Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Virol J ; 10: 229, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23842430

ABSTRACT

Vaccinia virus protein A33 (A33VACV) plays an important role in protection against orthopoxviruses, and hence is included in experimental multi-subunit smallpox vaccines. In this study we show that single-dose vaccination with recombinant Sindbis virus expressing A33VACV, is sufficient to protect mice against lethal challenge with vaccinia virus WR (VACV-WR) and ectromelia virus (ECTV) but not against cowpox virus (CPXV), a closely related orthopoxvirus. Moreover, a subunit vaccine based on the cowpox virus A33 ortholog (A33CPXV) failed to protect against cowpox and only partially protected mice against VACV-WR challenge. We mapped regions of sequence variation between A33VACV and A33CPXVand analyzed the role of such variations in protection. We identified a single protective region located between residues 104-120 that harbors a putative H-2Kd T cell epitope as well as a B cell epitope - a target for the neutralizing antibody MAb-1G10 that blocks spreading of extracellular virions. Both epitopes in A33CPXV are mutated and predicted to be non-functional. Whereas vaccination with A33VACV did not induce in-vivo CTL activity to the predicted epitope, inhibition of virus spread in-vitro, and protection from lethal VACV challenge pointed to the B cell epitope highlighting the critical role of residue L118 and of adjacent compensatory residues in protection. This epitope's critical role in protection, as well as its modifications within the orthopoxvirus genus should be taken in context with the failure of A33 to protect against CPXV as demonstrated here. These findings should be considered when developing new subunit vaccines and monoclonal antibody based therapeutics against orthopoxviruses, especially variola virus, the etiologic agent of smallpox.


Subject(s)
Cowpox virus/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Membrane Glycoproteins/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Adaptive Immunity , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Drug Carriers , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Variation , Genetic Vectors , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Sindbis Virus/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage
2.
Immunology ; 130(4): 527-35, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20331473

ABSTRACT

The protective efficacy and immunogenicity of a chimeric peptide against West Nile virus (WNV) was evaluated. This virus is the aetiological agent of West Nile fever, which has recently emerged in the western hemisphere. The rapid spread of WNV throughout North America, as well as the constantly changing epidemiology and transmission of the virus by blood transfusion and transplantation, have raised major public-health concerns. Currently, there are no effective treatments for WNV or vaccine for human use. We previously identified a novel, continuous B-cell epitope from domain III of the WNV envelope protein, termed Ep15. To test whether this epitope can protect against WNV infection, we synthesized a linear chimeric peptide composed of Ep15 and the heat-shock protein 60 peptide, p458. The p458 peptide is an effective carrier peptide for subunit vaccines against other infectious agents. We now report that mice immunized with the chimeric peptide, p458-Ep15, were resistant to lethal challenges with three different WNV strains. Moreover, their brains were free of viral genome and infectious virus. Mice immunized with Ep15 alone or with p431-Ep15, a control conjugate, were not protected. The chimeric p458-Ep15 peptide induced WNV-specific immunoglobulin G antibodies that neutralized the virus and induced the secretion of interferon-gammain vitro. Challenge of chimeric peptide-immunized mice considerably enhanced WNV-specific neutralizing antibodies. We conclude that this chimeric peptide can be used for formulation of a human vaccine against WNV.


Subject(s)
Chaperonin 60/immunology , Gene Products, env/immunology , Recombinant Fusion Proteins/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , Antibodies, Viral/immunology , Brain/virology , Cells, Cultured , Chlorocebus aethiops , Epitopes/immunology , Female , Genome, Viral , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Peptides/immunology , Spleen/immunology , Spleen/metabolism , West Nile Fever/virology
3.
BMC Infect Dis ; 9: 18, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19222853

ABSTRACT

BACKGROUND: West Nile Virus (WNV) is endemic in Israel and a significant level of antibodies is present in the population due to natural exposure. Anecdotal cases suggested that the presence of anti-WNV antibodies in intravenous immunoglobulin (IVIG) from Israeli donors (IVIG-IL) assisted the recovery of patients with severe WNV infection. METHODS: To enhance the therapeutic efficacy of IVIG-IL against WNV infection, OMRIX Biopharmaceuticals, Israel, have developed a strategy for selection of plasma units from a 10% fraction of Israeli blood donors with anti-WNV antibodies. Positive units were processed into pharmaceutical grade WNV IVIG (WNIG). Following inoculation with WNV, mice received i.p. injections of different doses (0.01-8 mg/mouse) of IVIG-IL or WNIG, according to the specific experimental protocol. RESULTS: WNIG was about 10 times more potent (per gr of IgG) than was regular IVIG-IL when tested by ELISA and neutralization assays. In a mouse lethal WNV infection model, prophylactic treatment with WNIG was at least 5-10-fold more potent as compared to treatment with IVIG-IL. Treatment with WNIG during active encephalitis, three or four days following WNV infection, had a significant protective effect. WNIG was also very effective in protecting immunosuppressed mice. Indeed, treatment of dexamethasone-immunosuppressed mice with 0.2 or 1.0 mg WNIG 4 h after virus infection, led to 100% survival. CONCLUSION: IVIG produced from selected plasma donated in WNV endemic regions can be used to produce WNV IVIG with superior activity for therapeutic and prophylactic measures.


Subject(s)
Antibodies, Viral/blood , Blood Donors , Immunization, Passive , Immunoglobulins, Intravenous/therapeutic use , West Nile Fever/drug therapy , Animals , Antibodies, Viral/therapeutic use , Chlorocebus aethiops , Female , Humans , Immunoglobulin G/therapeutic use , Israel/epidemiology , Mice , Mice, Inbred BALB C , Neutralization Tests , Vero Cells , West Nile Fever/epidemiology , West Nile virus/immunology
4.
J Virol Methods ; 141(2): 133-40, 2007 May.
Article in English | MEDLINE | ID: mdl-17215048

ABSTRACT

West Nile virus (WNV) is one of the major emerging infectious diseases in North America. WNV belongs to the genus Flavivirus, and its rapid and extensive global spread has highlighted the necessity for accurate and specific assays for diagnosis of WNV infection. This study presents the first phage displayed peptide based ELISA for detection of WNV immunoglobulin G (IgG). The Ep15 epitope, derived from the WNV E protein DIII, was cloned into a T7 phage display system that was then used as recombinant antigen in a chemiluminescent ELISA format. The phage concentration was optimized at 5 x 10(10)PFU/ml and was used directly after polyethylene glycol concentration. The assay shows a limit of detection at a serum titer of 1:51,200 and a dynamic range from 1:100 to 1:2000. A screen of a panel of 66 human sera samples, and comparison with a commercial kit, revealed a sensitivity of 67% and a specificity of 100%. Considering the ease of antigen preparation, its stability and the optimum display properties of the T7 bacteriophage, it is apparent that this approach can be useful for the preparation of highly sensitive and specific anti-WNV immunoglobulin diagnostic kits.


Subject(s)
Antibodies, Viral/blood , Bacteriophage T7 , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin G/blood , Peptide Library , West Nile virus/immunology , Bacteriophage T7/isolation & purification , Bacteriophage T7/metabolism , Epitopes/metabolism , Humans , Recombinant Proteins/metabolism , Sensitivity and Specificity , Viral Proteins/immunology , West Nile Fever/blood , West Nile Fever/diagnosis
5.
J Med Microbiol ; 48(5): 425-431, 1999 May.
Article in English | MEDLINE | ID: mdl-10229539

ABSTRACT

The protective effects of the hormones androstenediol (androstene-3beta, 17beta,-diol; AED) and dehydroepiandrosterone (5-androsten-3beta-ol-17-one; DHEA) on the pathophysiology of two lethal bacterial infections and endotoxin shock were examined. The infections included a gram-positive organism (Enterococcus faecalis) and a gram-negative organism (Pseudomonas aeruginosa). Both hormones protected mice from the lethal bacterial infections and from lipopolysaccharide (LPS) challenge. Treatment of animals lethally infected with P. aeruginosa with DHEA resulted in a 43% protection whereas treatment with AED gave a 67% protection. Both hormones also protected completely animals infected with an LD50 dose of E. faecalis. Similarly, the 88% mortality rate seen in LPS challenge was reduced to 17% and 8.5%, by treatment with DHEA and AED, respectively. The protective influences of both steroids were shown not to be directly antibacterial, but primarily an indirect antitoxin reaction. DHEA appears to mediate its protective effect by a mechanism that blocks the toxin-induced production of pathophysiological levels of tumour necrosis factor-alpha (TNF-alpha) and interleukin-1. AED usually had greater protective effects than DHEA; however, the AED effect was independent of TNF-alpha suppression, both in vivo and in vitro. The data suggest that both DHEA and AED may have a role in the neuro-endocrine regulation of antibacterial immune resistance.


Subject(s)
Androstenediol/therapeutic use , Dehydroepiandrosterone/therapeutic use , Gram-Positive Bacterial Infections/prevention & control , Lipopolysaccharides/toxicity , Pseudomonas Infections/prevention & control , Shock, Septic/prevention & control , Animals , Enterococcus faecalis , Female , Interleukin-1/biosynthesis , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Tumor Necrosis Factor-alpha/biosynthesis
7.
Vaccine ; 25(45): 7743-53, 2007 Nov 07.
Article in English | MEDLINE | ID: mdl-17928110

ABSTRACT

Since smallpox eradication by the WHO during the 1980s, potency of new vaccines is compared to vaccines that were used during the eradication campaign. In this work we characterize the tail scarification technique in mice as a model for scarification in humans. Similar to humans, mice develop "clinical take" which is dependent on the vaccination dose. Appearance of anti-Vaccinia IgM is followed by IgG antibodies 10 days post scarification and lasting more then 1(1/2) years. Mice with "clinical take" are 100% protected against lethal respiratory challenge (100LD(50)) of Vaccinia WR indicating that the "clinical take" can serve as a correlate of protective immunity. Reducing the vaccination dose and using Cowpox virus as a more virulent strain, enabled us to draw the limit of the vaccine potency in mice. Similar to humans, in revaccinated mice the development of "clinical take" was inversely correlated to the level of pre-existing antibodies. These results indicate that tail scarification of mice can be used as a model for evaluation of smallpox vaccines. High correlation between "clinical take" and protective immunity allows the use of visual inspection to evaluate vaccine potency.


Subject(s)
Antibodies, Viral/biosynthesis , Cicatrix/chemically induced , Smallpox Vaccine/adverse effects , Tail/drug effects , Vaccinia virus/immunology , Animals , Antibodies, Viral/analysis , Disease Models, Animal , Lethal Dose 50 , Mice , Smallpox Vaccine/immunology , T-Lymphocytes/immunology , Tail/pathology
8.
J Periodontal Res ; 38(2): 204-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12608916

ABSTRACT

Previous studies have shown that psychological stress plays a significant role in the outcome of infectious diseases, but data related to the effect of stress on periodontal infection is limited. The present study was designed to test the impact of emotional stress on the humoral immune response to the periodontal pathogen Porphyromonas gingivalis in a mouse model of local inflammation. Chambers constructed from titanium wire were implanted in the subcutaneous dorsolumbar region of mice. All mice were immunized with P. gingivalis followed by an intrachamber challenge with the bacteria. One group of mice was used as control, while the other two experienced experimental stress conditions (isolation/restraint stress). Stress-1 group was stressed during the immunization period, while Stress-2 group was stressed during the local challenge period. Chamber exudates and serum were collected and analyzed for levels of anti-P. gingivalis antibodies (IgG, IgG1 and IgG2a). The levels of serum antibodies to P. gingivalis were not different between the three tested groups, excluding increased levels of IgG2a in Stress-1 group at baseline. The levels of antibodies in the chamber exudates were significantly lower in the stressed groups at baseline, but higher at d 7. The IgG1 to IgG2a ratio was significantly higher in the control group compared with the two stressed groups. The findings of the present study suggest that chronic psychological stress had a marked impact on the localized response to P. gingivalis challenge. The lower IgG1/IgG2a ratio observed in the stress groups suggests elevated Th1 response during stress.


Subject(s)
Antibody Formation/immunology , Periodontitis/immunology , Porphyromonas gingivalis/immunology , Stress, Psychological/immunology , Analysis of Variance , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Chronic Disease , Diffusion Chambers, Culture , Disease Models, Animal , Exudates and Transudates/immunology , Female , Immunization , Immunoglobulin G/analysis , Immunoglobulin G/blood , Mice , Periodontitis/microbiology , Skin/immunology , Statistics as Topic , Th1 Cells/immunology , Time Factors
9.
J Infect Dis ; 188(1): 5-12, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12825165

ABSTRACT

West Nile virus (WNV) is a mosquito-borne disease found most commonly in Africa, West Asia, and the Middle East, where up to 40% of the human population possesses antibodies. It is an emerging disease in the United States. Humans infected with WNV develop a febrile illness that can progress to meningitis or encephalitis. In mice, WNV causes central nervous system infection, paralysis, encephalitis, and death. Currently, no specific therapy or vaccine has been approved for human use. We examined the prophylactic and therapeutic efficacy of pooled human plasma (PP) and intravenous immunoglobulin (IVIG) for treatment of WNV-infected mice. Full protection was achieved when the infected mice were treated with pooled plasma or IVIG obtained from healthy Israeli blood donors that contained WNV-specific antibodies. Similar treatments using PP or IVIG obtained from US blood donors had no protective effect. Recovery of the lethally infected mice was dependent on the dose and time of IVIG administration. These results indicate that antibodies play a major role in protection and recovery from WNV infection and that IVIG can be used as first-line therapy.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , West Nile Fever/drug therapy , West Nile Fever/prevention & control , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunization, Passive , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/immunology , Mice , Mice, Inbred BALB C , Time Factors , Virus Replication , West Nile Fever/immunology , West Nile virus/immunology , West Nile virus/physiology
10.
J Med Virol ; 66(4): 576-80, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11857540

ABSTRACT

There are contradictory reports regarding the effects of inhalation anesthetics on the immune system. Measurable immune responses have been studied in vitro, but little is known about the in vivo effects in the intact organism. We used an attenuated, non-neuroinvasive, nonlethal strain of the encephalitic West Nile virus, termed WN-25, which can become lethal in combination with environmental stressors, to study possible modulatory immune effects of inhalation anesthetics in mice. Both single short-term exposure and repeated exposure to halothane and nitrous oxide were studied. Exposure to 30% CO2 served as a positive control. Mortality, brain invasion, spleen weight, and antiviral antibodies served as the experimental endpoints. Halothane and nitrous oxide led to viral brain invasion, increased mortality, and suppressed immune response in a concentration- and time-dependent manner. Repeated exposures had a cumulative effect. Assessment of the stability of the viral attenuation did not demonstrate any alteration in the character of the virus, suggesting an increased access to the brain by inhalation anesthetics that led to the fatal encephalitis. These findings may be of special concern to populations at risk, such as operating room staff and patients undergoing general anesthesia in endemic areas of encephalitic virus species, in which subclinical infection may develop into an overt disease.


Subject(s)
Anesthetics, Inhalation/pharmacology , Brain/virology , Halothane/pharmacology , Nitrous Oxide/pharmacology , West Nile Fever/immunology , West Nile virus/pathogenicity , Anesthetics, Inhalation/administration & dosage , Animals , Antibodies, Viral/blood , Halothane/administration & dosage , Humans , Mice , Nitrous Oxide/administration & dosage , Virulence , West Nile Fever/mortality , West Nile Fever/virology , West Nile virus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL