Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Equine Vet J ; 51(6): 774-778, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30859618

ABSTRACT

BACKGROUND: Potomac horse fever (PHF) is a potentially fatal enterocolitis of horses caused by Neorickettsia risticii. The disease was originally recognised almost 40 years ago in the state of Maryland in the US. It is now known to occur in many areas of North America, as well as having been described in South America and Europe. Monocomponent PHF vaccines are available, but clinical protection with vaccination has been reported to be inconsistent. OBJECTIVES: This study was designed to assess the immunogenicity of a commercially available Potomac Horse Fever (PHF) vaccine when administered as either a monovalent PHF vaccine simultaneously co-administered with a separate monovalent Rabies vaccine or as a multivalent PHF/Rabies vaccine in horses. STUDY DESIGN: Randomised parallel group trial. METHODS: Ninety-one client or University owned horses participated in this open-label randomised study, with 45 horses receiving the monovalent vaccines at separate sites and 46 receiving the multivalent vaccine at a single site. Serum PHF IFA titres were determined twice prior to vaccination and at 1, 2 and 3 months after vaccination. RESULTS: Both vaccination protocols exhibited poor immunogenicity, with only one-third of all the animals demonstrating seroconversion, defined as an increase in titre of greater than 400 over baseline, at any time point after vaccination. The monovalent PHF vaccine exhibited significantly greater immunogenicity in terms of the number of horses exhibiting seroconversion, as compared to the multivalent vaccine, at one (20 vs. 11, P = 0.03) and two (18 vs. 9, p = 0.02) months post vaccination. The monovalent PHF vaccine also exhibited significantly greater immunogenicity in terms of the median (interquartile range) IFA titres, as compared to the multivalent vaccine, at one (800 [200-1600] vs. 400 [200-800], P = 0.009) and 2 months (400 [200-1600] vs. 400 [100-800], P = 0.02) post vaccination. There was no significant difference between groups at 3 months in either seroconversion rate or median IFA titers. MAIN LIMITATIONS: This study did not assess the actual protective effects of PHF vaccination but rather used the serologic response to vaccination as a surrogate biomarker of immunity. CONCLUSIONS: The multivalent PHF/Rabies vaccine exhibited lower immunogenicity as compared to the monovalent PHF vaccine co-administered with a separate Rabies vaccine.


Subject(s)
Anaplasmataceae Infections/veterinary , Horse Diseases/prevention & control , Neorickettsia risticii , Rabies Vaccines/immunology , Rabies/veterinary , Anaplasmataceae Infections/microbiology , Anaplasmataceae Infections/prevention & control , Animals , Antibodies, Bacterial/blood , Female , Horse Diseases/microbiology , Horses , Immunogenicity, Vaccine , Male , Rabies/prevention & control , Rickettsial Vaccines/immunology , Vaccination , Vaccines, Inactivated/immunology
2.
Infect Immun ; 87(2)2019 02.
Article in English | MEDLINE | ID: mdl-30396898

ABSTRACT

Rocky Mountain spotted fever (RMSF) is a potentially fatal tick-borne disease in people and dogs. RMSF is reported in the United States and several countries in North, Central, and South America. The causative agent of this disease, Rickettsia rickettsii, is transmitted by several species of ticks, including Dermacentor andersoni, Rhipicephalus sanguineus, and Amblyomma americanum RMSF clinical signs generally include fever, headache, nausea, vomiting, muscle pain, lack of appetite, and rash. If untreated, it can quickly progress into a life-threatening illness in people and dogs, with high fatality rates ranging from 30 to 80%. While RMSF has been known for over a century, recent epidemiological data suggest that the numbers of documented cases and the fatality rates remain high in people, particularly during the last two decades in parts of North America. Currently, there are no vaccines available to prevent RMSF in either dogs or people. In this study, we investigated the efficacies of two experimental vaccines, a subunit vaccine containing two recombinant outer membrane proteins as recombinant antigens (RCA) and a whole-cell inactivated antigen vaccine (WCA), in conferring protection against virulent R. rickettsii infection challenge in a newly established canine model for RMSF. Dogs vaccinated with WCA were protected from RMSF, whereas those receiving RCA developed disease similar to that of nonvaccinated R. rickettsii-infected dogs. WCA also reduced the pathogen loads to nearly undetected levels in the blood, lungs, liver, spleen, and brain and induced bacterial antigen-specific immune responses. This study provides the first evidence of the protective ability of WCA against RMSF in dogs.


