Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
2.
PLoS Pathog ; 18(12): e1010735, 2022 12.
Article in English | MEDLINE | ID: mdl-36534695

ABSTRACT

Inhalation anthrax has three clinical stages: early-prodromal, intermediate-progressive, and late-fulminant. We report the comprehensive characterization of anthrax toxins, including total protective antigen (PA), total lethal factor (LF), total edema factor (EF), and their toxin complexes, lethal toxin and edema toxin in plasma, during the course of inhalation anthrax in 23 cynomolgus macaques. The toxin kinetics were predominantly triphasic with an early rise (phase-1), a plateau/decline (phase-2), and a final rapid rise (phase-3). Eleven animals had shorter survival times, mean±standard deviation of 58.7±7.6 hours (fast progression), 11 animals had longer survival times, 113±34.4 hours (slow progression), and one animal survived. Median (lower-upper quartile) LF levels at the end-of-phase-1 were significantly higher in animals with fast progression [138 (54.9-326) ng/mL], than in those with slow progression [23.8 (15.6-26.3) ng/mL] (p = 0.0002), and the survivor (11.1 ng/mL). The differences were also observed for other toxins and bacteremia. Animals with slow progression had an extended phase-2 plateau, with low variability of LF levels across all time points and animals. Characterization of phase-2 toxin levels defined upper thresholds; critical levels for exiting phase-2 and entering the critical phase-3, 342 ng/mL (PA), 35.8 ng/mL (LF), and 1.10 ng/mL (EF). The thresholds were exceeded earlier in animals with fast progression (38.5±7.4 hours) and later in animals with slow progression (78.7±15.2 hours). Once the threshold was passed, toxin levels rose rapidly in both groups to the terminal stage. The time from threshold to terminal was rapid and similar; 20.8±7.4 hours for fast and 19.9±7.5 hours for slow progression. The three toxemic phases were aligned with the three clinical stages of anthrax for fast and slow progression which showed that anthrax progression is toxin- rather than time-dependent. This first comprehensive evaluation of anthrax toxins provides new insights into disease progression.


Subject(s)
Anthrax , Bacillus anthracis , Respiratory Tract Infections , Animals , Antigens, Bacterial , Macaca mulatta
3.
Clin Infect Dis ; 75(Suppl 3): S364-S372, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36251557

ABSTRACT

This report describes a 49-year-old male construction worker who acquired a Bacillus anthracis infection after working on a sheep farm. He experienced a severe respiratory infection, septic shock, and hemorrhagic meningoencephalitis with severe intracranial hypertension. After several weeks with multiple organ dysfunction syndrome, he responded favorably to antibiotic treatment. Three weeks into his hospitalization, an intracranial hemorrhage and cerebral edema led to an abrupt deterioration in his neurological status. A single dose of raxibacumab was added to his antimicrobial regimen on hospital day 27. His overall status, both clinical and radiographic, improved within a few days. He was discharged 2 months after admission and appears to have fully recovered.


Subject(s)
Anthrax , Bacillus anthracis , Meningitis , Animals , Anthrax/complications , Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Male , Meningitis/drug therapy , Respiratory Tract Infections , Sheep
4.
Clin Infect Dis ; 75(Suppl 3): S354-S363, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36251561

ABSTRACT

Bacillus anthracis has traditionally been considered the etiologic agent of anthrax. However, anthrax-like illness has been documented in welders and other metal workers infected with Bacillus cereus group spp. harboring pXO1 virulence genes that produce anthrax toxins. We present 2 recent cases of severe pneumonia in welders with B. cereus group infections and discuss potential risk factors for infection and treatment options, including antitoxin.


Subject(s)
Anthrax , Antitoxins , Bacillus anthracis , Anthrax/diagnosis , Anthrax/drug therapy , Bacillus anthracis/genetics , Bacillus cereus/genetics , Humans , Metal Workers , Plasmids
5.
J Infect Dis ; 223(2): 319-325, 2021 02 03.
Article in English | MEDLINE | ID: mdl-32697310

ABSTRACT

BACKGROUND: Inhalational anthrax is rare and clinical experience limited. Expert guidelines recommend treatment with combination antibiotics including protein synthesis-inhibitors to decrease toxin production and increase survival, although evidence is lacking. METHODS: Rhesus macaques exposed to an aerosol of Bacillus anthracis spores were treated with ciprofloxacin, clindamycin, or ciprofloxacin + clindamycin after becoming bacteremic. Circulating anthrax lethal factor and protective antigen were quantitated pretreatment and 1.5 and 12 hours after beginning antibiotics. RESULTS: In the clindamycin group, 8 of 11 (73%) survived demonstrating its efficacy for the first time in inhalational anthrax, compared to 9 of 9 (100%) with ciprofloxacin, and 8 of 11 (73%) with ciprofloxacin + clindamycin. These differences were not statistically significant. There were no significant differences between groups in lethal factor or protective antigen levels from pretreatment to 12 hours after starting antibiotics. Animals that died after clindamycin had a greater incidence of meningitis compared to those given ciprofloxacin or ciprofloxacin + clindamycin, but numbers of animals were very low and no definitive conclusion could be reached. CONCLUSION: Treatment of inhalational anthrax with clindamycin was as effective as ciprofloxacin in the nonhuman primate. Addition of clindamycin to ciprofloxacin did not enhance reduction of circulating toxin levels.


Subject(s)
Anthrax/blood , Anthrax/prevention & control , Antigens, Bacterial/blood , Bacillus anthracis/drug effects , Bacillus anthracis/physiology , Bacterial Toxins/blood , Ciprofloxacin/therapeutic use , Clindamycin/therapeutic use , Respiratory Tract Infections/blood , Respiratory Tract Infections/prevention & control , Animals , Anthrax/microbiology , Anthrax/mortality , Anti-Bacterial Agents/therapeutic use , Biomarkers , Ciprofloxacin/pharmacology , Clindamycin/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Macaca mulatta , Prognosis , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Treatment Outcome
6.
J Immunol ; 204(5): 1263-1273, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31932496

ABSTRACT

The poly-γ-glutamic acid (PGA) capsule produced by Bacillus anthracis is composed entirely of d-isomer glutamic acid, whereas nonpathogenic Bacillus species produce mixed d-, l-isomer PGAs. To determine if B. anthracis PGA confers a pathogenic advantage over other PGAs, we compared the responses of human innate immune cells to B. anthracis PGA and PGAs from nonpathogenic B. subtilis subsp. chungkookjang and B. licheniformis Monocytes and immature dendritic cells (iDCs) responded differentially to the PGAs, with B. anthracis PGA being least stimulatory and B. licheniformis PGA most stimulatory. All three elicited IL-8 and IL-6 from monocytes, but B. subtilis PGA also elicited IL-10 and TNF-α, whereas B. licheniformis PGA elicited all those plus IL-1ß. Similarly, all three PGAs elicited IL-8 from iDCs, but B. subtilis PGA also elicited IL-6, and B. licheniformis PGA elicited those plus IL-12p70, IL-10, IL-1ß, and TNF-α. Only B. licheniformis PGA induced dendritic cell maturation. TLR assays also yielded differential results. B. subtilis PGA and B. licheniformis PGA both elicited more TLR2 signal than B. anthracis PGA, but only responses to B. subtilis PGA were affected by a TLR6 neutralizing Ab. B. licheniformis PGA elicited more TLR4 signal than B. anthracis PGA, whereas B. subtilis PGA elicited none. B. anthracis PGA persisted longer in high m.w. form in monocyte and iDC cultures than the other PGAs. Reducing the m.w. of B. anthracis PGA reduced monocytes' cytokine responses. We conclude that B. anthracis PGA is recognized less effectively by innate immune cells than PGAs from nonpathogenic Bacillus species, resulting in failure to induce a robust host response, which may contribute to anthrax pathogenesis.


