Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pathogens ; 10(6)2021 May 22.
Article in English | MEDLINE | ID: mdl-34067337

ABSTRACT

Pneumonic tularemia is a highly debilitating and potentially fatal disease caused by inhalation of Francisella tularensis. Most of our current understanding of its pathogenesis is based on the highly virulent F. tularensis subsp. tularensis strain SCHU S4. However, multiple sources of SCHU S4 have been maintained and propagated independently over the years, potentially generating genetic variants with altered virulence. In this study, the virulence of four SCHU S4 stocks (NR-10492, NR-28534, NR-643 from BEI Resources and FTS-635 from Battelle Memorial Institute) along with another virulent subsp. tularensis strain, MA00-2987, were assessed in parallel. In the Fischer 344 rat model of pneumonic tularemia, NR-643 and FTS-635 were found to be highly attenuated compared to NR-10492, NR-28534, and MA00-2987. In the NZW rabbit model of pneumonic tularemia, NR-643 caused morbidity but not mortality even at a dose equivalent to 500x the LD50 for NR-10492. Genetic analyses revealed that NR-10492 and NR-28534 were identical to each other, and nearly identical to the reference SCHU S4 sequence. NR-643 and FTS-635 were identical to each other but were found to have nine regions of difference in the genomic sequence when compared to the published reference SCHU S4 sequence. Given the genetic differences and decreased virulence, NR-643/FTS-635 should be clearly designated as a separate SCHU S4 substrain and no longer utilized in efficacy studies to evaluate potential vaccines and therapeutics against tularemia.

2.
Methods Mol Biol ; 1700: 293-318, 2018.
Article in English | MEDLINE | ID: mdl-29177837

ABSTRACT

The resistance nodulation cell division (RND) family of proteins are inner membrane transporters that associate with periplasmic adaptor proteins and outer membrane porins to affect substrate transport from the cytosol and periplasm in Gram-negative bacteria. Various structurally diverse compounds are substrates of RND transporters. Along with their notable role in antibiotic resistance, these transporters are essential for niche colonization, quorum sensing, and virulence as well as for the removal of fatty acids and bile salts. As such, RNDs are an attractive target for antimicrobial development. However, while enhancing the utility of antibiotics with an RND inhibitor is an appealing concept, only a small core of chemotypes has been identified as efflux pump inhibitors (EPIs). Thus, our key objective is the development and validation of an efflux profiling and discovery strategy for RND model systems. Here we describe a flow cytometric dye accumulation assay that uses fluorescein diacetate (FDA) to interrogate the model Gram-negative pathogens Escherichia coli, Franscisella tularensis, and Burkholderia pseudomallei. Fluorochrome retention is increased in the presence of known efflux inhibitors and in RND deletion strains. The assay can be used in a high-throughput format to evaluate efflux of dye-substrate candidates and to screen chemical libraries for novel EPIs. Triaged compounds that inhibit efflux in pathogenic strains are tested for growth inhibition and antibiotic potentiation using microdilution culture plates in a select agent Biosafety Level-3 (BSL3) environment. This combined approach demonstrates the utility of flow cytometric analysis for efflux activity and provides a useful platform in which to characterize efflux in pathogenic Gram-negative bacteria. Screening small molecule libraries for novel EPI candidates offers the potential for the discovery of new classes of antibacterial compounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fluoresceins/metabolism , Gram-Negative Bacteria/growth & development , Membrane Transport Proteins/isolation & purification , Small Molecule Libraries/pharmacology , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Burkholderia pseudomallei/growth & development , Burkholderia pseudomallei/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Multiple, Bacterial , Escherichia coli/growth & development , Escherichia coli/metabolism , Flow Cytometry , Francisella tularensis/growth & development , Francisella tularensis/metabolism , Gram-Negative Bacteria/metabolism , Membrane Transport Proteins/metabolism , Substrate Specificity
3.
Am J Pathol ; 187(2): 252-267, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27939130

ABSTRACT

The inbred Fischer 344 rat is being evaluated for testing novel vaccines and therapeutics against pneumonic tularemia. Although primary pneumonic tularemia in humans typically occurs by inhalation of aerosolized bacteria, the rat model has relied on intratracheal inoculation of organisms because of safety and equipment issues. We now report the natural history of pneumonic tularemia in female Fischer 344 rats after nose-only inhalational exposure to lethal doses of aerosolized Francisella tularensis subspecies tularensis, strain SCHU S4. Our results are consistent with initial uptake of aerosolized SCHU S4 from the nasal cavity, lungs, and possibly the gastrointestinal tract. Bacteremia with hematogenous dissemination was first detected 2 days after exposure. Shortly thereafter, the infected rats exhibited fever, tachypnea, and hypertension that persisted for 24 to 36 hours and then rapidly decreased as animals succumbed to infection between days 5 and 8 after exposure. Tachycardia was observed briefly, but only after the core body temperature and blood pressure began to decrease as the animals were near death. Initial neutrophilic and histiocytic inflammation in affected tissues became progressively more fibrinous and necrotizing over time. At death, as many as 1010 colony-forming units were found in the lungs, spleen, and liver. Death was attributed to sepsis and disseminated intravascular coagulation. Overall, the pathogenesis of pneumonic tularemia in the female F344 rat model appears to replicate the disease in humans.


Subject(s)
Disease Models, Animal , Lung Diseases/microbiology , Lung Diseases/pathology , Tularemia/pathology , Animals , Female , Francisella tularensis , Rats , Rats, Inbred F344
4.
Lab Chip ; 16(21): 4142-4151, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27713988

ABSTRACT

Since the introduction of micro total analytical systems (µTASs), significant advances have been made toward development of lab-on-a-chip platforms capable of performing complex biological assays that can revolutionize public health, among other applications. However, use of these platforms in low-resource environments (e.g. developing countries) has yet to be realized as the majority of technologies used to control microfluidic flow rely on off-device hardware with non-negligible size, cost, power requirements and skill/training to operate. In this paper we describe a magnetic-adhesive based valve that is simple to construct and operate, and can be used to control fluid flow and store reagents within a microfluidic device. The design consists of a port connecting two chambers on different planes in the device that is closed by a neodymium disk magnet seated on a thin ring of adhesive. Bringing an external magnet into contact with the outer surface of the device unseats and displaces the valve magnet from the adhesive ring, exposing the port. Using this configuration, we demonstrate on-device reagent storage and on-demand transport and reaction of contents between chambers. This design requires no power or external instrumentation to operate, is extremely low cost ($0.20 materials cost per valve), can be used by individuals with no technical training, and requires only a hand-held magnet to actuate. Additionally, valve actuation does not compromise the integrity of the completely sealed microfluidic device, increasing safety for the operator when toxic or harmful substances are contained within. This valve concept has the potential to simplify design of µTASs, facilitating development of lab-on-a-chip systems that may be practical for use in point-of-care and low-resource settings.


Subject(s)
Adhesives , Health Resources/supply & distribution , Lab-On-A-Chip Devices , Magnets , Point-of-Care Systems
5.
Clin Vaccine Immunol ; 22(8): 902-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041039

ABSTRACT

The immunogenicity of Bacillus anthracis capsule (poly-γ-D-glutamic acid [PGA]) conjugated to recombinant B. anthracis protective antigen (rPA) or to tetanus toxoid (TT) was evaluated in two anthrax-naive juvenile chimpanzees. In a previous study of these conjugates, highly protective monoclonal antibodies (MAbs) against PGA were generated. This study examines the polyclonal antibody response of the same animals. Preimmune antibodies to PGA with titers of >10(3) were detected in the chimpanzees. The maximal titer of anti-PGA was induced within 1 to 2 weeks following the 1st immunization, with no booster effects following the 2nd and 3rd immunizations. Thus, the anti-PGA response in the chimpanzees resembled a secondary immune response. Screening of sera from nine unimmunized chimpanzees and six humans revealed antibodies to PGA in all samples, with an average titer of 10(3). An anti-PA response was also observed following immunization with PGA-rPA conjugate, similar to that seen following immunization with rPA alone. However, in contrast to anti-PGA, preimmune anti-PA antibody titers and those following the 1st immunization were ≤300, with the antibodies peaking above 10(4) following the 2nd immunization. The polyclonal anti-PGA shared the MAb 11D epitope and, similar to the MAbs, exerted opsonophagocytic killing of B. anthracis. Most important, the PGA-TT-induced antibodies protected mice from a lethal challenge with virulent B. anthracis spores. Our data support the use of PGA conjugates, especially PGA-rPA targeting both toxin and capsule, as expanded-spectrum anthrax vaccines.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Polyglutamic Acid/analogs & derivatives , Animals , Anthrax/immunology , Anthrax Vaccines/administration & dosage , Bacillus anthracis/physiology , Bacterial Toxins/immunology , Blood Bactericidal Activity , Disease Models, Animal , Female , Humans , Immunization, Passive , Mice, Inbred BALB C , Microbial Viability/drug effects , Opsonin Proteins/blood , Pan troglodytes , Polyglutamic Acid/immunology , Survival Analysis , Tetanus Toxoid/immunology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
6.
Langmuir ; 31(15): 4481-9, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25822668

ABSTRACT

A wide range of oligo-p-phenylene ethynylenes has been shown to exhibit good biocidal activity against both Gram-negative and Gram-positive bacteria. While cell death may occur in the dark, these biocidal compounds are far more effective in the light as a result of their ability to sensitize the production of cell-damaging reactive oxygen species. In these studies, the interactions of a specific cationic oligo-p-phenylene ethynylene with spore-forming Bacillus atrophaeus and Bacillus anthracis Sterne have been investigated. Flow cytometry assays are used to rapidly monitor cell death as well as spore germination. This compound effectively killed Bacillus anthracis Sterne vegetative cells (over 4 log reduction), presumably by severe perturbations of the bacterial cell wall and cytoplasmic membrane, while also acting as an effective spore germinant in the dark. While 2 log reduction of B. anthracis Sterne spores was observed, it is hypothesized that further killing could be achieved through enhanced germination.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Bacillus/drug effects , Photosensitizing Agents/pharmacology , Polymers/pharmacology , Spores, Bacterial/drug effects , Bacillus/growth & development , Bacillus/ultrastructure , Bacillus anthracis/growth & development , Bacillus anthracis/ultrastructure , Cell Wall/drug effects , Cell Wall/ultrastructure , Light , Microscopy, Electron, Scanning , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Spores, Bacterial/growth & development , Spores, Bacterial/ultrastructure
7.
Am J Pathol ; 184(12): 3205-16, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25285720

ABSTRACT

Inhalational anthrax is caused by inhalation of Bacillus anthracis spores. The ability of B. anthracis to cause anthrax is attributed to the plasmid-encoded A/B-type toxins, edema toxin (edema factor and protective antigen) and lethal toxin (lethal factor and protective antigen), and a poly-d-glutamic acid capsule. To better understand the contribution of these toxins to the disease pathophysiology in vivo, we used B. anthracis Ames strain and isogenic toxin deletion mutants derived from the Ames strain to examine the role of lethal toxin and edema toxin after pulmonary spore challenge of cynomolgus macaques. Lethal toxin, but not edema toxin, was required to induce sustained bacteremia and death after pulmonary challenge with spores delivered via bronchoscopy. After intravenous challenge with bacilli to model the systemic phase of infection, lethal toxin contributed to bacterial proliferation and subsequent host death to a greater extent than edema toxin. Deletion of protective antigen resulted in greater loss of virulence after intravenous challenge with bacilli than deletion of lethal toxin or edema toxin alone. These findings are consistent with the ability of anti-protective antigen antibodies to prevent anthrax and suggest that lethal factor is the dominant toxin that contributes to the escape of significant numbers of bacilli from the thoracic cavity to cause anthrax after inhalation challenge with spores.


Subject(s)
Anthrax/microbiology , Antigens, Bacterial/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/metabolism , Lung/microbiology , Respiratory Tract Infections/microbiology , Animals , Antibodies, Bacterial/blood , Female , Macaca , Male , Spores, Bacterial/pathogenicity , Virulence , Virulence Factors/metabolism
8.
PLoS Pathog ; 9(4): e1003306, 2013.
Article in English | MEDLINE | ID: mdl-23637599

ABSTRACT

Bacterial capsules are common targets for antibody-mediated immunity. The capsule of Bacillus anthracis is unusual among capsules because it is composed of a polymer of poly-γ-d-glutamic acid (γdPGA). We previously generated murine IgG3 monoclonal antibodies (mAbs) to γdPGA that were protective in a murine model of pulmonary anthrax. IgG3 antibodies are characteristic of the murine response to polysaccharide antigens. The goal of the present study was to produce subclass switch variants of the γdPGA mAbs (IgG3 → IgG1 → IgG2b → IgG2a) and assess the contribution of subclass to antibody affinity and protection. Subclass switch antibodies had identical variable regions but differed in their heavy chains. The results showed that a switch from the protective IgG3 to IgG1, IgG2b or IgG2a was accompanied by i) a loss of protective activity ii) a change in mAb binding to the capsular matrix, and iii) a loss of affinity. These results identify a role for the heavy chain constant region in mAb binding. Hybrid mAbs were constructed in which the CH1, CH2 or CH3 heavy chain constant domains from a non-protective, low binding IgG2b mAb were swapped into the protective IgG3 mAb. The IgG3 mAb that contained the CH1 domain from IgG2b showed no loss of affinity or protection. In contrast, swapping the CH2 or CH3 domains from IgG2b into IgG3 produced a reduction in affinity and a loss of protection. These studies identify a role for the constant region of IgG heavy chains in affinity and protection against an encapsulated bacterial pathogen.


Subject(s)
Anthrax/immunology , Bacillus anthracis/immunology , Immunoglobulin Constant Regions/immunology , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/immunology , Animals , Anthrax/microbiology , Antibodies, Monoclonal/immunology , Antibody Affinity , Antigen-Antibody Reactions , Bacterial Capsules/immunology , Glutamic Acid/immunology , Immunoglobulin Class Switching , Immunoglobulin G/chemistry , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary
9.
Infect Immun ; 80(7): 2414-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22526673

ABSTRACT

The development of therapeutics against biothreats requires that we understand the pathogenesis of the disease in relevant animal models. The rabbit model of inhalational anthrax is an important tool in the assessment of potential therapeutics against Bacillus anthracis. We investigated the roles of B. anthracis capsule and toxins in the pathogenesis of inhalational anthrax in rabbits by comparing infection with the Ames strain versus isogenic mutants with deletions of the genes for the capsule operon (capBCADE), lethal factor (lef), edema factor (cya), or protective antigen (pagA). The absence of capsule or protective antigen (PA) resulted in complete avirulence, while the presence of either edema toxin or lethal toxin plus capsule resulted in lethality. The absence of toxin did not influence the ability of B. anthracis to traffic to draining lymph nodes, but systemic dissemination required the presence of at least one of the toxins. Histopathology studies demonstrated minimal differences among lethal wild-type and single toxin mutant strains. When rabbits were coinfected with the Ames strain and the PA- mutant strain, the toxin produced by the Ames strain was not able to promote dissemination of the PA- mutant, suggesting that toxigenic action occurs in close proximity to secreting bacteria. Taken together, these findings suggest that a major role for toxins in the pathogenesis of anthrax is to enable the organism to overcome innate host effector mechanisms locally and that much of the damage during the later stages of infection is due to the interactions of the host with the massive bacterial burden.


Subject(s)
Anthrax/microbiology , Anthrax/pathology , Antigens, Bacterial/biosynthesis , Bacillus anthracis/pathogenicity , Bacterial Toxins/biosynthesis , Virulence Factors/biosynthesis , Animals , Anthrax/mortality , Antigens, Bacterial/genetics , Bacterial Capsules/genetics , Bacterial Toxins/genetics , Disease Models, Animal , Female , Gene Deletion , Histocytochemistry , Rabbits , Survival Analysis , Virulence
10.
Diagn Microbiol Infect Dis ; 64(2): 229-32, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19345041

ABSTRACT

Latex agglutination has been used to detect capsular polysaccharides from a variety of bacteria in body fluids. A latex agglutination assay was constructed for detection of the poly-gamma-D-glutamic acid (gammaDPGA) capsular polypeptide of Bacillus anthracis in serum from animal models of pulmonary anthrax. The assay was able to detect gammaDPGA in serum from infected animals at concentrations of 100 to 200 ng/mL.


Subject(s)
Anthrax/diagnosis , Antigens, Bacterial/analysis , Bacillus anthracis/isolation & purification , Bacterial Capsules/chemistry , Polyglutamic Acid/analogs & derivatives , Animals , Antigens, Bacterial/immunology , Bacillus anthracis/chemistry , Bacillus anthracis/immunology , Bacterial Capsules/immunology , Latex Fixation Tests/methods , Mice , Mice, Inbred BALB C , Polyglutamic Acid/analysis , Polyglutamic Acid/immunology , Serum/chemistry
11.
Infect Immun ; 77(4): 1649-63, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19168734

ABSTRACT

Bacillus anthracis is the causative agent of anthrax. We have developed a novel whole-bacterial-cell anthrax vaccine utilizing B. anthracis that is killed but metabolically active (KBMA). Vaccine strains that are asporogenic and nucleotide excision repair deficient were engineered by deleting the spoIIE and uvrAB genes, rendering B. anthracis extremely sensitive to photochemical inactivation with S-59 psoralen and UV light. We also introduced point mutations into the lef and cya genes, which allowed inactive but immunogenic toxins to be produced. Photochemically inactivated vaccine strains maintained a high degree of metabolic activity and secreted protective antigen (PA), lethal factor, and edema factor. KBMA B. anthracis vaccines were avirulent in mice and induced less injection site inflammation than recombinant PA adsorbed to aluminum hydroxide gel. KBMA B. anthracis-vaccinated animals produced antibodies against numerous anthrax antigens, including high levels of anti-PA and toxin-neutralizing antibodies. Vaccination with KBMA B. anthracis fully protected mice against challenge with lethal doses of toxinogenic unencapsulated Sterne 7702 spores and rabbits against challenge with lethal pneumonic doses of fully virulent Ames strain spores. Guinea pigs vaccinated with KBMA B. anthracis were partially protected against lethal Ames spore challenge, which was comparable to vaccination with the licensed vaccine anthrax vaccine adsorbed. These data demonstrate that KBMA anthrax vaccines are well tolerated and elicit potent protective immune responses. The use of KBMA vaccines may be broadly applicable to bacterial pathogens, especially those for which the correlates of protective immunity are unknown.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Antibodies, Bacterial/blood , Bacillus anthracis , Vaccines, Inactivated/immunology , Animals , Anthrax/microbiology , Anthrax/prevention & control , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/genetics , Antigens, Bacterial/immunology , Bacillus anthracis/genetics , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacillus anthracis/radiation effects , Female , Furocoumarins , Guinea Pigs , Immunity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mutation , Rabbits , Spores, Bacterial/genetics , Ultraviolet Rays , Vaccination , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Virulence
12.
Protein J ; 27(5): 292-302, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18649128

ABSTRACT

This article reports the design of a bivalent protein ligand with dual use in therapy and diagnosis of anthrax caused by Bacillus anthracis. The ligand specifically binds to PA and thereby blocks the intracellular delivery of LF and EF toxins that, respectively, cause cell lysis and edema. The ligand is a chimeric scaffold with two PA-binding domains (called VWA) linked to an IgG-Fc frame. Molecular modeling and binding measurements reveal that the VWA-Fc dimer binds to PA with high affinity (K(D)=0.2 nM). An in vitro bio-luminescence assay shows that VWA-Fc (at nanomolar concentration) protects mouse macrophages from lysis by PA/LF. In vivo studies demonstrate that VWA-Fc at low doses (approximately 50 microg/animal) are able to rescue animals from lethal doses of PA/LF and B. anthracis spores. Finally, VWA-Fc is utilized as the capture molecule in the sensitive (down to 30 picomolar) detection of PA using surface plasmon resonance.


Subject(s)
Anthrax/diagnosis , Anthrax/therapy , Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Animals , Antigens, Bacterial/genetics , Cell Line , Humans , Ligands , Mice , Models, Molecular , Protein Structure, Quaternary , Sensitivity and Specificity
13.
Infect Immun ; 75(1): 152-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17060470

ABSTRACT

Bacillus anthracis is surrounded by a polypeptide capsule composed of poly-gamma-d-glutamic acid (gammaDPGA). In a previous study, we reported that a monoclonal antibody (MAb F26G3) reactive with the capsular polypeptide is protective in a murine model of pulmonary anthrax. The present study examined a library of six MAbs generated from mice immunized with gammaDPGA. Evaluation of MAb binding to the capsule by a capsular "quellung" type reaction showed a striking diversity in capsular effects. Most MAbs produced a rim type reaction that was characterized by a sharp increase followed directly by a decrease in refractive index at the capsular edge. Some MAbs produced a second capsular reaction well beneath the capsular edge, suggesting complexity in capsular architecture. Binding of MAbs to soluble gammaDPGA was assessed by a fluorescence perturbation assay in which a change in the MAb intrinsic fluorescence produced by ligand binding was used as a reporter for antigen-antibody interaction. The MAbs differed considerably in the complexity of the binding curves. MAbs producing rim type capsule reactions typically produced the more complex binding isotherms. Finally, the protective activity of the MAbs was compared in a murine model of pulmonary anthrax. One MAb was markedly less protective than the remaining five MAbs. Characteristics of the more protective MAbs included a relatively high affinity, an immunoglobulin G3 isotype, and a complex binding isotherm in the fluorescence perturbation assay. Given the relatively monotonous structure of gammaDPGA, the results demonstrate a striking diversity in the antigen binding behavior of gammaDPGA antibodies.


Subject(s)
Anthrax/immunology , Antibodies, Monoclonal/immunology , Bacterial Capsules/immunology , Glutamic Acid/immunology , Animals , Anthrax/prevention & control , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Bacillus anthracis/immunology , Bacterial Capsules/chemistry , Disease Models, Animal , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/prevention & control , Mice , Mice, Inbred BALB C , Peptides/immunology , Polymerase Chain Reaction
14.
Vaccine ; 24(31-32): 5872-80, 2006 Jul 26.
Article in English | MEDLINE | ID: mdl-16790303

ABSTRACT

Because of the high failure rate of antibiotic treatment in patients with anthrax there is a need for additional therapies such as passive immunization with therapeutic antibodies. In this study, we used codon-optimized plasmid DNAs (DNA vaccines) encoding Bacillus anthracis protective antigen (PA) to immunize rabbits for producing anti-anthrax antibodies for use in passive immunotherapy. The antisera generated with these DNA vaccines were of high titer as measured by ELISA. The antisera were also able to protect J774 macrophage cells by neutralizing the cytotoxic effect of exogenously added anthrax lethal toxin, and of the toxin released by B. anthracis (Sterne strain) spores following infection. In addition, the antisera passively protected mice against pulmonary challenge with an approximate 50 LD50 dose of B. anthracis (Sterne strain) spores. The protection in mice was obtained when the antiserum was given 1h before or 1h after challenge. We further demonstrated that IgG and F(ab')2 components purified from anti-PA rabbit hyperimmune sera retained similar levels of neutralizing activities against both exogenously added B. anthracis lethal toxin and toxin produced by B. anthracis (Sterne strain) spores. The high titer antisera we produced will enable an immunization strategy to supplement antibiotic therapy for improving the survival of patients with anthrax.


Subject(s)
Anthrax Vaccines/therapeutic use , Anthrax/prevention & control , Bacillus anthracis , Immune Sera , Immunization, Passive/methods , Pneumonia, Bacterial/prevention & control , Vaccines, DNA/therapeutic use , Animals , Anthrax/immunology , Anthrax Vaccines/immunology , Bacillus anthracis/immunology , Female , Immune Sera/immunology , Mice , Mice, Inbred DBA , Pneumonia, Bacterial/immunology , Rabbits , Vaccines, DNA/immunology
15.
Proc Natl Acad Sci U S A ; 101(14): 5042-7, 2004 Apr 06.
Article in English | MEDLINE | ID: mdl-15051894

ABSTRACT

Bacillus anthracis is surrounded by an antiphagocytic polypeptide capsule composed of poly gamma-D-glutamic acid (gammaDPGA). gammaDPGA has been identified recently as a potential target for vaccine development. Studies of the role of gammaDPGA in disease have been hampered by the poor Ab response to this antigen and the lack of immunochemical reagents. As a consequence, neither the extent of gammaDPGA production during anthrax nor the protective activity of gammaDPGA Abs in inhalation anthrax are known. Here we report production of IgG Abs to gammaDPGA in mice following an immunization regimen using gammaDPGA in combination with agonist mAbs to CD40. mAbs were produced that are specific for gammaDPGA. Passive immunization with gammaDPGA mAbs protected >90% of mice in a pulmonary model of anthrax that was lethal in control mice (P < 0.0001). Use of gammaDPGA mAb in an antigen detection immunoassay found that the appearance of gammaDPGA in serum coincided with the emergence of bacteremia. These studies identify CD40 stimulation as a means for production of Ab and generation of mAbs against a weakly immunogenic antigen and demonstrate that the capsule is an effective target for immunoprotection and for antigen detection in the diagnosis of anthrax.


Subject(s)
Anthrax/diagnosis , Antibodies, Monoclonal/immunology , Antigens, Bacterial/blood , Bacillus anthracis/immunology , Animals , Anthrax/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Enzyme-Linked Immunosorbent Assay , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...