Subject(s)
Antigens, Bacterial/immunology , Dog Diseases , Rickettsia rickettsii/immunology , Rickettsial Vaccines/immunology , Rocky Mountain Spotted Fever , Animals , Bacterial Outer Membrane Proteins/immunology , Dog Diseases/immunology , Dog Diseases/microbiology , Dog Diseases/prevention & control , Dogs , Recombinant Proteins/immunology , Rocky Mountain Spotted Fever/immunology , Rocky Mountain Spotted Fever/prevention & control , Rocky Mountain Spotted Fever/veterinary
3.
Adv Healthc Mater ; 7(11): e1701381, 2018 06.
Article in English | MEDLINE | ID: mdl-29663698

ABSTRACT

Dissolving microneedles (DMNs) have been widely studied in medical applications due to their pain-free administration, superior efficiency, and safe drug delivery. In skin vaccination, preserving the activity of the encapsulated antigen is an important consideration, as antigen activity is lost during DMN fabrication because of various stress factors. These stress factors vary between fabrication methods and each method affects the antigen's activity to different degrees. In this study, the activity of encapsulated antigens delivered by DMNs is compared between two recently developed DMN fabrication methods; droplet-born air blowing (DAB) and centrifugal lithography (CL) for a model scrub typhus vaccine antigen, ScaA. Although the in vitro analysis of ScaA-loaded DMNs (ScaA-DMNs) does not show any differences in physical properties depending on the fabrication methods, the immunogenicity of the CL-produced ScaA-DMN is significantly higher based on cytokine measurement and humoral immunity. DAB and CL differ in their solidification conditions, suggesting that solidification factors critically affect the encapsulated antigen's activity. ScaA-DMNs may also be stably stored for 4 weeks at room temperature. In conclusion, CL is a superior DMN fabrication method compared with DAB, and this study proves that DMN is feasible and practical for skin vaccination.


Subject(s)
Antigens, Bacterial/pharmacology , Needles , Rickettsial Vaccines/pharmacology , Skin/immunology , Vaccination/instrumentation , Vaccination/methods , Animals , Antigens, Bacterial/immunology , Injections, Intradermal , Mice , Rickettsial Vaccines/immunology , Swine
4.
Acta Trop ; 182: 100-110, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29474831

ABSTRACT

BACKGROUND: Antibiotic resistance is increasing rapidly in pathogenic organisms, creating more complications for treatment of diseases. Rocky Mountain spotted fever (RMSF) is a neglected tropical disease in humans caused by Rickettsia rickettsii for which no effective therapeutic is available. Subtractive genomics methods facilitate the characterization of non-homologous essential proteins that could be targeted for the discovery of potential therapeutic compounds against R. rickettsii to combat RMSF. Present study followed an in-silico based methodology, involving scanning and filtering the complete proteome of Rickettsia rickettsii by using several prioritization parameters in the search of potential candidates for drug development. Further the putative targets were subjected to series of molecular dockings with ligands obtained from PDB ligand database to identify suitable potential inhibitors. The comparative genomic analysis revealed 606 non-homologous proteins and 233 essential non-homologous proteins of R. rickettsii. The metabolic pathway analysis predicted 120 proteins as putative drug targets, out of which 56 proteins were found to be associated with metabolic pathways unique to the bacteria and further subcellular localization analysis revealed that 9 proteins as potential drug targets which are secretion proteins, involved in peptidoglycan biosynthesis, folate biosynthesis and bacterial secretion system. As secretion proteins are more feasible as vaccine candidates, we have selected a most potential target i.e. tolC, an outer membrane efflux protein that belongs to type I secretion system and has major role in pathogen survival as well as MDR persistence. So for case study, we have modelled the three dimensional structure of tolC (tunnel protein). The model was further subjected to virtual screening and in-silico docking. The study identified three potential inhibitors having PDB Id 19V, 6Q8 and 39H. Further we have suggested that the above study would be most important while considering the selection of candidate targets and drug or vaccine designing against R. rickettsii.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Drug Discovery/methods , Molecular Targeted Therapy/methods , Rickettsia rickettsii/genetics , Rickettsial Vaccines/genetics , Bacterial Outer Membrane Proteins/drug effects , Bacterial Outer Membrane Proteins/immunology , Comparative Genomic Hybridization , Genomics , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Proteome/analysis , Rickettsia rickettsii/chemistry , Rickettsia rickettsii/drug effects , Rickettsia rickettsii/immunology , Rickettsial Vaccines/immunology , Rocky Mountain Spotted Fever/drug therapy , Rocky Mountain Spotted Fever/microbiology
5.
PLoS One ; 11(2): e0148229, 2016.
Article in English | MEDLINE | ID: mdl-26841025