Subject(s)
Bacillus anthracis/immunology , Bacillus licheniformis/immunology , Bacillus subtilis/immunology , Dendritic Cells/immunology , Immunity, Innate , Macrophages/immunology , Monocytes/immunology , Polyglutamic Acid/immunology , Cytokines/immunology , Female , Humans , Male
7.
Anal Bioanal Chem ; 411(12): 2493-2509, 2019 May.
Article in English | MEDLINE | ID: mdl-30911800

ABSTRACT

Inhalation of Bacillus anthracis spores can cause a rapidly progressing fatal infection. B. anthracis secretes three protein toxins: lethal factor (LF), edema factor (EF), and protective antigen (PA). EF and LF may circulate as free or PA-bound forms. Both free EF (EF) and PA-bound-EF (ETx) have adenylyl cyclase activity converting ATP to cAMP. We developed an adenylyl cyclase activity-based method for detecting and quantifying total EF (EF+ETx) in plasma. The three-step method includes magnetic immunocapture with monoclonal antibodies, reaction with ATP generating cAMP, and quantification of cAMP by isotope-dilution HPLC-MS/MS. Total EF was quantified from 5PL regression of cAMP vs ETx concentration. The detection limit was 20 fg/mL (225 zeptomoles/mL for the 89 kDa protein). Relative standard deviations for controls with 0.3, 6.0, and 90 pg/mL were 11.7-16.6% with 91.2-99.5% accuracy. The method demonstrated 100% specificity in 238 human serum/plasma samples collected from unexposed healthy individuals, and 100% sensitivity in samples from 3 human and 5 rhesus macaques with inhalation anthrax. Analysis of EF in the rhesus macaques showed that it was detected earlier post-exposure than B. anthracis by culture and PCR. Similar to LF, the kinetics of EF over the course of infection were triphasic, with an initial rise (phase-1), decline (phase-2), and final rapid rise (phase-3). EF levels were ~ 2-4 orders of magnitude lower than LF during phase-1 and phase-2 and only ~ 6-fold lower at death/euthanasia. Analysis of EF improves early diagnosis and adds to our understanding of anthrax toxemia throughout infection. The LF/EF ratio may also indicate the stage of infection and need for advanced treatments.


Subject(s)
Anthrax/pathology , Antigens, Bacterial/blood , Bacillus anthracis/pathogenicity , Bacterial Toxins/blood , Chromatography, High Pressure Liquid/methods , Respiratory Tract Infections/pathology , Tandem Mass Spectrometry/methods , Toxemia/pathology , Adenosine Triphosphate/metabolism , Animals , Anthrax/blood , Case-Control Studies , Cyclic AMP/biosynthesis , Disease Progression , Enzyme-Linked Immunosorbent Assay , Humans , Limit of Detection , Macaca mulatta , Polymerase Chain Reaction , Respiratory Tract Infections/blood , Toxemia/blood , Toxemia/microbiology
8.
Analyst ; 144(7): 2264-2274, 2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30810119

ABSTRACT

Anthrax protective antigen (83 kDa, PA83) is an essential component of two major binary toxins produced by Bacillus anthracis, lethal toxin (LTx) and edema toxin (ETx). During infection, LTx and ETx contribute to immune collapse, endothelial dysfunction, hemorrhage and high mortality. Following protease cleavage on cell receptors or in circulation, the 20 kDa (PA20) N-terminus is released, activating the 63 kDa (PA63) form which binds lethal factor (LF) and edema factor (EF), facilitating their entry into their cellular targets. Several ELISA-based PA methods previously developed are primarily qualitative or semi-quantitative. Here, we combined protein immunocapture, tryptic digestion and isotope dilution liquid chromatography-mass spectrometry (LC-MS/MS), to develop a highly selective and sensitive method for detection and accurate quantification of total-PA (PA83 + PA63) and PA83. Two tryptic peptides in the 63 kDa region measure total-PA and three in the 20 kDa region measure PA83 alone. Detection limits range from 1.3-2.9 ng mL-1 PA in 100 µL of plasma. Spiked recovery experiments with combinations of PA83, PA63, LF and EF in plasma showed that PA63 and PA83 were quantified accurately against the PA83 standard and that LF and EF did not interfere with accuracy. Applied to a study of inhalation anthrax in rhesus macaques, total-PA suggested triphasic kinetics, similar to that previously observed for LF and EF. This study is the first to report circulating PA83 in inhalation anthrax, typically at less than 4% of the levels of PA63, providing the first evidence that activated PA63 is the primary form of PA throughout infection.


