level of the causative agents showed that the isolated viruses were recombinant oral polio vaccine (OPV)-derived polioviruses. To allow rapid identification and detailed analysis of such recombinant polioviruses, a robust full-length reverse transcriptase-PCR (RT-PCR) was developed using SuperScript II (RT) and expand (PCR). Without extensive purification, it was possible to amplify and characterize the full-length genomes of all selected vaccine, wild-type, and recombinant vaccine-derived polioviruses within a week. Endonuclease nuclease analysis (SpeI) of the full-length amplicons allowed easy discrimination between recombinant and non-recombinant polioviruses. Furthermore, sequence analysis of cloned full-length amplicons of a recombinant vaccine-derived poliovirus strain showed that the quasi-species nature of a viral stock is preserved during the RT-PCR procedure. This robust and rapid RT-PCR method will allow rapid characterization of (recombinant) poliovirus strains in case of a local poliomyelitis outbreak, and will help to assess the risk of the appearance of such strains after wild-type poliovirus has been eradicated globally.


Subject(s)
Genome, Viral , Poliovirus Vaccine, Oral , Poliovirus/genetics , Poliovirus/isolation & purification , RNA, Viral/genetics , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction/methods , Genes, Viral , Humans , Poliomyelitis/epidemiology , Poliomyelitis/virology , Poliovirus/classification , Polymorphism, Restriction Fragment Length , RNA, Viral/analysis , Vaccines, Synthetic/genetics , Vaccines, Synthetic/virology
17.
J Gen Virol ; 83(Pt 6): 1377-1386, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12029153

ABSTRACT

An efficient antigen expression system using a recombinant Sendai virus (SeV) has been established recently and its potential to induce resistance against immunodeficiency virus infections in macaques has been shown. SeV replication has been well characterized in mice, the natural host, but not in primates, including humans. Here, primary SeV replication was investigated in macaques. After intranasal immunization with a recombinant SeV expressing simian immunodeficiency virus Gag protein, SeV-Gag, robust gag expression was observed in the nasal mucosa and much lower but significant levels of gag expression were observed in the local retropharyngeal and submandibular lymph nodes (LN). Expression peaked within a week and lasted at least up to 13 days after immunization. SeV-Gag was isolated from nasal swabs consistently at day 4 but not at all at day 13. Gag expression was undetectable in the lung as well as in remote lymphoid tissues, such as the thymus, spleen and inguinal LN, indicating that the spread of the virus was more restricted in macaques than in mice. SeV-specific T cells were detectable in SeV-immunized macaques at day 7. Finally, no naive macaques showed significant levels of anti-SeV antibodies in the plasma, even after living in a cage together with an acutely SeV-infected macaque for 5 weeks, indicating that SeV transmission from SeV-infected macaques to naive ones was inefficient. None of the SeV-immunized macaques displayed appreciable clinical manifestations. These results support the idea that this system may be used safely in primates, including humans.


Subject(s)
Genetic Vectors/physiology , Macaca/virology , Sendai virus/physiology , Virus Replication , Animals , Antibodies, Viral/blood , Gene Products, gag/analysis , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , Genetic Vectors/genetics , Immunization , Lymph Nodes/metabolism , Macaca/blood , Macaca fascicularis , Macaca mulatta , Nasal Mucosa/virology , Recombination, Genetic , Respirovirus Infections/transmission , Sendai virus/genetics , Sendai virus/immunology , T-Lymphocytes/immunology , Vaccines, Synthetic/virology
18.
Curr Protoc Immunol ; Chapter 20: Unit 20.1, 2001 May.
Article in English | MEDLINE | ID: mdl-18432774

ABSTRACT

This unit details protocols for in vivo models of subcutaneous growth and pulmonary metastases of B16 melanoma. Therapeutic approaches include the use of B16.GM-CSF and rVVmTRP-1 to induce autoimmune vitiligo and tumor protection. The induction and use of gp 100-specific therapeutic cytotoxic T lymphocytes (CTL) are discussed. Methods are also included for CTL induction, isolation and testing, CTL maintenance, and adoptive transfer. Support protocols detail the testing of mouse sera for presence of MDA-specific antibodies by immunoblotting and ELISA, respectively. Additional sections, including growing B16 melanoma, enumerating pulmonary metastases, and use of recombinant viruses for vaccination, are discussed together with safety concerns.


Subject(s)
Disease Models, Animal , Melanoma, Experimental , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunotherapy , Lung Neoplasms/secondary , Melanocytes/cytology , Melanocytes/enzymology , Melanocytes/immunology , Melanocytes/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Mice , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology
19.
Mol Med Today ; 6(1): 28-35, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10637572

ABSTRACT

The application of modern molecular techniques has profoundly influenced our understanding of virus function. As a consequence, virus biology is being directly applied to medical research. It is a reflection of the current pace of virology that we are now beginning to think of our ancient foes as useful and beneficial tools.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , RNA Viruses/genetics , Vaccines, Synthetic/virology , Alphavirus/genetics , Animals , Flavivirus/genetics , Humans , Picornaviridae/genetics , Replicon
SELECTION OF CITATIONS
SEARCH DETAIL