ABSTRACT

Ehrlichia chaffeensis is a tick-borne rickettsial pathogen and the causative agent of human monocytic ehrlichiosis. Transmitted by the Amblyomma americanum tick, E. chaffeensis also causes disease in several other vertebrate species including white-tailed deer and dogs. We have recently described the generation of an attenuated mutant strain of E. chaffeensis, with a mutation in the Ech_0660 gene, which is able to confer protection from secondary, intravenous-administered, wild-type E. chaffeensis infection in dogs. Here, we extend our previous results, demonstrating that vaccination with the Ech_0660 mutant protects dogs from physiologic, tick-transmitted, secondary challenge with wild-type E. chaffeensis; and describing, for the first time, the cellular and humoral immune responses induced by Ech_0660 mutant vaccination and wild-type E. chaffeensis infection in the canine host. Both vaccination and infection induced a rise in E. chaffeensis-specific antibody titers and a significant Th1 response in peripheral blood as measured by E. chaffeensis antigen-dependent CD4+ T cell proliferation and IFNγ production. Further, we describe for the first time significant IL-17 production by peripheral blood leukocytes from both Ech_0660 mutant vaccinated animals and control animals infected with wild-type E. chaffeensis, suggesting a previously unrecognized role for IL-17 and Th17 cells in the immune response to rickettsial pathogens. Our results are a critical first step towards defining the role of the immune system in vaccine-induced protection from E. chaffeensis infection in an incidental host; and confirm the potential of the attenuated mutant clone, Ech_0660, to be used as a vaccine candidate for protection against tick-transmitted E. chaffeensis infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ehrlichia chaffeensis/immunology , Ehrlichiosis/veterinary , Rickettsial Vaccines/immunology , Vaccination/veterinary , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cell Proliferation , Dogs , Ehrlichia chaffeensis/genetics , Ehrlichiosis/immunology , Ehrlichiosis/microbiology , Ehrlichiosis/prevention & control , Insect Vectors/microbiology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Ticks/microbiology
6.
Vaccine ; 33(8): 985-92, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25597943

ABSTRACT

BACKGROUND: Two surface proteins of Rickettsia rickettsii, outer membrane protein B (OmpB) and adhesion 2 (Adr2), have been recognized as protective antigens. Herein, the immunization with both OmpB and Adr2 was performed in mice so as to explore whether their combination could induce an enhanced immunoprotection against R. rickettsii infection. METHODS: C3H/HeN mice were immunized with recombinant protein rAdr2 or/and rOmp-4, a fragment derived from OmpB, and then mice were challenged with R. rickettsii. After which rickettsial loads in mice were measured by quantitative PCR. The specific antibodies in mouse sera were determined by ELISA and antigen-specific cytokines secretion by mouse T cells were analyzed in vitro. RESULTS: After challenge with R. rickettsii, the mice immunized with rAdr2 or/and rOmpB-4 had significant lower rickettsial load in livers, spleens, or lungs compared to PBS mock-immunized mice. Particularly, the load in lungs of mice immunized with both rAdr2 and rOmpB-4 was significantly lower than that with either of them. High levels of specific antibodies were detected in sera from mice immunized with rAdr2 or/and rOmpB-4, but the ratios of specific IgG2a to IgG1 induced by their combination were significantly higher than that by either rAdr2 or rOmpB-4. Following stimulation with rAdr2 or/and rOmpB-4, the INF-γ secreted by CD4(+) T cells from infected mice was significantly higher than that by cognate cells from uninfected mice. And the TNF-α secreted by CD4(+) or CD8(+) T cells from infected mice was markedly greater than that by cognate cells from uninfected mice after stimulation by their combination but not either of them. CONCLUSION: The combination of rAdr2 and rOmpB-4 conferred an enhanced protection against R. rickettsii infection in mice, which was mainly dependent on a stronger Th1-oriented immunoresponse with greater INF-γ and TNF-α secretion by antigen-specific T cells and specific IgG2a elicited by the combination.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Recombinant Fusion Proteins/immunology , Rickettsia rickettsii/immunology , Rocky Mountain Spotted Fever/prevention & control , Adhesins, Bacterial/genetics , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Cytokines/biosynthesis , Disease Models, Animal , Immunization , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C3H , Rickettsia rickettsii/genetics , Rickettsial Vaccines/genetics , Rickettsial Vaccines/immunology , Rocky Mountain Spotted Fever/immunology , Rocky Mountain Spotted Fever/metabolism , Rocky Mountain Spotted Fever/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Vaccine ; 32(39): 4968-76, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25010827