Subject(s)
Antigens, Bacterial/blood , Bacillus anthracis/immunology , Bacterial Toxins/blood , Immunoassay/methods , Limit of Detection , Mass Spectrometry , Animals , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Macaca mulatta
9.
Anal Biochem ; 543: 97-107, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29224733

ABSTRACT

Anthrax lethal factor (LF) is a zinc-dependent endoprotease and a critical virulence factor for Bacillus anthracis, the causative agent of anthrax. The mass spectrometry (MS) method for total-LF quantification includes three steps; 1) LF specific antibody capture/concentration, 2) LF-specific hydrolysis of a peptide substrate, and 3) detection and quantification of LF-cleaved peptides by isotope-dilution MALDI-TOF/MS. Recombinant LF spiked plasma was used for calibration and quality control (QC) materials. Specificity was 100% from analysis of serum and plasma from 383 non-infected humans, 31 rabbits, and 24 rhesus macaques. Sensitivity was 100% from 32 human clinical anthrax cases including infections by inhalation, ingestion, cutaneous and injection exposures and experimental infections for 29 rabbits and 24 rhesus macaques with inhalation anthrax. Robustness evaluation included sample storage, serum and plasma, antimicrobial and antitoxin effects and long-term performance. Data from 100 independent runs gave detection limits 0.01 ng/mL (111 amol/mL) for the 4-h method and 0.0027 ng/mL (30 amol/mL) for an alternate 20-h method. QC precision ranged from 7.7 to 14.8% coefficient of variation and accuracy from 0.2 to 9.8% error. The validated LF MS method provides sensitive quantification of anthrax total-LF using a robust high throughput platform for early diagnosis and evaluation of therapeutics during an anthrax emergency.


Subject(s)
Anthrax/diagnosis , Anthrax/drug therapy , Antigens, Bacterial/analysis , Bacillus anthracis/chemistry , Bacterial Toxins/analysis , Animals , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Calibration , Humans , Macaca mulatta , Quality Control , Rabbits , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Emerg Infect Dis ; 23(1): 56-65, 2017 01.
Article in English | MEDLINE | ID: mdl-27983504

ABSTRACT

We studied anthrax immune globulin intravenous (AIG-IV) use from a 2009-2010 outbreak of Bacillus anthracis soft tissue infection in injection drug users in Scotland, UK, and we compared findings from 15 AIG-IV recipients with findings from 28 nonrecipients. Death rates did not differ significantly between recipients and nonrecipients (33% vs. 21%). However, whereas only 8 (27%) of 30 patients at low risk for death (admission sequential organ failure assessment score of 0-5) received AIG-IV, 7 (54%) of the 13 patients at high risk for death (sequential organ failure assessment score of 6-11) received treatment. AIG-IV recipients had surgery more often and, among survivors, had longer hospital stays than did nonrecipients. AIG-IV recipients were sicker than nonrecipients. This difference and the small number of higher risk patients confound assessment of AIG-IV effectiveness in this outbreak.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Antitoxins/therapeutic use , Disease Outbreaks , Immunoglobulin G/therapeutic use , Soft Tissue Infections/drug therapy , Substance Abuse, Intravenous/drug therapy , Adult , Anthrax/epidemiology , Anthrax/microbiology , Anthrax/mortality , Bacillus anthracis/pathogenicity , Bacillus anthracis/physiology , Drug Therapy, Combination , Drug Users , Female , Heroin/administration & dosage , Humans , Male , Scotland/epidemiology , Soft Tissue Infections/epidemiology , Soft Tissue Infections/microbiology , Soft Tissue Infections/mortality , Substance Abuse, Intravenous/epidemiology , Substance Abuse, Intravenous/microbiology , Substance Abuse, Intravenous/mortality , Survival Analysis , Treatment Outcome
11.
PLoS One ; 11(6): e0156987, 2016.
Article in English | MEDLINE | ID: mdl-27257909

ABSTRACT

Bacillus cereus isolates have been described harboring Bacillus anthracis toxin genes, most notably B. cereus G9241, and capable of causing severe and fatal pneumonias. This report describes the characterization of a B. cereus isolate, BcFL2013, associated with a naturally occurring cutaneous lesion resembling an anthrax eschar. Similar to G9241, BcFL2013 is positive for the B. anthracis pXO1 toxin genes, has a multi-locus sequence type of 78, and a pagA sequence type of 9. Whole genome sequencing confirms the similarity to G9241. In addition to the chromosome having an average nucleotide identity of 99.98% when compared to G9241, BcFL2013 harbors three plasmids with varying homology to the G9241 plasmids (pBCXO1, pBC210 and pBFH_1). This is also the first report to include serologic testing of patient specimens associated with this type of B. cereus infection which resulted in the detection of anthrax lethal factor toxemia, a quantifiable serum antibody response to protective antigen (PA), and lethal toxin neutralization activity.


Subject(s)
Anthrax/metabolism , Anthrax/microbiology , Antigens, Bacterial/metabolism , Bacillus cereus/metabolism , Bacillus cereus/pathogenicity , Bacterial Toxins/metabolism , Aged , Anthrax/pathology , Bacillus cereus/isolation & purification , Humans , Male
12.
Health Secur ; 13(6): 365-77, 2015.
Article in English | MEDLINE | ID: mdl-26690378

ABSTRACT

Concern about use of anthrax as a bioweapon prompted development of novel anthrax antitoxins for treatment. Clinical guidelines for the treatment of anthrax recommend antitoxin therapy in combination with intravenous antimicrobials; however, a large-scale or mass anthrax incident may exceed antitoxin availability and create a need for judicious antitoxin use. We conducted a systematic review of antitoxin treatment of inhalation anthrax in humans and experimental animals to inform antitoxin recommendations during a large-scale or mass anthrax incident. A comprehensive search of 11 databases and the FDA website was conducted to identify relevant animal studies and human reports: 28 animal studies and 3 human cases were identified. Antitoxin monotherapy at or shortly after symptom onset demonstrates increased survival compared to no treatment in animals. With early treatment, survival did not differ between antimicrobial monotherapy and antimicrobial-antitoxin therapy in nonhuman primates and rabbits. With delayed treatment, antitoxin-antimicrobial treatment increased rabbit survival. Among human cases, addition of antitoxin to combination antimicrobial treatment was associated with survival in 2 of the 3 cases treated. Despite the paucity of human data, limited animal data suggest that adjunctive antitoxin therapy may improve survival. Delayed treatment studies suggest improved survival with combined antitoxin-antimicrobial therapy, although a survival difference compared with antimicrobial therapy alone was not demonstrated statistically. In a mass anthrax incident with limited antitoxin supplies, antitoxin treatment of individuals who have not demonstrated a clinical benefit from antimicrobials, or those who present with more severe illness, may be warranted. Additional pathophysiology studies are needed, and a point-of-care assay correlating toxin levels with clinical status may provide important information to guide antitoxin use during a large-scale anthrax incident.


Subject(s)
Anthrax/drug therapy , Antibodies, Monoclonal/therapeutic use , Antitoxins/therapeutic use , Immunoglobulin G/therapeutic use , Respiratory Tract Infections/drug therapy , Administration, Intravenous , Animals , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, Bacterial/immunology , Bioterrorism , Drug Therapy, Combination , Humans , Immunoglobulins, Intravenous/therapeutic use , Mass Casualty Incidents , Rabbits
13.
Toxins (Basel) ; 7(9): 3497-511, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-26404376

ABSTRACT

Mass spectrometry has recently become a powerful technique for bacterial identification. Mass spectrometry approaches generally rely upon introduction of the bacteria into a matrix-assisted laser-desorption time-of-flight (MALDI-TOF) mass spectrometer with mass spectrometric recognition of proteins specific to that organism that form a reliable fingerprint. With some bacteria, such as Bacillus anthracis and Clostridium botulinum, the health threat posed by these organisms is not the organism itself, but rather the protein toxins produced by the organisms. One such example is botulinum neurotoxin (BoNT), a potent neurotoxin produced by C. botulinum. There are seven known serotypes of BoNT, A-G, and many of the serotypes can be further differentiated into toxin variants, which are up to 99.9% identical in some cases. Mass spectrometric proteomic techniques have been established to differentiate the serotype or toxin variant of BoNT produced by varied strains of C. botulinum. Detection of potent biological toxins requires high analytical sensitivity and mass spectrometry based methods have been developed to determine the enzymatic activity of BoNT and the anthrax lethal toxins produced by B. anthracis. This enzymatic activity, unique for each toxin, is assessed with detection of the toxin-induced cleavage of strategically designed peptide substrates by MALDI-TOF mass spectrometry offering unparalleled specificity. Furthermore, activity assays allow for the assessment of the biological activity of a toxin and its potential health risk. Such methods have become important diagnostics for botulism and anthrax. Here, we review mass spectrometry based methods for the enzymatic activity of BoNT and the anthrax lethal factor toxin.


Subject(s)
Bacterial Proteins/analysis , Bacterial Toxins/analysis , Antigens, Bacterial/analysis , Bacillus anthracis/metabolism , Botulinum Toxins, Type A/analysis , Clostridium botulinum/metabolism , Neurotoxins/analysis , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Anal Bioanal Chem ; 407(10): 2847-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25673244

ABSTRACT

Inhalation anthrax has a rapid progression and high fatality rate. Pathology and death from inhalation of Bacillus anthracis spores are attributed to the actions of secreted protein toxins. Protective antigen (PA) binds and imports the catalytic component lethal factor (LF), a zinc endoprotease, and edema factor (EF), an adenylyl cyclase, into susceptible cells. PA-LF is termed lethal toxin (LTx) and PA-EF, edema toxin. As the universal transporter for both toxins, PA is an important target for vaccination and immunotherapeutic intervention. However, its quantification has been limited to methods of relatively low analytic sensitivity. Quantification of LTx may be more clinically relevant than LF or PA alone because LTx is the toxic form that acts on cells. A method was developed for LTx-specific quantification in plasma using anti-PA IgG magnetic immunoprecipitation of PA and quantification of LF activity that co-purified with PA. The method was fast (<4 h total time to detection), sensitive at 0.033 ng/mL LTx in plasma for the fast analysis (0.0075 ng/mL LTx in plasma for an 18 h reaction), precise (6.3-9.9% coefficient of variation), and accurate (0.1-12.7%error; n ≥ 25). Diagnostic sensitivity was 100% (n = 27 animal/clinical cases). Diagnostic specificity was 100% (n = 141). LTx was detected post-antibiotic treatment in 6/6 treated rhesus macaques and 3/3 clinical cases of inhalation anthrax and as long as 8 days post-treatment. Over the course of infection in two rhesus macaques, LTx was first detected at 0.101 and 0.237 ng/mL at 36 h post-exposure and increased to 1147 and 12,107 ng/mL in late-stage anthrax. This demonstrated the importance of LTx as a diagnostic and therapeutic target. This method provides a sensitive, accurate tool for anthrax toxin detection and evaluation of PA-directed therapeutics.


Subject(s)
Anthrax/diagnosis , Antigens, Bacterial/analysis , Bacterial Toxins/analysis , Respiratory Tract Infections/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Animals , Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Antibody Specificity , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Toxins/blood , Bacterial Toxins/immunology , Humans , Immunoprecipitation/methods , Macaca mulatta , Magnetics , Molecular Sequence Data , Reproducibility of Results , Respiratory Tract Infections/drug therapy , Sensitivity and Specificity , Time Factors
15.
J Immunol Methods ; 408: 78-88, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24857756

ABSTRACT

Inhalation anthrax is a rare but acute infectious disease following adsorption of Bacillus anthracis spores through the lungs. The disease has a high fatality rate if untreated, but early and correct diagnosis has a significant impact on case patient recovery. The early symptoms of inhalation anthrax are, however, non-specific and current anthrax diagnostics are primarily dependent upon culture and confirmatory real-time PCR. Consequently, there may be a significant delay in diagnosis and targeted treatment. Rapid, culture-independent diagnostic tests are therefore needed, particularly in the context of a large scale emergency response. The aim of this study was to evaluate the ability of monoclonal antibodies to detect anthrax toxin proteins that are secreted early in the course of B. anthracis infection using a time-resolved fluorescence (TRF) immunoassay. We selected monoclonal antibodies that could detect protective antigen (PA), as PA83 and also PA63 and LF in the lethal toxin complex. The assay reliable detection limit (RDL) was 6.63×10(-6)µM (0.551ng/ml) for PA83 and 2.51×10(-5)µM (1.58ng/ml) for PA63. Despite variable precision and accuracy of the assay, PA was detected in 9 out of 10 sera samples from anthrax confirmed case patients with cutaneous (n=7), inhalation (n=2), and gastrointestinal (n=1) disease. Anthrax Immune Globulin (AIG), which has been used in treatment of clinical anthrax, interfered with detection of PA. This study demonstrates a culture-independent method of diagnosing anthrax through the use of monoclonal antibodies to detect PA and LF in the lethal toxin complex.


Subject(s)
Anthrax/diagnosis , Antibodies, Monoclonal , Antigens, Bacterial/blood , Bacillus anthracis/immunology , Bacterial Toxins/blood , Europium , Immunoassay/methods , Anthrax/blood , Anthrax/immunology , Anthrax Vaccines/immunology , Biomarkers/blood , Case-Control Studies , Early Diagnosis , Fluoroimmunoassay , Humans , Limit of Detection , Predictive Value of Tests , Reproducibility of Results
16.
Emerg Infect Dis ; 20(2): 310-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24447456

ABSTRACT

Bacillus anthracis was identified in a 61-year-old man hospitalized in Minnesota, USA. Cooperation between the hospital and the state health agency enhanced prompt identification of the pathogen. Treatment comprising antimicrobial drugs, anthrax immune globulin, and pleural drainage led to full recovery; however, the role of passive immunization in anthrax treatment requires further evaluation.


Subject(s)
Anthrax/microbiology , Antibodies, Bacterial/blood , Antibodies, Viral/blood , Antigens, Bacterial/blood , Bacillus anthracis/isolation & purification , Bacterial Toxins/blood , Respiratory Tract Infections/microbiology , Anthrax/diagnosis , Anthrax/immunology , Anthrax/therapy , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/pathogenicity , Drainage, Postural , Drug Administration Schedule , Humans , Immunoglobulins, Intravenous/therapeutic use , Male , Middle Aged , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/immunology , Respiratory Tract Infections/therapy , Treatment Outcome
17.
Cell Microbiol ; 16(4): 504-18, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24152301

ABSTRACT

Polymorphonuclear leucocytes (PMNs) play a protective role during Bacillus anthracis infection. However, B. anthracis is able to subvert the PMN response effectively as evidenced by the high mortality rates of anthrax. One major virulence factor produced by B. anthracis, lethal toxin (LT), is necessary for dissemination in the BSL2 model of mouse infection. While human and mouse PMNs kill vegetative B. anthracis, short in vitro half-lives of PMNs have made it difficult to determine how or if LT alters their bactericidal function. Additionally, the role of LT intoxication on PMN's ability to migrate to inflammatory signals remains controversial. LF concentrations in both serum and major organs were determined from mice infected with B. anthracis Sterne strain at defined stages of infection to guide subsequent administration of purified toxin. Bactericidal activity of PMNs assessed using ex vivo cell culture assays showed significant defects in killing B. anthracis. In vivo PMN recruitment to inflammatory stimuli was significantly impaired at 24 h as assessed by real-time analysis of light-producing PMNs within the mouse. The observations described above suggest that LT serves dual functions; it both attenuates accumulation of PMNs at sites of inflammation and impairs PMNs bactericidal activity against vegetative B. anthracis.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Bacterial/toxicity , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Neutrophils/drug effects , Neutrophils/immunology , Animal Structures/chemistry , Animals , Anthrax/immunology , Anthrax/microbiology , Antigens, Bacterial/analysis , Bacterial Toxins/analysis , Cells, Cultured , Disease Models, Animal , Humans , Mice , Serum/chemistry
18.
PLoS One ; 7(2): e30201, 2012.
Article in English | MEDLINE | ID: mdl-22393351

ABSTRACT

Anthrax is caused by infection with Bacillus anthracis, a spore-forming gram-positive bacterium. A major virulence factor for B. anthracis is an immunomodulatory tripartite exotoxin that has been reported to alter immune cell chemotaxis and activation. It has been proposed that B. anthracis infections initiate through entry of spores into the regional draining lymph nodes where they germinate, grow, and disseminate systemically via the efferent lymphatics. If this model holds true, it would be predicted that surgical removal of infected tissues, debridement, would have little effect on the systemic dissemination of bacteria. This model was tested through the development of a mouse debridement model. It was found that removal of the site of subcutaneous infection in the ear increased the likelihood of survival and reduced the quantity of spores in the draining cervical lymph nodes (cLN). At the time of debridement 12 hours post-injection measurable levels of exotoxins were present in the ear, cLN, and serum, yet leukocytes within the cLN were activated; countering the concept that exotoxins inhibit the early inflammatory response to promote bacterial growth. We conclude that the initial entry of spores into the draining lymph node of cutaneous infections alone is not sufficient to cause systemic disease and that debridement should be considered as an adjunct to antibiotic therapy.


Subject(s)
Anthrax/surgery , Debridement/methods , Skin Diseases/surgery , Animals , Anthrax/drug therapy , Anthrax/microbiology , Anti-Bacterial Agents/therapeutic use , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Disease Models, Animal , Female , Flow Cytometry/methods , Lymph Nodes/metabolism , Lymph Nodes/microbiology , Mice , Skin Diseases/drug therapy , Skin Diseases/microbiology , Spores, Bacterial/metabolism , Stem Cells/metabolism
19.
J Infect Dis ; 204(9): 1321-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21908727

ABSTRACT

Cutaneous anthrax outbreaks occurred in Bangladesh from August to October 2009. As part of the epidemiological response and to confirm anthrax diagnoses, serum samples were collected from suspected case patients with observed cutaneous lesions. Anthrax lethal factor (LF), anti-protective antigen (anti-PA) immunoglobulin G (IgG), and anthrax lethal toxin neutralization activity (TNA) levels were determined in acute and convalescent serum of 26 case patients with suspected cutaneous anthrax from the first and largest of these outbreaks. LF (0.005-1.264 ng/mL) was detected in acute serum from 18 of 26 individuals. Anti-PA IgG and TNA were detected in sera from the same 18 individuals and ranged from 10.0 to 679.5 µg/mL and 27 to 593 units, respectively. Seroconversion to serum anti-PA and TNA was found only in case patients with measurable toxemia. This is the first report of quantitative analysis of serum LF in cutaneous anthrax and the first to associate acute stage toxemia with subsequent antitoxin antibody responses.


Subject(s)
Anthrax/epidemiology , Anthrax/immunology , Antigens, Bacterial/immunology , Antitoxins/blood , Bacterial Toxins/immunology , Disease Outbreaks , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bangladesh/epidemiology , Humans , Immunoglobulin G/blood , Skin Diseases, Bacterial
20.
Clin Vaccine Immunol ; 18(5): 743-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21389148

ABSTRACT

The Bacillus anthracis exosporium protein BclA contains an O-linked antigenic tetrasaccharide whose terminal sugar is known as anthrose (J. M. Daubenspeck et al., J. Biol. Chem. 279:30945-30953, 2004). We hypothesized that serologic responses to anthrose may have diagnostic value in confirming exposure to aerosolized B. anthracis. We evaluated the serologic responses to a synthetic anthrose-containing trisaccharide (ATS) in a group of five rhesus macaques that survived inhalation anthrax following exposure to B. anthracis Ames spores. Two of five animals (RM2 and RM3) were treated with ciprofloxacin starting at 48 hours postexposure and two (RM4 and RM5) at 72 h postexposure; one animal (RM1) was untreated. Infection was confirmed by blood culture and detection of anthrax toxin lethal factor (LF) in plasma. Anti-ATS IgG responses were determined at 14, 21, 28, and 35 days postexposure, with preexposure serum as a control. All animals, irrespective of ciprofloxacin treatment, mounted a specific, measurable anti-ATS IgG response. The earliest detectable responses were on days 14 (RM1, RM2, and RM5), 21 (RM4), and 28 (RM3). Specificity of the anti-ATS responses was demonstrated by competitive-inhibition enzyme immunoassay (CIEIA), in which a 2-fold (wt/wt) excess of carbohydrate in a bovine serum albumin (BSA) conjugate of the oligosaccharide (ATS-BSA) effected >94% inhibition, whereas a structural analog lacking the 3-hydroxy-3-methyl-butyryl moiety at the C-4" of the anthrosyl residue had no inhibition activity. These data suggest that anti-ATS antibody responses may be used to identify aerosol exposure to B. anthracis spores. The anti-ATS antibody responses were detectable during administration of ciprofloxacin.


Subject(s)
Antibodies, Bacterial/blood , Carbohydrates/immunology , Membrane Glycoproteins/immunology , Primate Diseases/diagnosis , Spores, Bacterial/immunology , Animals , Anthrax/diagnosis , Anthrax/immunology , Anthrax/veterinary , Antigens, Bacterial/immunology , Macaca mulatta , Primate Diseases/immunology , Skin Diseases, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL