Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 134: 182-191, 2016 10.
Article in English | MEDLINE | ID: mdl-27637905

ABSTRACT

The control of smallpox was achieved using live vaccinia virus (VV) vaccine, which successfully eradicated the disease worldwide. As the variola virus no longer exists as a natural infection agent, mass vaccination was discontinued after 1980. However, emergence of smallpox outbreaks caused by accidental or deliberate release of variola virus has stimulated new research for second-generation vaccine development based on attenuated VV strains. Considering the closely related animal poxviruses that also arise as zoonoses, and the increasing number of unvaccinated or immunocompromised people, a safer and more effective vaccine is still required. With this aim, new vectors based on avian poxviruses that cannot replicate in mammals should improve the safety of conventional vaccines, and protect from zoonotic orthopoxvirus diseases, such as cowpox and monkeypox. In this study, DNA and fowlpox (FP) recombinants that expressed the VV L1R, A27L, A33R, and B5R genes were generated (4DNAmix, 4FPmix, respectively) and tested in mice using novel administration routes. Mice were primed with 4DNAmix by electroporation, and boosted with 4FPmix applied intranasally. The lethal VVIHD-J strain was then administered by intranasal challenge. All of the mice receiving 4DNAmix followed by 4FPmix, and 20% of the mice immunized only with 4FPmix, were protected. The induction of specific humoral and cellular immune responses directly correlated with this protection. In particular, higher anti-A27 antibodies and IFNγ-producing T lymphocytes were measured in the blood and spleen of the protected mice, as compared to controls. VVIHD-J neutralizing antibodies in sera from the protected mice suggest that the prime/boost vaccination regimen with 4DNAmix plus 4FPmix may be an effective and safe mode to induce protection against smallpox and poxvirus zoonotic infections. The electroporation/intranasal administration routes contributed to effective immune responses and mouse survival.


Subject(s)
Antibodies, Neutralizing/blood , Electroporation , Fowlpox/genetics , Smallpox Vaccine/administration & dosage , Vaccination/methods , Vaccinia virus/genetics , Animals , Immunity, Cellular , Immunity, Humoral , Interferon-gamma/blood , Interferon-gamma/immunology , Mice , Mpox (monkeypox)/prevention & control , Neutralization Tests , Smallpox/prevention & control , Smallpox Vaccine/genetics , Smallpox Vaccine/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/pathogenicity , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
2.
Sci Total Environ ; 539: 395-400, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26372942

ABSTRACT

Dreissena polymorpha is a widespread filter-feeder species, resistant to a broad range of environmental conditions and different types of pollutants,which has recently colonized Italian freshwaters. Although widely used to monitor pollution in freshwater environments, this species is also an important food source for some fish and water birds. It can also be used to concentrate or remove particulate organic matter to interrupt avian-to-human transmission of pollutants and control health risks for animals and humans. In this study, the accumulation/inactivation in D. polymorpha of human health-related spiked enteric viruses was described. The removal of endogenous Escherichia coli, the classical indicator of fecal contamination,was tested as well.Our preliminary lab-scale results demonstrate that zebra mussels can reduce significantly poliovirus titer after 24 h and rotavirus titer after 8 h. E. coli counts were also reduced in the presence of zebra mussels by about 1.5 log after 4 h and nearly completely after 24 h. The fate of the two enteric viruses after concentration by zebra mussels was also investigated after mechanical disruption of the tissues. To our knowledge, the accumulation from water and inactivation of human health-related enteric viruses by zebra mussels has never been reported.


Subject(s)
Dreissena/virology , Enterovirus , Environmental Monitoring , Escherichia coli , Fresh Water/microbiology , Fresh Water/virology , Water Pollutants/analysis , Animals , Biodegradation, Environmental , Dreissena/microbiology
3.
J Transl Med ; 13: 80, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25763880

ABSTRACT

BACKGROUND: Considering the high number of new cases of cervical cancer each year that are caused by human papilloma viruses (HPVs), the development of an effective vaccine for prevention and therapy of HPV-associated cancers, and in particular against the high-risk HPV-16 genotype, remains a priority. Vaccines expressing the E6 and E7 proteins that are detectable in all HPV-positive pre-cancerous and cancer cells might support the treatment of HPV-related lesions and clear already established tumors. METHODS: In this study, DNA and fowlpox virus recombinants expressing the E6F47R mutant of the HPV-16 E6 oncoprotein were generated, and their correct expression verified by RT-PCR, Western blotting and immunofluorescence. Immunization protocols were tested in a preventive or therapeutic pre-clinical mouse model of HPV-16 tumorigenicity using heterologous (DNA/FP) or homologous (DNA/DNA and FP/FP) prime/boost regimens. The immune responses and therapeutic efficacy were evaluated by ELISA, ELISPOT assays, and challenge with TC-1* cells. RESULTS: In the preventive protocol, while an anti-E6-specific humoral response was just detectable, a specific CD8(+) cytotoxic T-cell response was elicited in immunized mice. After the challenge, there was a delay in cancer appearance and a significant reduction of tumor volume in the two groups of E6-immunized mice, thus confirming the pivotal role of the CD8(+) T-cell response in the control of tumor growth in the absence of E6-specific antibodies. In the therapeutic protocol, in-vivo experiments resulted in a higher number of tumor-free mice after the homologous DNA/DNA or heterologous DNA/FP immunization. CONCLUSIONS: These data establish a preliminary indication for the prevention and treatment of HPV-related tumors by the use of DNA and avipox constructs as safe and effective immunogens following a prime/boost strategy. The combined use of recombinants expressing both E6 and E7 proteins might improve the antitumor efficacy, and should represent an important approach to control HPV-associated cancers.


Subject(s)
Cancer Vaccines/immunology , DNA, Recombinant/metabolism , Fowlpox/metabolism , Human papillomavirus 16/immunology , Immunization, Secondary , Neoplasms/immunology , Oncogene Proteins, Viral/genetics , Repressor Proteins/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Chick Embryo , Female , Humans , Immunity, Humoral/immunology , Mice, Inbred C57BL , Neoplasms/pathology , Transgenes , Vaccination , Virus Replication
4.
Antiviral Res ; 116: 67-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25660110

ABSTRACT

Human papilloma virus (HPV)-16 is the prevalent genotype associated with cervical tumours. Virus-like-particle (VLP)-based vaccines have proven to be effective in limiting new infections of high-risk HPVs, but their high cost has hampered their use, especially in the poor developing countries. Avipox-based recombinants are replication-restricted to avian species and represent efficient and safe vectors also for immunocompromised hosts, as they can elicit a complete immune response. A new fowlpox virus recombinant encoding HPV-L1 (FPL1) was engineered and evaluated side-by-side with a FP recombinant co-expressing L1 and green fluorescent protein (FPL1GFP) for correct expression of L1 in vitro in different cell lines, as confirmed by Western blotting, immunofluorescence, real-time PCR, and electron microscopy. Mice were also immunised to determine its immunogenicity. Here, we demonstrate that the FPL1 recombinant better expresses L1 in the absence of GFP, correctly assembles structured capsomers into VLPs, and elicits an immune response in a preclinical animal model. To our knowledge, this is the first report of HPV VLPs assembled in eukaryotic cells using an avipox recombinant.


Subject(s)
Capsid Proteins/immunology , Capsid Proteins/metabolism , Fowlpox virus/genetics , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/metabolism , Papillomavirus Vaccines/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Blotting, Western , Capsid Proteins/genetics , Cell Line , Fluorescent Antibody Technique , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Mice , Microscopy, Electron , Oncogene Proteins, Viral/genetics , Papillomavirus Vaccines/genetics , Real-Time Polymerase Chain Reaction , Transgenes
5.
Virus Res ; 178(2): 374-82, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24050999

