Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Antimicrob Agents Chemother ; 60(8): 4878-85, 2016 08.
Article in English | MEDLINE | ID: mdl-27270276

ABSTRACT

Protective antigen (PA)-based vaccines are effective in preventing the development of fatal anthrax disease both in humans and in relevant animal models. The Bacillus anthracis toxins lethal toxin (lethal factor [LF] plus PA) and edema toxin (edema factor [EF] plus PA) are essential for the establishment of the infection, as inactivation of these toxins results in attenuation of the pathogen. Since the toxins reach high toxemia levels at the bacteremic stages of the disease, the CDC's recommendations include combining antibiotic treatment with antitoxin (anti-PA) immunotherapy. We demonstrate here that while treatment with a highly potent neutralizing monoclonal antibody was highly efficient as postexposure prophylaxis treatment, it failed to protect rabbits with any detectable bacteremia (≥10 CFU/ml). In addition, we show that while PA vaccination was effective against a subcutaneous spore challenge, it failed to protect rabbits against systemic challenges (intravenous injection of vegetative bacteria) with the wild-type Vollum strain or a toxin-deficient mutant. To test the possibility that additional proteins, which are secreted by the bacteria under pathogenicity-stimulating conditions in vitro, may contribute to the vaccine's potency, we immunized rabbits with a secreted protein fraction from a toxin-null mutant. The antiserum raised against the secreted fraction reacts with the bacteria in an immunofluorescence assay. Immunization with the secreted protein fraction did not protect the rabbits against a systemic challenge with the fully pathogenic bacteria. Full protection was obtained only by a combined vaccination with PA and the secreted protein fraction. Therefore, these results indicate that an effective antiserum treatment in advanced stages of anthrax must include toxin-neutralizing antibodies in combination with antibodies against bacterial cell targets.


Subject(s)
Anthrax/immunology , Antigens, Bacterial/immunology , Antitoxins/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Animals , Anthrax/microbiology , Anthrax Vaccines/immunology , Antibodies, Bacterial/immunology , Female , Immune Sera/immunology , Rabbits , Spores, Bacterial/immunology , Vaccination/methods
2.
Antimicrob Agents Chemother ; 59(12): 7497-503, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26392505

ABSTRACT

Respiratory anthrax is a fatal disease in the absence of early treatment with antibiotics. Rabbits are highly susceptible to infection with Bacillus anthracis spores by intranasal instillation, succumbing within 2 to 4 days postinfection. This study aims to test the efficiency of antibiotic therapy to treat systemic anthrax in this relevant animal model. Delaying the initiation of antibiotic administration to more than 24 h postinfection resulted in animals with systemic anthrax in various degrees of bacteremia and toxemia. As the onset of symptoms in humans was reported to start on days 1 to 7 postexposure, delaying the initiation of treatment by 24 to 48 h (time frame for mass distribution of antibiotics) may result in sick populations. We evaluated the efficacy of antibiotic administration as a function of bacteremia levels at the time of treatment initiation. Here we compare the efficacy of treatment with clarithromycin, amoxicillin-clavulanic acid (Augmentin), imipenem, vancomycin, rifampin, and linezolid to the previously reported efficacy of doxycycline and ciprofloxacin. We demonstrate that treatment with amoxicillin-clavulanic acid, imipenem, vancomycin, and linezolid were as effective as doxycycline and ciprofloxacin, curing rabbits exhibiting bacteremia levels of up to 10(5) CFU/ml. Clarithromycin and rifampin were shown to be effective only as a postexposure prophylactic treatment but failed to treat the systemic (bacteremic) phase of anthrax. Furthermore, we evaluate the contribution of combined treatment of clindamycin and ciprofloxacin, which demonstrated improvement in efficacy compared to ciprofloxacin alone.


Subject(s)
Amoxicillin-Potassium Clavulanate Combination/pharmacology , Anthrax/drug therapy , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Bacteremia/drug therapy , Ciprofloxacin/pharmacology , Doxycycline/pharmacology , Respiratory Tract Infections/drug therapy , Animals , Anthrax/microbiology , Anthrax/mortality , Anthrax/pathology , Bacillus anthracis/pathogenicity , Bacillus anthracis/physiology , Bacteremia/microbiology , Bacteremia/mortality , Bacteremia/pathology , Clarithromycin/pharmacology , Disease Models, Animal , Drug Combinations , Drug Synergism , Humans , Imipenem/pharmacology , Linezolid/pharmacology , Male , Microbial Sensitivity Tests , Rabbits , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Respiratory Tract Infections/pathology , Rifampin/pharmacology , Spores, Bacterial/drug effects , Spores, Bacterial/pathogenicity , Spores, Bacterial/physiology , Survival Analysis , Vancomycin/pharmacology
3.
Mol Syst Biol ; 10: 720, 2014.
Article in English | MEDLINE | ID: mdl-24586061