ABSTRACT

Rickettsial agents are some of the most lethal pathogens known to man. Among them, Rickettsia prowazekii is a select agent with potential use for bioterrorism; yet, there is no anti-Rickettsia vaccine commercially available. Owing to the obligate intracellular lifestyle of rickettsiae, CD8(+) T cells are indispensable for protective cellular immunity. Furthermore, T cells can mediate cross-protective immunity between different pathogenic Rickettsia, a finding consistent with the remarkable similarity among rickettsial genomes. However, Rickettsia T cell antigens remain unidentified. In the present study, we report an algorithm that allowed us to identify and validate four novel R. prowazekii vaccine antigen candidates recognized by CD8(+) T cells from a set of twelve in silico-defined protein targets. Our results highlight the importance of combining proteasome-processing as well as MHC class-I-binding predictions. The novel rickettsial vaccine candidate antigens, RP778, RP739, RP598, and RP403, protected mice against a lethal challenge with Rickettsia typhi, which is indicative of cross-protective immunity within the typhus group rickettsiae. Together, our findings validate a reverse vaccinology approach as a viable strategy to identify protective rickettsial antigens and highlight the feasibility of a subunit vaccine that triggers T-cell-mediated cross-protection among diverse rickettsiae.


Subject(s)
Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Protection , Rickettsia prowazekii/immunology , Algorithms , Animals , Histocompatibility Antigens Class I/immunology , Mice , Rickettsial Vaccines/immunology
8.
PLoS One ; 8(10): e76253, 2013.
Article in English | MEDLINE | ID: mdl-24146844

ABSTRACT

Rickettsia prowazekii has been tested for biological warfare due to the high mortality that it produces after aerosol transmission of very low numbers of rickettsiae. Epidemic typhus, the infection caused by these obligately intracellular bacteria, continues to be a threat because it is difficult to diagnose due to initial non-specific symptoms and the lack of commercial diagnostic tests that are sensitive and specific during the initial clinical presentation. A vaccine to prevent epidemic typhus would constitute an effective deterrent to the weaponization of R. prowazekii; however, an effective and safe vaccine is not currently available. Due to the cytoplasmic niche of Rickettsia, CD8(+) T-cells are critical effectors of immunity; however, the identification of antigens recognized by these cells has not been systematically addressed. To help close this gap, we designed an antigen discovery strategy that uses cell-based vaccination with antigen presenting cells expressing microbe's proteins targeted to the MHC class I presentation pathway. We report the use of this method to discover a protective T-cell rickettsial antigen, RP884, among a test subset of rickettsial proteins.


Subject(s)
Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , Rickettsia prowazekii/immunology , Typhus, Epidemic Louse-Borne/immunology , Typhus, Epidemic Louse-Borne/prevention & control , Animals , Antigen-Presenting Cells/immunology , Bacterial Proteins/metabolism , CD8-Positive T-Lymphocytes/microbiology , Computational Biology , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Genetic Vectors/metabolism , Mice , Reproducibility of Results , Rickettsia prowazekii/genetics , Rickettsial Vaccines/immunology , Typhus, Epidemic Louse-Borne/microbiology
9.
Vaccine ; 31(35): 3617-22, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23664994

ABSTRACT

Despite significant economic losses resulting from infection with Anaplasma marginale, a tick-transmitted rickettsial pathogen of cattle, available vaccines provide, at best, only partial protection against clinical disease. The green-fluorescent protein expressing mutant of the A. marginale St. Maries strain is a live, marked vaccine candidate (AmStM-GFP). To test whether AmStM-GFP is safe and provides clinical protection, a group of calves was vaccinated, and clinical parameters, including percent parasitized erythrocytes (PPE), packed cell volume (PCV) and days required to reach peak bacteremia, were measured following inoculation and following tick challenge with wild type St. Maries strain (AmStM). These clinical parameters were compared to those obtained during infection with the A. marginale subsp. centrale vaccine strain (A. centrale) or wild type AmStM. AmStM-GFP resulted in similar clinical parameters to A. centrale, but had a lower maximum PPE, smaller drop in PCV and took longer to reach peak bacteremia than wild type AmStM. AmStM-GFP provided clinical protection, yielding a stable PCV and low bacteremia following challenge, whereas A. centrale only afforded partial clinical protection.


Subject(s)
Anaplasma marginale/immunology , Anaplasmosis/immunology , Cattle Diseases/immunology , Rickettsial Vaccines/immunology , Vaccination/veterinary , Anaplasma marginale/genetics , Anaplasmosis/microbiology , Anaplasmosis/prevention & control , Animals , Antibodies, Bacterial/blood , Bacteremia/microbiology , Cattle , Cattle Diseases/prevention & control , Cross Protection/immunology , Erythrocytes/microbiology , Green Fluorescent Proteins/genetics , Male , Rickettsial Vaccines/adverse effects
10.
FEMS Immunol Med Microbiol ; 64(1): 115-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22066488

ABSTRACT

The capacity to identify immunogens for vaccine development by genome-wide screening has been markedly enhanced by the availability of microbial genome sequences coupled to proteomic and bioinformatic analysis. Critical to this approach is in vivo testing in the context of a natural host­pathogen relationship, one that includes genetic diversity in the host as well as among pathogen strains. We aggregate the results of three independent genome-wide screens using in vivo immunization and protection against Anaplasma marginale as a model for discovery of vaccine antigens for rickettsial pathogens. In silico analysis identified 62 outer membrane proteins (Omp) from the 949 predicted proteins in the A. marginale genome. These 62 Omps were reduced to 10 vaccine candidates by two independent genome-wide screens using IgG2 from vaccinates protected from challenge following vaccination with outer membranes (screen 1) or bacterial surface complexes (screen 2). Omps with broadly conserved epitopes were identified by immunization with a live heterologous vaccine, A. marginale ssp. centrale (screen 3), reducing the candidates to three. The genome-wide screens identified Omps that have orthologs broadly conserved among rickettsial pathogens, highlighted the importance of identifying immunologically subdominant antigens, and supported the use of reverse vaccinology approaches in vaccine development for rickettsial diseases.


Subject(s)
Anaplasma marginale/immunology , Antigens, Bacterial/immunology , Genome, Bacterial , Rickettsial Vaccines/immunology , Anaplasma marginale/genetics , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Computational Biology/methods , Humans , Mass Screening
11.
Infect Immun ; 79(6): 2303-13, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21444665

ABSTRACT

Pathogenic rickettsiae are the causative agents of Rocky Mountain spotted fever, typhus, and other human diseases with high mortality and an important impact on society. Although survivors of rickettsial infections are considered immune to disease, the molecular basis of this immunity or the identification of protective antigens that enable vaccine development was hitherto not known. By exploring the molecular pathogenesis of Rickettsia conorii, the agent of Mediterranean spotted fever, we report here that the autotransporter protein, rickettsial outer membrane protein B (rOmpB), constitutes a protective antigen for this group of pathogens. A recombinant, purified rOmpB passenger domain fragment comprised of amino acids 36 to 1334 is sufficient to elicit humoral immune responses that protect animals against lethal disease. Protective immunity requires folded antigen and production of antibodies that recognize conformational epitopes on the rickettsial surface. Monoclonal antibodies (MAbs) 5C7.27 and 5C7.31, which specifically recognize a conformation present in the folded, intact rOmpB passenger domain, are sufficient to confer immunity in vivo. Analyses in vitro indicate this protection involves a mechanism of complement-mediated killing in mammalian blood, a means of rickettsial clearance that has not been previously described. Considering the evolutionary conservation of rOmpB and its crucial contribution to bacterial invasion of host cells, we propose that rOmpB antibody-mediated killing confers immunity to rickettsial infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Boutonneuse Fever/immunology , Rickettsia conorii/immunology , Animals , Antibodies, Monoclonal/immunology , Boutonneuse Fever/microbiology , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Flow Cytometry , HeLa Cells , Humans , Male , Mice , Mice, Inbred C3H , Polymerase Chain Reaction , Rickettsial Vaccines/immunology , Vero Cells
12.
Vaccine ; 29(4): 712-20, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21115057

ABSTRACT

An efficacious, low cost vaccine against typhoid fever, especially for young children, would make a major impact on disease burden in developing countries. The virulence capsular polysaccharide of Salmonella Typhi (Vi) coupled to recombinant mutant Pseudomonas aeruginosa exoprotein A (Vi-rEPA) has been shown to be highly efficacious. We investigated the use of carrier proteins included in infant vaccines, standardized the conjugation process and developed key assays required for routine lot release at production scale. Vi from a BSL1 organism, Citrobacter freundii, strain WR7011, was used as an alternative to Vi from S. Typhi. We showed that Vi conjugated to CRM(197), a non-toxic mutant of diphtheria toxin, widely used in commercial vaccines, was produced at high yield. Vi-CRM(197) proved immunogenic in animal studies, even without adjuvant. Thus, Vi-CRM(197) appears to be a suitable candidate for the development of a commercially viable, effective typhoid vaccine for developing countries.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Proteins/administration & dosage , Polysaccharides, Bacterial/immunology , Rickettsial Vaccines/immunology , Typhoid Fever/prevention & control , Animals , Antibodies, Bacterial/blood , Citrobacter freundii/chemistry , Citrobacter freundii/immunology , Female , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/isolation & purification , Rickettsial Vaccines/administration & dosage , Salmonella typhi/chemistry , Salmonella typhi/immunology , Typhoid Fever/immunology , Vaccination/methods , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
13.
Vaccine ; 27 Suppl 4: D52-5, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19837287

ABSTRACT

Rickettsia prowazekii, R. rickettsii, R. conorii, and R. typhi are serious biologic weapon threats because of high infectivity of low dose aerosols, stable small particle aerosol infectivity, virulence causing severe disease, difficulty in establishing a timely diagnosis, ineffectiveness of usual empiric treatments, potential for engineered complete antimicrobial resistance, lower level of immunity, availability of the agents in nature, and feasibility of propagation, stabilization, and dispersal. Infection induces long-term immunity, killed rickettsial vaccines stimulate incomplete protection, and a live attenuated mutant stimulates strong immunity but reverts to virulence. Prospects for rational development of a safe, effective live attenuated vaccine are excellent.


Subject(s)
Rickettsia Infections/prevention & control , Rickettsia/immunology , Rickettsial Vaccines/immunology , Animals , Biological Warfare/prevention & control , Biological Warfare Agents , Humans , Rickettsia/pathogenicity , Vaccines, Attenuated , Virulence
14.
Clin Vaccine Immunol ; 15(9): 1461-71, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18650399

ABSTRACT

Typhoid caused by Salmonella enterica serovar Typhi remains a major health concern worldwide. The emergence of multidrug-resistant strains of Salmonella with increased virulence, communicability, and survivability leading to increased morbidity and mortality has further complicated its management. Currently available vaccines for typhoid have less-than-desired efficacy and certain unacceptable side effects, making it pertinent to search for new immunogens suitable for vaccine formulation. The outer membrane proteins (OMPs) of Salmonella have been considered possible candidates for conferring protection against typhoid. OMPs interface the cell with the environment, thus representing important virulence factors with a significant role in the pathobiology of gram-negative bacteria and bacterial adaptation. An OMP of Salmonella enterica serovar Typhimurium with an apparent molecular mass of 49 kDa that is highly immunogenic, evokes humoral and cell-mediated immune responses, and confers 100% protection to immunized rats against challenge with very high doses (up to 100 times the 50% lethal dose) of Salmonella enterica serovar Typhimurium has been identified. Further, very efficient clearance of bacteria from the reticuloendothelial systems of immunized animals was seen. This protein is recognized by the antibodies present in serum of typhoid patients. When sodium dodecyl sulfate-polyacrylamide gel electrophoresis gel-eluted protein was further analyzed by high-performance liquid chromatography (HPLC) and two-dimensional electrophoresis, two polypeptides with the same molecular weight were resolved. These have different isoelectric points and gave two peaks with different retention times in reverse-phase HPLC. However, only one of the two bands interacted with patient serum. The immunogenicity studies (enzyme-linked immunosorbent assay and delayed-type hypersensitivity [DTH]) indicated that the immunoreactive protein evoked a strong immune response in rats. The N-terminal sequencing and analysis of the homology of this protein with sequences in the protein database of Salmonella resulted in a match with the N-terminal sequences of a protein in Salmonella enterica serovar Typhi (CT18 and Ty2 strains). The homology search further revealed it to be a hypothetical protein, whose gene had unidentified open reading frames in Salmonella serovar Typhi encoding 447 amino acid residues, corresponding to a molecular mass of 49 kDa. The nucleotide sequence of the encoding gene was deduced, and the gene was amplified by PCR using appropriate primers. An amplified 1.3-kb band was purified and sequenced to confirm its identity. These OMPs provide promising targets for the development of a candidate vaccine against typhoid.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Rickettsial Vaccines/immunology , Salmonella typhimurium/immunology , Typhoid Fever/prevention & control , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/isolation & purification , Chromatography, High Pressure Liquid , Colony Count, Microbial , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Genes, Bacterial , Humans , Hypersensitivity, Delayed , Isoelectric Point , Lethal Dose 50 , Liver/microbiology , Mice , Molecular Weight , Mononuclear Phagocyte System/microbiology , Rats , Sequence Analysis, DNA , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Vaccines, Subunit/immunology
15.
Biochim Biophys Acta ; 1774(3): 373-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17301007

ABSTRACT

Rickettsia prowazekii, an obligate intracellular Gram-negative bacterium, is the etiologic agent of epidemic typhus. We analyzed the proteome of the virulent Breinl strain of R. prowazekii purified from infected egg yolk sacs. Total proteins from purified R. prowazekii Breinl strain were reduced by dithiothreitol, alkylated by iodoacetic acid and digested with trypsin followed by analysis with an integrated two-dimensional liquid chromatography and mass spectrometry system (2D-LC/MS/MS). A comparison was made using previously analyzed proteome of the Madrid E strain and current analysis of the Breinl strain. For Breinl 251 proteins were identified, representing 30% of the total protein-encoding genes, using a shotgun 2D-LC/MS/MS proteomic approach. This result is identical to that of Madrid E strain. Among the identified proteins, 33 from Breinl and 37 from Madrid E have an unknown function. A methyltransferase, RP028/RP027, whose gene is mutated in the avirulent Madrid E strain but not in the virulent Breinl strain, was only detectable in the Breinl strain, consistent with the genetic mutation in Madrid E. This result suggests the possible relationship between this gene product and the virulence of the strains.


Subject(s)
Proteomics , Rickettsia prowazekii/metabolism , Rickettsia prowazekii/pathogenicity , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA, Bacterial/genetics , Molecular Sequence Data , Protein Array Analysis , Rickettsia prowazekii/chemistry , Rickettsia prowazekii/classification , Rickettsial Vaccines/genetics , Rickettsial Vaccines/immunology , Tandem Mass Spectrometry , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Virulence/physiology
16.
Ann N Y Acad Sci ; 1063: 130-42, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16481504

ABSTRACT

The gene fragments encoding outer membrane protein 1 (P1) and heat-shock protein B (HspB) amplified from genomic DNA of Coxiella burnetii Xinqiao by PCR were inserted into prokaryotic expression vector pQE30 to construct recombinant expression plasmids pQE30/p1 and pQE30/hspB, respectively. The p1 fragment from pQE30/p1 was ligated with hspB of pQE30/hspB to construct pQE30/p1-hspB. Recombinant proteins, P1, HspB, and P1-HspB, were expressed in Escherichia coli cells transformed with pQE30/p1, pQE30/hspB, and pQE30/p1-hspB, respectively. The purified recombinant proteins and whole-cell antigen (WCA) of C. burnetii were used to immunize BALB/c mice. The antibody detection, T-cell proliferation assay, and cytokine detection demonstrated that the animals immunized with P1-HspB or WCA exhibited stronger humoral and cellular immune responses compared with animals immunized with P1 or HspB individually. Seven days after challenge of 10-fold 50% infection dose of C. burnetii, mice were euthanized and their spleens were collected. The splenic weights of mice immunized with P1-HspB or WCA were significantly lighter than that of mice immunized with P1 or HspB. By real-time PCR assay, the coxiella loads of spleens of mice immunized with P1-HspB or WCA were also significantly lower than that of mice immunized with P1 or HspB. The data from this study indicate that fusion antigen P1-HspB is a good immunogen for eliciting immunoresponses against C. burnetii, and it may be a more suitable candidate for preparing subunit vaccine against Q fever.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Heat-Shock Proteins/immunology , Q Fever/prevention & control , Recombinant Fusion Proteins/immunology , Rickettsial Vaccines/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Chick Embryo , Heat-Shock Proteins/administration & dosage , Heat-Shock Proteins/genetics , Immunity, Cellular , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Organ Size , Q Fever/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Rickettsial Vaccines/genetics , Spleen/microbiology , Spleen/physiology , Ticks/microbiology
17.
Ann N Y Acad Sci ; 1063: 143-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16481505

ABSTRACT

Previous attempts to develop Q fever vaccines were less successful in that the vaccines caused unacceptable side effects or failed to be protective. In this study, we tested the efficacy of a mixture of eight recombinant Coxiella burnetii (C. b.) proteins in sublethal challenge infections with mice. Eight potential C. b. virulence genes (Omp, Pmm, HspB, Fbp, Orf410, Crc, CbMip, and MucZ) were overexpressed in E. coli as his-tagged fusion proteins and partially purified. All recombinant proteins but rPmm proved to be antigenic in BALB/c mice when administered as protein mixtures. For efficacy testing, mice were immunized with an adjuvanted mixture of the eight recombinant proteins and subsequently challenged intraperitoneally with the C. b. isolate Nine Mile RSA493 (1.8 x 10(8) C. b.). Only animals vaccinated with the licensed Q fever vaccine Q-Vax (vaccination control) exhibited milder symptoms and minor gain of spleen and liver weights. In summary, clinical examinations and dissection of mice immunized with the eight recombinant C. b. proteins did not indicate a protective immune response after test infection.


Subject(s)
Antigens, Bacterial/immunology , Q Fever/prevention & control , Rickettsial Vaccines/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Female , Liver/physiology , Mice , Mice, Inbred BALB C , Organ Size , Q Fever/immunology , Q Fever/microbiology , Q Fever/physiopathology , Rickettsial Vaccines/administration & dosage , Rickettsial Vaccines/genetics , Spleen/physiology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
18.
Ann N Y Acad Sci ; 1063: 171-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16481510

ABSTRACT

The Balb/c mice were infected with Coxiella burnetii and samples of blood and major organs from the infected mice were collected on days 1 to 14 after infection. The DNAs extracted from the samples were detected by a developed real-time quantitative PCR specific for C. burnetii and high loads of C. burnetii were found in spleens, livers, and lungs, particularly in spleens. The Balb/c mice were immunized with whole cell antigen (WCA) of C. burnetii and coxiella loads in spleens of mice were assessed by the real-time quantitative PCR on day 7 after challenge with C. burnetii. The analysis suggested that phase I whole cells were excellent immunogen that elicited complete protection against coxiella infection by two-booster but not one-booster immunization. The results suggest that the combination of Balb/c model and the real-time quantitative PCR assay is a reliable and sensitive way to evaluate the efficiency of vaccines against Q fever.


Subject(s)
Coxiella burnetii/immunology , Disease Models, Animal , Q Fever/immunology , Q Fever/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Rickettsial Vaccines/immunology , Animals , Dose-Response Relationship, Immunologic , Immunization, Secondary , Male , Mice , Mice, Inbred BALB C , Rickettsial Vaccines/administration & dosage , Splenic Diseases/immunology , Splenic Diseases/microbiology , Ticks/microbiology
20.
Ann N Y Acad Sci ; 1063: 266-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16481526

ABSTRACT

Orientia tsutsugamushi is an obligate intracellular bacterium that is the causative agent of scrub typhus. To develop an effective vaccine to prevent or ameliorate scrub typhus, knowledge of the protective immune response to O. tsutsugamushi needs to be ascertained. Our laboratory has demonstrated that the DNA vaccine vector pVR1012 carrying the O. tsutsugamushi Karp strain 47-kDa protein gene (p47Kp) consistently provides outbred mice protection against homologous challenge.


Subject(s)
Orientia tsutsugamushi/immunology , Rickettsial Vaccines/immunology , Scrub Typhus/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Bacterial/blood , Female , Interferon-gamma/biosynthesis , Mice , Rickettsial Vaccines/administration & dosage , Scrub Typhus/prevention & control , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Time Factors , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...