ABSTRACT

The first-generation smallpox vaccine was based on live vaccinia virus (VV) and it successfully eradicated the disease worldwide. Therefore, it was not administered any more after 1980, as smallpox no longer existed as a natural infection. However, emerging threats by terrorist organisations has prompted new programmes for second-generation vaccine development based on attenuated VV strains, which have been shown to cause rare but serious adverse events in immunocompromised patients. Considering the closely related animal poxviruses that might also be used as bioweapons, and the increasing number of unvaccinated young people and AIDS-affected immunocompromised subjects, a safer and more effective smallpox vaccine is still required. New avipoxvirus-based vectors should improve the safety of conventional vaccines, and protect from newly emerging zoonotic orthopoxvirus diseases and from the threat of deliberate release of variola or monkeypox virus in a bioterrorist attack. In this study, DNA and fowlpox recombinants expressing the L1R, A27L, A33R and B5R genes were constructed and evaluated in a pre-clinical trial in mouse, following six prime/boost immunisation regimens, to compare their immunogenicity and protective efficacy against a challenge with the lethal VV IHD-J strain. Although higher numbers of VV-specific IFNγ-producing T lymphocytes were observed in the protected mice, the cytotoxic T-lymphocyte response and the presence of neutralising antibodies did not always correlate with protection. In spite of previous successful results in mice, rabbits and monkeys, where SIV/HIV transgenes were expressed by the fowlpox vector, the immune response elicited by these recombinants was low, and most of the mice were not protected.


Subject(s)
Fowlpox virus/genetics , Mpox (monkeypox)/prevention & control , Smallpox Vaccine/immunology , Vaccines, DNA/immunology , Vaccinia virus/immunology , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytotoxicity, Immunologic , Female , Genetic Vectors , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mpox (monkeypox)/immunology , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/genetics , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccinia virus/genetics , Viral Proteins/genetics
6.
J Transl Med ; 11: 95, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23578094

ABSTRACT

BACKGROUND: The traditional smallpox vaccine, administered by scarification, was discontinued in the general population from 1980, because of the absence of new smallpox cases. However, the development of an effective prophylactic vaccine against smallpox is still necessary, to protect from the threat of deliberate release of the variola virus for bioterrorism and from new zoonotic infections, and to improve the safety of the traditional vaccine. Preventive vaccination still remains the most effective control and new vectors have been developed to generate recombinant vaccines against smallpox that induce the same immunogenicity as the traditional one. As protective antibodies are mainly directed against the surface proteins of the two infectious forms of vaccinia, the intracellular mature virions and the extracellular virions, combined proteins from these viral forms can be used to better elicit a complete and protective immunity. METHODS: Four novel viral recombinants were constructed based on the fowlpox genetic background, which independently express the vaccinia virus L1 and A27 proteins present on the mature virions, and the A33 and B5 proteins present on the extracellular virions. The correct expression of the transgenes was determined by RT-PCR, Western blotting, and immunofluorescence. RESULTS AND CONCLUSIONS: Using immunoprecipitation and Western blotting, the ability of the proteins expressed by the four novel FPL1R, FPA27L, FPA33R and FPB5R recombinants to be recognized by VV-specific hyperimmune mouse sera was demonstrated. By neutralisation assays, recombinant virus particles released by infected chick embryo fibroblasts were shown not be recognised by hyperimmune sera. This thus demonstrates that the L1R, A27L, A33R and B5R gene products are not inserted into the new viral progeny. Fowlpox virus replicates only in avian species, but it is permissive for entry and transgene expression in mammalian cells, while being immunologically non-cross-reactive with vaccinia virus. These recombinants might therefore represent safer and more promising immunogens that can circumvent neutralisation by vector-generated immunity in smallpox-vaccine-experienced humans.


Subject(s)
Fowlpox virus/genetics , Smallpox Vaccine/genetics , Vaccines, Synthetic/genetics , Vaccinia virus/genetics , Viral Vaccines/genetics , Animals , Chick Embryo , Chlorocebus aethiops , Fibroblasts/metabolism , Genes, Viral , Humans , Mice , Microscopy, Fluorescence , Neutralization Tests , Smallpox Vaccine/immunology , Transgenes , Vaccines, Synthetic/immunology , Vero Cells
7.
J Virol Methods ; 187(1): 172-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23000750

ABSTRACT

The development of an effective prophylactic vaccine is still necessary to improve the safety of the conventional although-discontinued smallpox vaccine, and to protect from the threat of deliberate release of variola virus. This need also arises from the number of new cases of animal orthopoxvirus infections each year, and to reduce the risk to animal handlers. Fowlpox (FP) recombinants only replicate in avian species and have been developed against human infectious diseases, as they can elicit an effective immune response, are not cross-reactive immunologically with vaccinia, and represent safer and more promising immunogens for immunocompromised individuals. The aim of this study was the characterisation of two new fowlpox recombinants expressing the A33R vaccinia virus gene either alone (FP(A33R)) or with the green fluorescent protein (FP(A33R-GFP)) to verify whether GFP can affect the expression of the transgene. The results show that both FP(A33R) and FP(A33R-GFP) can express A33R correctly, but A33R mRNA and protein synthesis are higher by FP(A33R) than by FP(A33R-GFP). Therefore, GFP co-expression does not prevent, but can reduce the level of a vaccine protein, and may affect the protective efficacy of the immune response.


Subject(s)
Fowlpox virus/genetics , Fowlpox virus/metabolism , Green Fluorescent Proteins/genetics , Membrane Glycoproteins/genetics , Recombinant Proteins/biosynthesis , Viral Envelope Proteins/genetics , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , Fowlpox virus/immunology , Green Fluorescent Proteins/biosynthesis , Humans , Membrane Glycoproteins/biosynthesis , Orthopoxvirus/immunology , Poxviridae Infections/immunology , Poxviridae Infections/prevention & control , Protein Biosynthesis , RNA, Messenger/biosynthesis , Recombinant Proteins/genetics , Vaccinia virus/immunology , Vero Cells , Viral Envelope Proteins/biosynthesis , Virus Replication
8.
Virus Res ; 170(1-2): 44-52, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22951311

ABSTRACT

Development of effective therapeutic vaccines against human papilloma virus (HPV) infections remains a priority, considering the high number of new cases of cervical cancer each year by high-risk HPVs, in particular by HPV-16. Vaccines expressing the E7 oncoprotein, which is detectable in all HPV-positive pre-cancerous and cancer cells, might clear already established tumors and support the treatment of HPV-related lesions. In this study, DNA or fowlpox virus recombinants expressing the harmless variant E7GGG of the HPV-16 E7 oncoprotein (DNA(E7GGG) and FP(E7GGG)) were generated. Two immunization regimens were tested in a pre-clinical mouse model by homologous (FP/FP) or heterologous (DNA/FP) prime-boost protocols to evaluate the immune response and therapeutic efficacy of the proposed HPV-16 vaccine. Low levels of anti-E7-specific antibodies were elicited after immunization, and in vivo experiments resulted in a higher number of tumor-free mice after the heterologous immunization. These results establish a preliminary indication for therapy of HPV-related tumors by the combined use of DNA and avipox recombinants, which might represent safer immunogens than vaccinia-based vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Fowlpox virus/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/therapeutic use , Vaccines, DNA/therapeutic use , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line , Female , Gene Expression , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Humans , Immunity, Cellular , Immunity, Humoral , Immunization Schedule , Immunotherapy , Mice , Mutant Proteins/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/virology , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/genetics , Plasmids/genetics , Transgenes
9.
J Transl Med ; 9: 190, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22053827

ABSTRACT

BACKGROUND: Human papilloma virus (HPV)-16 is the most prevalent high-risk mucosal genotype. Virus-like-particle (VLP)-based immunogens developed recently have proven to be successful as prophylactic HPV vaccines, but are still too expensive for developing countries. Although vaccinia viruses expressing the HPV-16 L1 protein (HPV-L1) have been studied, fowlpox-based recombinants represent efficient and safer vectors for immunocompromised hosts due to their ability to elicit a complete immune response and their natural host-range restriction to avian species. METHODS: A new fowlpox virus recombinant encoding HPV-L1 (FPL1) was engineered and evaluated for the correct expression of HPV-L1 in vitro, using RT-PCR, immunoprecipitation, Western blotting, electron microscopy, immunofluorescence, and real-time PCR assays. RESULTS: The FPL1 recombinant correctly expresses HPV-L1 in mammalian cells, which are non-permissive for the replication of this vector. CONCLUSION: This FPL1 recombinant represents an appropriate immunogen for expression of HPV-L1 in human cells. The final aim is to develop a safe, immunogenic, and less expensive prophylactic vaccine against HPV.


Subject(s)
Capsid Proteins/metabolism , Fowlpox virus/genetics , Genetic Techniques , Human papillomavirus 16/metabolism , Oncogene Proteins, Viral/metabolism , Recombination, Genetic/genetics , Animals , Blotting, Western , Capsid Proteins/genetics , Cell Line , Fluorescent Antibody Technique , Fowlpox virus/ultrastructure , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Mammals , Oncogene Proteins, Viral/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Time Factors , Virion/metabolism , Virion/ultrastructure
10.
Antiviral Res ; 88(1): 53-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20643163

ABSTRACT

Canarypox and fowlpox viruses represent alternative vaccine vectors due to their natural host-range restriction to avian species. Although they cannot replicate in mammals, they correctly express transgenes in human cells and elicit a complete immune response in vaccinated subjects. Several studies have evaluated their genomic differences and protective efficacy in preclinical trials, but detailed information is not available for their transgene expression, cytokine modulation and abortive replication in mammals. This study demonstrates that the heterologous HIV gag/pol and env genes are more efficiently expressed by fowlpox in non-immune and immune cells. The production of retrovirus-like particles, the longer transgene expression, and a balanced cytokine induction may confer to fowlpox-based recombinants the ability to elicit a better immune response.


Subject(s)
AIDS Vaccines , Canarypox virus , Fowlpox virus , Genetic Vectors , HIV-1/genetics , Vaccines, Synthetic , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , B7-1 Antigen/analysis , Canarypox virus/genetics , Canarypox virus/immunology , Canarypox virus/physiology , Canarypox virus/ultrastructure , Cell Line , Cytokines/immunology , Dendritic Cells/immunology , Fluorescent Antibody Technique , Fowlpox virus/genetics , Fowlpox virus/immunology , Fowlpox virus/physiology , Fowlpox virus/ultrastructure , Gene Expression , Genes, env , Genes, gag , Genes, pol , HIV-1/immunology , Humans , Immunization , Macrophages/immunology , Microscopy, Electron, Transmission , Transgenes , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virus Replication/genetics
11.
Arch Virol ; 155(6): 915-24, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20379750

ABSTRACT

Due to their natural host-range restriction to avian species, canarypox virus (CP) and fowlpox virus (FP) represent efficient and safe vaccine vectors, as they correctly express transgenes in human cells, elicit complete immune responses, and show protective efficacy in preclinical animal models. At present, no information is available on the differences in the abortive replication of these two avipox viruses in mammalian cells. In the present study, the replicative cycles of CP and FP, wild-type and recombinants, are compared in permissive and non-permissive cells, using transmission electron microscopy. We demonstrate that in non-permissive cells, the replicative cycle is more advanced in FP than in CP, that human cells, whether immune or not, are less permissive to avipox replication than monkey cells, and that the presence of virus-like particles only occurs after FP infection. Overall, these data suggest that the use of FP recombinants is more appropriate than the use of CP for eliciting an immune response.


Subject(s)
Canarypox virus/ultrastructure , Fowlpox virus/ultrastructure , Genetic Vectors , Vaccines, Synthetic , Animals , Canarypox virus/genetics , Canarypox virus/immunology , Canarypox virus/physiology , Cell Line , Fowlpox virus/genetics , Fowlpox virus/immunology , Fowlpox virus/physiology , Genetic Vectors/genetics , Genetic Vectors/immunology , Genetic Vectors/physiology , Genetic Vectors/ultrastructure , Humans , Microscopy, Electron, Transmission , Transgenes/physiology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/ultrastructure , Virion/physiology , Virion/ultrastructure , Virus Replication
12.
J Transl Med ; 8: 40, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20409340

ABSTRACT

BACKGROUND: Around half million new cases of cervical cancer arise each year, making the development of an effective therapeutic vaccine against HPV a high priority. As the E6 and E7 oncoproteins are expressed in all HPV-16 tumour cells, vaccines expressing these proteins might clear an already established tumour and support the treatment of HPV-related precancerous lesions. METHODS: Three different immunisation regimens were tested in a pre-clinical trial in rabbits to evaluate the humoral and cell-mediated responses of a putative HPV-16 vaccine. Fowlpoxvirus (FP) recombinants separately expressing the HPV-16 E6 (FPE6) and E7 (FPE7) transgenes were used for priming, followed by E7 protein boosting. RESULTS: All of the protocols were effective in eliciting a high antibody response. This was also confirmed by interleukin-4 production, which increased after simultaneous priming with both FPE6 and FPE7 and after E7 protein boost. A cell-mediated immune response was also detected in most of the animals. CONCLUSION: These results establish a preliminary profile for the therapy with the combined use of avipox recombinants, which may represent safer immunogens than vaccinia-based vectors in immuno-compromised individuals, as they express the transgenes in most mammalian cells in the absence of a productive replication.


Subject(s)
Fowlpox virus/genetics , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/therapy , Animals , Antibody Formation/immunology , Antibody Specificity/immunology , Cell Extracts , Cell Line, Tumor , Chickens , Female , Humans , Mice , Rabbits , Recombination, Genetic , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Uterine Cervical Neoplasms/virology
13.
J Virol Methods ; 158(1-2): 184-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19428588

ABSTRACT

Human papilloma virus (HPV)-16 is the most prevalent high-risk mucosal genotype and the expression of the E6 and E7 proteins, which can bind to the p53 and p105Rb host cell-cycle regulatory proteins, is related to its tumorigenicity. Virus-like-particle (VLP)-based immunogens developed recently are successful as prophylactic HPV vaccines. However, given the high number of individuals infected already with HPV and the absence of expression of the L1 structural protein in HPV-infected or HPV-transformed cells, an efficient therapeutic vaccine targeting the non-structural E6 and E7 oncoproteins is required. In this study, two new fowlpox virus (FPV) recombinants encoding the HPV-16 E6 and E7 proteins were engineered and evaluated for their correct expression in vitro, with the final aim of developing a therapeutic vaccine against HPV-related cervical tumors. Although vaccinia viruses expressing the HPV-16 and HPV-18 E6 and E7 oncoproteins have already been studied, due to their natural host-range restriction to avian species and their ability to elicit a complete immune response, FPV recombinants may represent efficient and safer vectors also for immunocompromised hosts. The results indicate that FPV recombinants can express correctly the E6 and E7 oncoproteins, and they should represent appropriate vectors for the expression of these oncoproteins in human cells.


Subject(s)
Fowlpox virus/genetics , Gene Expression , Genetic Vectors , Oncogene Proteins, Viral/genetics , Papillomavirus Vaccines/genetics , Repressor Proteins/genetics , Animals , Cell Line , Humans , Papillomavirus E7 Proteins , Vaccines, Synthetic/genetics
14.
J Virol Methods ; 155(1): 77-81, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18955084

ABSTRACT

Although several techniques are available to evaluate cell-mediated immunity, numerous difficulties have prevented their use in rabbits. Cytotoxic T-lymphocyte (CTL) assays have been used to determine the ex vivo cytolytic activity of CD8+ T-lymphocytes in immunization protocols. However, this assay cannot be performed with rabbit peripheral blood mononuclear cell (PBMC) targets because of their high spontaneous (51)Cr release. To overcome this intrinsic difficulty shown by rabbit cells, syngeneic normal and SV40-immortalized cells were prepared from skin biopsies. The results show that: (i) skin-derived rabbit fibroblasts can be used as target cells after infection with a fowlpox virus recombinant; (ii) SV40-immortalized skin fibroblasts appear to be more appropriate for repeated assays; (iii) antigen-expanded T-cells and fresh PBMCs can be used as effectors with a similar efficiency; and (iv) dissociation of adherent skin fibroblast target cells with EDTA is to be preferred over TrypLE enzymatic treatment.


Subject(s)
Cytotoxicity Tests, Immunologic , Epidermis/immunology , Epidermis/virology , Major Histocompatibility Complex/physiology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Transformed , Cell Transformation, Viral , Epidermal Cells , Fibroblasts/immunology , Fibroblasts/virology , Fowlpox virus/genetics , Fowlpox virus/immunology , Lymphocyte Activation , Rabbits , Recombination, Genetic , Simian virus 40/physiology , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...