ABSTRACT

Hundreds of immune cell types work in coordination to maintain tissue homeostasis. Upon infection, dramatic changes occur with the localization, migration, and proliferation of the immune cells to first alert the body of the danger, confine it to limit spreading, and finally extinguish the threat and bring the tissue back to homeostasis. Since current technologies can follow the dynamics of only a limited number of cell types, we have yet to grasp the full complexity of global in vivo cell dynamics in normal developmental processes and disease. Here, we devise a computational method, digital cell quantification (DCQ), which combines genome-wide gene expression data with an immune cell compendium to infer in vivo changes in the quantities of 213 immune cell subpopulations. DCQ was applied to study global immune cell dynamics in mice lungs at ten time points during 7 days of flu infection. We find dramatic changes in quantities of 70 immune cell types, including various innate, adaptive, and progenitor immune cells. We focus on the previously unreported dynamics of four immune dendritic cell subtypes and suggest a specific role for CD103(+) CD11b(-) DCs in early stages of disease and CD8(+) pDC in late stages of flu infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Influenza, Human/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CD11b Antigen/immunology , Flow Cytometry , Humans , Influenza, Human/metabolism , Influenza, Human/pathology , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Lung/immunology , Mice , Transcriptome/immunology
4.
Immunol Rev ; 239(1): 221-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21198675

ABSTRACT

The lethal anthrax disease is caused by spores of the gram-positive Bacillus anthracis, a member of the cereus group of bacilli. Although the disease is very rare in the Western world, development of anthrax countermeasures gains increasing attention due to the potential use of B. anthracis spores as a bio-terror weapon. Protective antigen (PA), the non-toxic subunit of the bacterial secreted exotoxin, fulfills the role of recognizing a specific receptor and mediating the entry of the toxin into the host target cells. PA elicits a protective immune response and represents the basis for all current anthrax vaccines. Anti-PA neutralizing antibodies are useful correlates for protection and for vaccine efficacy evaluation. Post exposure anti-toxemic and anti-bacteremic prophylactic treatment of anthrax requires prolonged antibiotic administration. Shorter efficient postexposure treatments may require active or passive immunization, in addition to antibiotics. Although anthrax is acknowledged as a toxinogenic disease, additional factors, other than the bacterial toxin, may be involved in the virulence of B. anthracis and may be needed for the long-lasting protection conferred by PA immunization. The search for such novel factors is the focus of several high throughput genomic and proteomic studies that are already leading to identification of novel targets for therapeutics, for vaccine candidates, as well as biomarkers for detection and diagnosis.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Animals , Anthrax/therapy , Antibodies, Bacterial/immunology , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Biological Warfare , Disease Models, Animal , Guinea Pigs , Humans , Mice , Rabbits , Rats , Spores, Bacterial/immunology , Spores, Bacterial/pathogenicity , Vaccines, DNA
5.
Infect Immun ; 80(8): 2623-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22585968

ABSTRACT

The virulence of Bacillus anthracis, the causative agent of anthrax, stems from its antiphagocytic capsule, encoded by pXO2, and the tripartite toxins encoded by pXO1. The accepted paradigm states that anthrax is both an invasive and toxinogenic disease and that the toxins play major roles in pathogenicity. We tested this assumption by a systematic study of mutants with combined deletions of the pag, lef, and cya genes, encoding protective antigen (PA), lethal factor (LF), and edema factor (EF), respectively. The resulting seven mutants (single, double, and triple) were evaluated following subcutaneous (s.c.) and intranasal (i.n.) inoculation in rabbits and guinea pigs. In the rabbit model, virulence is completely dependent on the presence of PA. Any mutant bearing a pag deletion behaved like a pXO1-cured mutant, exhibiting complete loss of virulence with attenuation indices of over 2,500,000 or 1,250 in the s.c. or i.n. route of infection, respectively. In marked contrast, in guinea pigs, deletion of pag or even of all three toxin components resulted in relatively moderate attenuation, whereas the pXO1-cured bacteria showed complete attenuation. The results indicate that a pXO1-encoded factor(s), other than the toxins, has a major contribution to the virulence mechanism of B. anthracis in the guinea pig model. These unexpected toxin-dependent and toxin-independent manifestations of pathogenicity in different animal models emphasize the importance and need for a comprehensive evaluation of B. anthracis virulence in general and in particular for the design of relevant next-generation anthrax vaccines.


Subject(s)
Anthrax/microbiology , Antigens, Bacterial/toxicity , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacillus anthracis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , DNA, Bacterial/genetics , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Genotype , Guinea Pigs , Polymerase Chain Reaction , Rabbits , Virulence
6.
Microb Pathog ; 52(1): 55-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22020310

ABSTRACT

Bacillus anthracis secretes three major components, which assemble into two bipartite toxins: lethal toxin (LT), composed of lethal factor (LF) and protective antigen (PA) and edema toxin (ET), composed of edema factor (EF) and PA. EF is a potent calmodulin-dependent adenylate cyclase, which is internalized into the target cell following PA binding. Once inside the cell, EF elevates cAMP levels, interrupting intracellular signaling. Effects of ET were demonstrated on monocytes, neutrophils and T-cells. In an earlier work we demonstrated that a deletion of LF in a fully virulent strain had no effect in guinea pigs and a significant, but not major, effect in the rabbit model. These results suggested that EF might play an important role in the development of infection and mortality following exposure to B. anthracis spores. To evaluate the role of EF in B. anthracis pathogenicity we deleted the cya gene, which encodes the EF protein, in the fully virulent Vollum strain. The Δcya mutant was fully virulent in the guinea pig model as determined by LD(50) experiments. In the rabbit model, when infected subcutaneously, the absence of EF had no effect on the virulence of the mutant. However an increase of two orders of magnitude in the LD(50) was demonstrated when the rabbits were infected by intranasal instillation accompanied with partial mortality and increased mean time to death. These results argue that in the guinea pig model the presence of one of the toxins, ET or LT is sufficient for the development of the infection. In the rabbit model ET plays a role in respiratory infection, most probably mediating the early steps of host colonization.


Subject(s)
Anthrax/microbiology , Antigens, Bacterial/genetics , Bacillus anthracis/pathogenicity , Bacterial Toxins/genetics , Gene Deletion , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Anthrax/mortality , Antigens, Bacterial/metabolism , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Disease Models, Animal , Female , Guinea Pigs , Humans , Rabbits , Virulence
7.
Antimicrob Agents Chemother ; 55(4): 1533-42, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21263056

ABSTRACT

Respiratory anthrax, in the absence of early antibiotic treatment, is a fatal disease. This study aimed to test the efficiency of antibiotic therapy in curing infected animals and those sick with anthrax. Postexposure prophylaxis (24 h postinfection [p.i.]) of guinea pigs infected intranasally with Bacillus anthracis Vollum spores with doxycycline, ofloxacin, imipenem, and gentamicin conferred protection. However, upon termination of treatment, the animals died from respiratory anthrax. Combined treatment with antibiotics and active vaccination with a protective antigen-based vaccine leads to full protection even after cessation of treatment. Delaying the initiation of antibiotic administration to over 24 h p.i. resulted in treatment of animals with anthrax exhibiting various degrees of bacteremia and toxemia. Treatment with doxycycline or ciprofloxacin cured sick guinea pigs and rabbits exhibiting bacteremia levels up to 10(5) CFU/ml. Addition of anti-protective antigen (PA) antibodies augmented the efficiency of protection, allowing the cure of guinea pigs and rabbits with 10- to 20-fold-higher bacteremia levels, up to 7 × 10(5) CFU/ml and 2 × 10(6) CFU/ml, respectively. Treatment with ciprofloxacin and a monoclonal anti-PA antibody rescued rabbits with bacteremia levels up to 4 × 10(6) CFU/ml. During antibiotic administration, all surviving animals developed a protective immune response against development of a fatal disease and subcutaneous challenge with Vollum spores. In conclusion, these results demonstrate that antibiotic treatment can prevent the development of fatal disease in respiratory-anthrax-infected animals and can cure animals after disease establishment. A therapeutic time window of 40 h to 48 h from infection to initiation of efficient antibiotic-mediated cure was observed.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/drug effects , Bacillus anthracis/pathogenicity , Animals , Anthrax/immunology , Anthrax/microbiology , Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacteremia/drug therapy , Bacteremia/immunology , Bacteremia/microbiology , Doxycycline/therapeutic use , Enzyme-Linked Immunosorbent Assay , Gentamicins/therapeutic use , Guinea Pigs , Imipenem/therapeutic use , Ofloxacin/therapeutic use , Rabbits
8.
Microb Pathog ; 51(5): 345-51, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21791242

ABSTRACT

The major virulence factor of Bacillus anthracis is the tripartite anthrax toxin, comprising the protective antigen (PA), lethal factor (LF) and edema factor (EF). The LF of B. anthracis is a metalloprotease that has been shown to play an important role in pathogenicity. Deletion of this gene (lef) in the Sterne strain was reported to dramatically reduce the pathogenicity of this strain in mice, and was reported to be as dramatic as the deletion of PA. We evaluated the effect on pathogenicity of the lef deletion in the fully virulent Vollum strain in guinea pigs and NZW rabbits by either subcutaneous injection or intranasal instillation. In guinea pigs, no major differences between the mutant strain and the wild type could be detected in the LD(50) or mean time to death values. On the other hand, the lef deletion caused death of 50-70% of all rabbits infected with the mutant spores at doses equivalent or higher than the wild type LD(50). The surviving rabbits, which were infected with spore doses higher than the wild type LD(50), developed a protective immune response that conferred resistance to challenge with the wild type strain. These findings may indicate that the mutant lacking the LF is capable of host colonization which causes death in 50-70% of the animals and a protective immune response in the others. These results indicate that unlike the data obtained in mice, the LF mutation does not abolish B. anthracis pathogenicity.


Subject(s)
Anthrax/microbiology , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/metabolism , Animals , Antigens, Bacterial/genetics , Bacillus anthracis/genetics , Bacterial Toxins/genetics , Disease Models, Animal , Female , Guinea Pigs , Humans , Mice , Rabbits , Species Specificity , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Cell Host Microbe ; 20(4): 458-470, 2016 Oct 12.
Article in English | MEDLINE | ID: mdl-27736644

ABSTRACT

Mounting an effective immune response, while also protecting tissue integrity, is critical for host survival. We used a combined genomic and proteomic approach to investigate the role of extracellular matrix (ECM) proteolysis in achieving this balance in the lung during influenza virus infection. We identified the membrane-tethered matrix metalloprotease MT1-MMP as a prominent host-ECM-remodeling collagenase in influenza infection. Selective inhibition of MT1-MMP protected the tissue from infection-related structural and compositional tissue damage. MT1-MMP inhibition did not significantly alter the immune response or cytokine expression. The available flu therapeutic Oseltamivir did not prevent lung ECM damage and was less effective than anti-MT1-MMP in influenza virus Streptococcus pneumoniae coinfection paradigms. Combination therapy of Oseltamivir with anti-MT1-MMP showed a strong synergistic effect and resulted in complete recovery of infected mice. This study highlights the importance of tissue resilience in surviving infection and the potential of such host-pathogen therapy combinations for respiratory infections.


Subject(s)
Extracellular Matrix/metabolism , Lung/pathology , Matrix Metalloproteinase 14/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae/growth & development , Animals , Antiviral Agents/therapeutic use , Disease Models, Animal , Female , Gene Expression Profiling , Genomics , Lung/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Oseltamivir/therapeutic use , Protease Inhibitors/therapeutic use , Proteolysis , Proteome/analysis , Proteomics , Survival Analysis , Treatment Outcome
10.
FEMS Microbiol Lett ; 244(1): 199-205, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15727841

ABSTRACT

Classification and differentiation of Bacillus anthracis isolates by genetic markers play an important role in anthrax research. We used a PCR based method--Random Amplification of Polymorphic DNA (RAPD)--to identify genetic markers in B. anthracis strains. Twenty-five differential genetic markers were identified which divided the strains into five different groups. Three selected RAPD-markers were cloned and sequenced. The five RAPD-derived genotypes could be defined by integration of these three markers. This system offers a simple non-expensive method to classify B. anthracis strains in laboratories involved in the research of this bacterium.


Subject(s)
Bacillus anthracis/genetics , DNA, Bacterial/genetics , Bacillus/classification , Bacillus/genetics , Bacillus/isolation & purification , Bacillus anthracis/classification , Bacillus anthracis/isolation & purification , Base Sequence , Cloning, Molecular , Databases, Nucleic Acid , Genetic Markers , Genotype , Phylogeny , Polymorphism, Genetic , Random Amplified Polymorphic DNA Technique , Species Specificity
11.
PLoS One ; 9(1): e84947, 2014.
Article in English | MEDLINE | ID: mdl-24416317

ABSTRACT

The accepted paradigm states that anthrax is both an invasive and toxinogenic disease and that the toxins play a major role in pathogenicity. In the guinea pig (GP) model we have previously shown that deletion of all three toxin components results in a relatively moderate attenuation in virulence, indicating that B. anthracis possesses an additional toxin-independent virulence mechanism. To characterize this toxin-independent mechanism in anthrax disease, we developed a new rabbit model by intravenous injection (IV) of B. anthracis encapsulated vegetative cells, artificially creating bacteremia. Using this model we were able to demonstrate that also in rabbits, B. anthracis mutants lacking the toxins are capable of killing the host within 24 hours. This virulent trait depends on the activity of AtxA in the presence of pXO2, as, in the absence of the toxin genes, deletion of either component abolishes virulence. Furthermore, this IV virulence depends mainly on AtxA rather than the whole pXO1. A similar pattern was shown in the GP model using subcutaneous (SC) administration of spores of the mutant strains, demonstrating the generality of the phenomenon. The virulent strains showed higher bacteremia levels and more efficient tissue dissemination; however our interpretation is that tissue dissemination per se is not the main determinant of virulence whose exact nature requires further elucidation.


Subject(s)
Bacillus anthracis/pathogenicity , Animals , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacterial Capsules/metabolism , Female , Mutation , Rabbits , Spores, Bacterial/genetics , Spores, Bacterial/metabolism , Spores, Bacterial/pathogenicity , Toxins, Biological/genetics , Toxins, Biological/metabolism , Virulence
12.
PLoS One ; 9(11): e112319, 2014.
Article in English | MEDLINE | ID: mdl-25375158

ABSTRACT

Infection of the central nervous system is considered a complication of Anthrax and was reported in humans and non-human primates. Previously we have reported that Bacillus anthracis possesses a toxin-independent virulent trait that, like the toxins, is regulated by the major virulence regulator, AtxA, in the presence of pXO2. This toxin-independent lethal trait is exhibited in rabbits and Guinea pigs following significant bacteremia and organ dissemination. Various findings, including meningitis seen in humans and primates, suggested that the CNS is a possible target for this AtxA-mediated activity. In order to penetrate into the brain tissue, the bacteria have to overcome the barriers isolating the CNS from the blood stream. Taking a systematic genetic approach, we compared intracranial (IC) inoculation and IV/SC inoculation for the outcome of the infection in rabbits/GP, respectively. The outstanding difference between the two models is exhibited by the encapsulated strain VollumΔpXO1, which is lethal when injected IC, but asymptomatic when inoculated IV/SC. The findings demonstrate that there is an apparent bottleneck in the ability of mutants to penetrate into the brain. Any mutant carrying either pXO1 or pXO2 will kill the host upon IC injection, but only those carrying AtxA either on pXO1 or in the chromosome in the background of pXO2 can penetrate into the brain following peripheral inoculation. The findings were corroborated by histological examination by H&E staining and immunofluorescence of rabbits' brains following IV and IC inoculations. These findings may have major implications on future research both on B. anthracis pathogenicity and on vaccine development.


Subject(s)
Anthrax , Antigens, Bacterial , Bacillus anthracis , Bacterial Proteins , Bacterial Toxins , Brain/metabolism , Trans-Activators , Animals , Anthrax/genetics , Anthrax/metabolism , Anthrax/pathology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Brain/pathology , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/metabolism , Guinea Pigs , Humans , Rabbits , Trans-Activators/genetics , Trans-Activators/metabolism
13.
Innate Immun ; 15(1): 43-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19201824

ABSTRACT

The involvement of TLR2 receptor in the innate response to infection with Bacillus anthracis was investigated. We studied the response to virulent or attenuated Vollum strains in either in vitro assays using macrophage cultures, or in an in vivo model comparing the sensitivity of Syrian hamster cells (expressing normal TLR2) to Chinese hamster cells (lacking functional TLR2) to infection by the various B. anthracis strains. Phagocytosis experiments with murine cell cultures or primary macrophages from both hamster strains, using virulent or attenuated Tox(+)Cap(-), Tox(-)Cap(+) or Tox(-)Cap(-) spores indicated that the secretion of TNF-alpha was induced by all the bacterial spores and purified spore antigens. In contrast, capsular antigens induce secretion of TNF-alpha only by Syrian hamster macrophages indicating the involvement of a functional TLR2 in macrophage activation. Challenge experiments with both hamster strains by intranasal spore inoculation, indicated that, while both strains are equally sensitive to infection with the virulent strain, the Chinese hamster demonstrated a higher sensitivity to infection with the toxinogenic or encapsulated strains. In conclusion, our findings imply that TLR2 has an important role in the attempt of the innate immunity to control B. anthracis infection, although TNF-alpha secretion was found to be mediated by both TLR2-dependent and TLR2-independent pathways.


Subject(s)
Anthrax/immunology , Bacillus anthracis/immunology , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/immunology , Animals , Anthrax/microbiology , Cell Line , Cricetinae , Cricetulus , Cytokines/biosynthesis , Cytokines/immunology , Immunity, Innate , Macrophages/immunology , Macrophages/microbiology , Mesocricetus , Mice , Phagocytosis/immunology , Spores, Bacterial/immunology , Tumor Necrosis Factor-alpha/biosynthesis
14.
Infect Immun ; 74(1): 394-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16368995

ABSTRACT

Correlates between immunological parameters and protection against Bacillus anthracis infection in animals vaccinated with protective antigen (PA)-based vaccines could provide surrogate markers to evaluate the putative protective efficiency of immunization in humans. In previous studies we demonstrated that neutralizing antibody levels serve as correlates for protection in guinea pigs (S. Reuveny et al., Infect. Immun. 69:2888-2893, 2001; H. Marcus et al., Infect. Immun. 72:3471-3477, 2004). In this study we evaluated similar correlates for protection by active and passive immunization of New Zealand White rabbits. Full immunization and partial immunization were achieved by single and multiple injections of standard and diluted doses of a PA-based vaccine. Passive immunization was carried out by injection of immune sera from rabbits vaccinated with PA-based vaccine prior to challenge with B. anthracis spores. Immunized rabbits were challenged by intranasal spore instillation with one of two virulent strains (strains Vollum and ATCC 6605). The immune competence was estimated by measuring the level of total anti-PA antibodies, the neutralizing antibody titers, and the conferred protective immunity. The results indicate that total anti-PA antibody titers greater than 1 x 10(5) conferred protection, whereas lower titers (between 10(4) and 10(5)) provided partial protection but failed to predict protection. Neutralizing antibody titers between 500 and 800 provided partial protection, while titers higher than 1,000 conferred protection. In conclusion, this study emphasizes that regardless of the immunization regimen or the time of challenge, neutralizing antibody titers are better predictors of protection than total anti-PA titers.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Bacillus anthracis/immunology , Administration, Intranasal , Animals , Anthrax/mortality , Anthrax/prevention & control , Antigens, Bacterial/immunology , Rabbits , Spores, Bacterial/immunology
15.
Infect Immun ; 74(10): 5871-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16988266

ABSTRACT

The most aggressive form of anthrax results from inhalation of airborne spores of Bacillus anthracis and usually progresses unnoticed in the early stages because of unspecific symptoms. The only reliable marker of anthrax is development of bacteremia, which increases with disease progress. Rapid diagnosis of anthrax is imperative for efficient treatment and cure. Herein we demonstrate that the presence and level of a bacterial antigen, the protective antigen (PA), a component of B. anthracis toxins, in host sera can serve as a reliable marker of infection. This was tested in two animal models of inhalation anthrax, rabbits and guinea pigs infected by intranasal instillation of Vollum spores. In both models, we demonstrated qualitative and quantitative correlations between levels of bacteremia and PA concentrations in the sera of sick animals. The average time to death in infected animals was about 16 h after the appearance of bacteremia, leaving a small therapeutic window. As the time required for immunodetection of PA can be very short, the use of this marker will be beneficial for faster diagnosis and treatment of inhalation anthrax.


Subject(s)
Anthrax/diagnosis , Bacteremia/diagnosis , Pneumonia, Bacterial/diagnosis , Animals , Antigens, Bacterial/blood , Bacterial Toxins/blood , Biomarkers/blood , Disease Models, Animal , Guinea Pigs , Rabbits
16.
Infect Immun ; 74(7): 3987-4001, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16790772

ABSTRACT

Bacillus anthracis proteins that possess antigenic properties and are able to evoke an immune response were identified by a reductive genomic-serologic screen of a set of in silico-preselected open reading frames (ORFs). The screen included in vitro expression of the selected ORFs by coupled transcription and translation of linear PCR-generated DNA fragments, followed by immunoprecipitation with antisera from B. anthracis-infected animals. Of the 197 selected ORFs, 161 were chromosomal and 36 were on plasmids pXO1 and pXO2, and 138 of the 197 ORFs had putative functional annotations (known ORFs) and 59 had no assigned functions (unknown ORFs). A total of 129 of the known ORFs (93%) could be expressed, whereas only 38 (64%) of the unknown ORFs were successfully expressed. All 167 expressed polypeptides were subjected to immunoprecipitation with the anti-B. anthracis antisera, which revealed 52 seroreactive immunogens, only 1 of which was encoded by an unknown ORF. The high percentage of seroreactive ORFs among the functionally annotated ORFs (37%; 51/129) attests to the predictive value of the bioinformatic strategy used for vaccine candidate selection. Furthermore, the experimental findings suggest that surface-anchored proteins and adhesins or transporters, such as cell wall hydrolases, proteins involved in iron acquisition, and amino acid and oligopeptide transporters, have great potential to be immunogenic. Most of the seroreactive ORFs that were tested as DNA vaccines indeed appeared to induce a humoral response in mice. We list more than 30 novel B. anthracis immunoreactive virulence-related proteins which could be useful in diagnosis, pathogenesis studies, and future anthrax vaccine development.


Subject(s)
Anthrax Vaccines/genetics , Anthrax Vaccines/immunology , Anthrax/microbiology , Bacillus anthracis/immunology , Genome, Bacterial/immunology , Genomics , Open Reading Frames/immunology , Vaccines, DNA/immunology , Animals , Anthrax/immunology , Anthrax/prevention & control , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/physiology , Bacillus anthracis/genetics , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/immunology , Computational Biology/methods , Guinea Pigs , Immune Sera/blood , Immune Sera/genetics , Mice , Mice, Inbred ICR , Open Reading Frames/genetics , Open Reading Frames/physiology , Rabbits , Vaccines, DNA/genetics
17.
Mol Microbiol ; 58(2): 533-51, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16194238

ABSTRACT

Here we describe the characterization of a lipoprotein previously proposed as a potential Bacillus anthracis virulence determinant and vaccine candidate. This protein, designated MntA, is the solute-binding component of a manganese ion ATP-binding cassette transporter. Coupled proteomic-serological screen of a fully virulent wild-type B. anthracis Vollum strain, confirmed that MntA is expressed both in vitro and during infection. Expression of MntA is shown to be independent of the virulence plasmids pXO1 and pXO2. An mntA deletion, generated by allelic replacement, results in complete loss of MntA expression and its phenotypic analysis revealed: (i) impaired growth in rich media, alleviated by manganese supplementation; (ii) increased sensitivity to oxidative stress; and (iii) delayed release from cultured macrophages. The DeltamntA mutant expresses the anthrax-associated classical virulence factors, lethal toxin and capsule, in vitro as well as in vivo, and yet the mutation resulted in severe attenuation; a 10(4)-fold drop in LD(50) in a guinea pig model. MntA expressed in trans allowed to restore, almost completely, the virulence of the DeltamntA B. anthracis strain. We propose that MntA is a novel B. anthracis virulence determinant essential for the development of anthrax disease, and that B. anthracisDeltamntA strains have the potential to serve as platform for future live attenuated vaccines.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Proteins/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Animals , Bacillus anthracis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cells, Cultured , Female , Guinea Pigs , Humans , Iron/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Macrophages/cytology , Macrophages/metabolism , Manganese/metabolism , Molecular Sequence Data , Plasmids/genetics , Plasmids/metabolism , Proteome/analysis , Sequence Alignment
18.
Infect Immun ; 71(3): 1352-60, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12595452

ABSTRACT

The genes that encode the enterotoxigenic Escherichia coli (ETEC) CS4 fimbriae, csaA, -B, -C, -E, and -D', were isolated from strain E11881A. The csa operon encodes a 17-kDa major fimbrial subunit (CsaB), a 40-kDa tip-associated protein (CsaE), a 27-kDa chaperone-like protein (CsaA), a 97-kDa usher-like protein (CsaC), and a deleted regulatory protein (CsaD'). The predicted amino acid sequences of the CS4 proteins are highly homologous to structural and assembly proteins of other ETEC fimbriae, including CS1 and CS2, and to CFA/I in particular. The csaA, -B, -C, -E operon was cloned on a stabilized plasmid downstream from an osomotically regulated ompC promoter. pGA2-CS4 directs production of CS4 fimbriae in both E. coli DH5alpha and Shigella flexneri 2a vaccine strain CVD 1204, as detected by Western blot analysis and bacterial agglutination with anti-CS4 immune sera. Electron-microscopic examination of Shigella expressing CS4 confirmed the presence of fimbriae on the bacterial surface. Guinea pigs immunized with CVD 1204(pGA2-CS4) showed serum and mucosal antibody responses to both the Shigella vector and the ETEC fimbria CS4. Among the seven most prevalent fimbrial antigens of human ETEC, CS4 is the last to be cloned and sequenced. These findings pave the way for CS4 to be included in multivalent ETEC vaccines, including an attenuated Shigella live-vector-based ETEC vaccine.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Vaccines/immunology , Fimbriae Proteins/genetics , Shigella/genetics , Vaccines, Synthetic/immunology , Animals , Cloning, Molecular , Escherichia coli Proteins/immunology , Fimbriae Proteins/immunology , Genetic Vectors , Guinea Pigs , Immunization , Operon
19.
Vaccine ; 21(5-6): 333-40, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12531629

ABSTRACT

Shigella and enterotoxigenic Escherichia coli (ETEC) continue to be important causes of diarrheal disease in infants and young children in developing countries and are major etiologic agents of traveler's diarrhea. Since attenuated strains of Shigella have been developed as live oral vaccines against shigellosis, we have adapted these attenuated Shigella strains to serve as carriers of ETEC antigens, thereby constituting a hybrid vaccine. Since protective immunity against ETEC is largely directed against fimbrial antigens (of which there are multiple antigenic types), we have individually expressed four different ETEC fimbriae, including CFA/I, CS2, CS3, and CS4, using deltaguaBA attenuated Shigella vaccine strain CVD 1204 as a prototype live vector. Following mucosal (intranasal) immunization of guinea pigs, serum IgG and mucosal IgA responses were elicited against each fimbrial type. An additional strain was constructed expressing a detoxified version of the human ETEC variant of heat labile toxin (LThK63). Following mucosal immunization of guinea pigs with a mixed inoculum containing five Shigella strains each expressing a different ETEC antigen, immune responses were observed against each ETEC antigen plus the Shigella vector.


Subject(s)
Bacterial Toxins/biosynthesis , Bacterial Toxins/immunology , Enterotoxins/biosynthesis , Enterotoxins/immunology , Escherichia coli Proteins , Escherichia coli Vaccines/immunology , Escherichia coli/immunology , Mutation/immunology , Shigella Vaccines/immunology , Shigella/immunology , Animals , Blotting, Western , Cloning, Molecular , Dysentery, Bacillary/immunology , Dysentery, Bacillary/prevention & control , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Guinea Pigs , Hemagglutination Inhibition Tests , Immunization , Molecular Biology , Plasmids/immunology , Shigella/genetics , Shigella/metabolism , Species Specificity
20.
Infect Immun ; 70(2): 544-60, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11796581

ABSTRACT

The efficacy of passive immunization as a postexposure prophylactic measure for treatment of guinea pigs intranasally infected with Bacillus anthracis spores was evaluated. Antisera directed either against the lethal toxin components (PA or LF) or against a toxinogenic strain (Sterne) were used for this evaluation. All antisera exhibited high enzyme-linked immunosorbent assay titers against the corresponding antigens, high titers of neutralization of cytotoxicity activity in an in vitro mouse macrophages cell line (J774A.1), as well as in vivo neutralization of toxicity when administered either directly to Fisher rats prior to challenge with the lethal toxin or after incubation with the lethal toxin. In these tests, anti-LF antiserum exhibited the highest neutralization efficiency, followed by anti-Sterne and anti-PA. The time dependence and antibody dose necessary for conferring postexposure protection by the various antibodies of guinea pigs infected with 25 50% lethal doses of Vollum spores was examined. Rabbit anti-PA serum was found to be the most effective. Intraperitoneal injections of anti-PA serum given 24 h postinfection protected 90% of the infected animals, whereas anti-Sterne and anti-LF were less effective. These results further emphasizes the importance of anti-PA antibodies in conferring protection against B. anthracis infection and demonstrated the ability of such antibodies to be effectively applied as an efficient postexposure treatment against anthrax disease.


Subject(s)
Anthrax Vaccines/therapeutic use , Anthrax/prevention & control , Antibodies, Bacterial/therapeutic use , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Immunization, Passive , Animals , Anthrax Vaccines/immunology , Bacillus anthracis/immunology , Cell Line , Female , Guinea Pigs , Immunization, Passive/methods , Macrophages/cytology , Male , Mice , Rabbits , Rats , Rats, Inbred F344 , Spores, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL