Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Antimicrob Chemother ; 78(3): 810-816, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36738250

RESUMEN

OBJECTIVES: To evaluate the in vitro activity and in vivo efficacy of delafloxacin against Bacillus anthracis, the causative agent of anthrax. METHODS: MICs were obtained according to CLSI guidelines for 30 virulent isolates and 14 attenuated antibiotic-resistant strains. For the in vivo efficacy study, mice were administered delafloxacin (30-62.5 mg/kg) subcutaneously, or ciprofloxacin (30 mg/kg) intraperitoneally beginning at either 24 or 48 ±â€Š1 h post-challenge (post-exposure prophylaxis) and continued every 12 h for 14 days with study termination on day 30. The mean inhaled dose in the study was approximately 103 × LD50 equivalents, and the range was 87-120 × LD50. RESULTS: Delafloxacin (MIC90 = 0.004 mg/L) was 16-fold more potent than ciprofloxacin (MIC90 = 0.06 mg/L) against a 30-strain set of virulent B. anthracis. Against a panel of attenuated antibiotic-resistant strains, delafloxacin demonstrated potency ≥128-fold over that observed with ciprofloxacin. When evaluated in vivo, mice treated with all delafloxacin doses tested at 24 h post-challenge demonstrated equivalent survival compared with mice treated with the positive control ciprofloxacin. Because of the high challenge dose of spores, mice treated at 48 h showed rapid and high mortality in all groups including the positive control. Surviving animals in all delafloxacin- and ciprofloxacin-treated groups (24 and 48 h) showed complete splenic clearance of infection and <2.2 × 103 cfu/g lung tissue. CONCLUSIONS: Given the high bar set by the 100 × LD50 challenge dose in this study, the results from delafloxacin treatment are promising for the treatment of inhaled anthrax.


Asunto(s)
Carbunco , Bacillus anthracis , Animales , Ratones , Carbunco/tratamiento farmacológico , Antibacterianos/uso terapéutico , Ciprofloxacina , Pruebas de Sensibilidad Microbiana
2.
PLoS Pathog ; 16(8): e1008836, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32866212

RESUMEN

Anthrax is a major zoonotic disease of wildlife, and in places like West Africa, it can be caused by Bacillus anthracis in arid nonsylvatic savannahs, and by B. cereus biovar anthracis (Bcbva) in sylvatic rainforests. Bcbva-caused anthrax has been implicated in as much as 38% of mortality in rainforest ecosystems, where insects can enhance the transmission of anthrax-causing bacteria. While anthrax is well-characterized in mammals, its transmission by insects points to an unidentified anthrax-resistance mechanism in its vectors. In mammals, a secreted anthrax toxin component, 83 kDa Protective Antigen (PA83), binds to cell-surface receptors and is cleaved by furin into an evolutionary-conserved PA20 and a pore-forming PA63 subunits. We show that PA20 increases the resistance of Drosophila flies and Culex mosquitoes to bacterial challenges, without directly affecting the bacterial growth. We further show that the PA83 loop known to be cleaved by furin to release PA20 from PA63 is, in part, responsible for the PA20-mediated protection. We found that PA20 binds directly to the Toll activating peptidoglycan-recognition protein-SA (PGRP-SA) and that the Toll/NF-κB pathway is necessary for the PA20-mediated protection of infected flies. This effect of PA20 on innate immunity may also exist in mammals: we show that PA20 binds to human PGRP-SA ortholog. Moreover, the constitutive activity of Imd/NF-κB pathway in MAPKK Dsor1 mutant flies is sufficient to confer the protection from bacterial infections in a manner that is independent of PA20 treatment. Lastly, Clostridium septicum alpha toxin protects flies from anthrax-causing bacteria, showing that other pathogens may help insects resist anthrax. The mechanism of anthrax resistance in insects has direct implications on insect-mediated anthrax transmission for wildlife management, and with potential for applications, such as reducing the sensitivity of pollinating insects to bacterial pathogens.


Asunto(s)
Vacunas contra el Carbunco/administración & dosificación , Carbunco/tratamiento farmacológico , Antígenos Bacterianos/administración & dosificación , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/administración & dosificación , Drosophila melanogaster/crecimiento & desarrollo , Mosquitos Vectores/microbiología , Sustancias Protectoras/administración & dosificación , Animales , Carbunco/microbiología , Culex , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Femenino , Masculino
3.
J Infect Dis ; 223(2): 319-325, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32697310

RESUMEN

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.


Asunto(s)
Carbunco/sangre , Carbunco/prevención & control , Antígenos Bacterianos/sangre , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/fisiología , Toxinas Bacterianas/sangre , Ciprofloxacina/uso terapéutico , Clindamicina/uso terapéutico , Infecciones del Sistema Respiratorio/sangre , Infecciones del Sistema Respiratorio/prevención & control , Animales , Carbunco/microbiología , Carbunco/mortalidad , Antibacterianos/uso terapéutico , Biomarcadores , Ciprofloxacina/farmacología , Clindamicina/farmacología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Macaca mulatta , Pronóstico , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/mortalidad , Resultado del Tratamiento
4.
Microb Pathog ; 155: 104919, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33915206

RESUMEN

Burkholderia mallei is a gram-negative obligate animal pathogen that causes glanders, a highly contagious and potentially fatal disease of solipeds including horses, mules, and donkeys. Humans are also susceptible, and exposure can result in a wide range of clinical forms, i.e., subclinical infection, chronic forms with remission and exacerbation, or acute and potentially lethal septicemia and/or pneumonia. Due to intrinsic antibiotic resistance and the ability of the organisms to survive intracellularly, current treatment regimens are protracted and complicated; and no vaccine is available. As a consequence of these issues, and since B. mallei is infectious by the aerosol route, B. mallei is regarded as a major potential biothreat agent. To develop optimal medical countermeasures and diagnostic tests, well characterized animal models of human glanders are needed. The goal of this study was to perform a head-to-head comparison of models employing three commonly used nonhuman primate (NHP) species, the African green monkey (AGM), Rhesus macaque, and the Cynomolgus macaque. The natural history of infection and in vitro clinical, histopathological, immunochemical, and bacteriological parameters were examined. The AGMs were the most susceptible NHP to B. mallei; five of six expired within 14 days. Although none of the Rhesus or Cynomolgus macaques succumbed, the Rhesus monkeys exhibited abnormal signs and clinical findings associated with B. mallei infection; and the latter may be useful for modeling chronic B. mallei infection. Based on the disease progression observations, gross and histochemical pathology, and humoral and cellular immune response findings, the AGM appears to be the optimal model of acute, lethal glanders infection. AGM models of infection by B. pseudomallei, the etiologic agent of melioidosis, have been characterized recently. Thus, the selection of the AGM species provides the research community with a single NHP model for investigations on acute, severe, inhalational melioidosis and glanders.


Asunto(s)
Burkholderia mallei , Burkholderia pseudomallei , Muermo , Melioidosis , Aerosoles , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Muermo/diagnóstico , Caballos , Macaca mulatta
5.
BMC Immunol ; 21(1): 5, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32013893

RESUMEN

BACKGROUND: Melioidosis is endemic in Southeast Asia and Northern Australia and is caused by the Gram-negative, facultative intracellular pathogen Burkholderia pseudomallei. Diagnosis of melioidosis is often difficult because of the protean clinical presentation of the disease, and it may mimic other diseases, such as tuberculosis. There are many different strains of B. pseudomallei that have been isolated from patients with melioidosis, but it was not clear if they could cause a similar disease in a chronic BALB/c murine model of melioidosis. Hence, we wanted to examine chronically infected mice exposed to different strains of B. pseudomallei to determine if there were differences in the host immune response to the pathogen. RESULTS: We identified common host immune responses exhibited in chronically infected BALB/c mice, although there was some heterogeneity in the host response in chronically infected mice after exposure to different strains of B. pseudomallei. They all displayed pyogranulomatous lesions in their spleens with a large influx of monocytes/macrophages, NK cells, and neutrophils identified by flow cytometry. Sera from chronically infected mice by ELISA exhibited elevated IgG titers to the pathogen, and we detected by Luminex micro-bead array technology a significant increase in the expression of inflammatory cytokines/chemokines, such as IFN-γ, IL-1α, IL-1ß, KC, and MIG. By immunohistochemical and in situ RNA hybridization analysis we found that the increased expression of proinflammatory cytokines (IL-1α, IL-1ß, TNF-α, IFN-γ) was confined primarily to the area with the pathogen within pyogranulomatous lesions. We also found that cultured splenocytes from chronically infected mice could express IFN-γ, TNF-α, and MIP-1α ex vivo without the need for additional exogenous stimulation. In addition by flow cytometry, we detected significant amounts of intracellular expression of TNF-α and IFN-γ without a protein transport blocker in monocytes/macrophages, NK cells, and neutrophils but not in CD4+ or CD8+ T cells in splenocytes from chronically infected mice. CONCLUSION: Taken together the common features we have identified in chronically infected mice when 10 different human clinical strains of B. pseudomallei were examined could serve as biomarkers when evaluating potential therapeutic agents in mice for the treatment of chronic melioidosis in humans.


Asunto(s)
Burkholderia pseudomallei/fisiología , Interferón gamma/metabolismo , Melioidosis/inmunología , Bazo/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos BALB C
6.
Microb Pathog ; 142: 104050, 2020 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-32050093

RESUMEN

The misuse of infectious disease pathogens as agents of deliberate attack on civilians and military personnel is a serious national security concern, which is exacerbated by the emergence of natural or genetically engineered multidrug resistant strains. In this study, the therapeutic potential of combinations of an antibiotic and a broad-spectrum antimicrobial peptide (AMP) was evaluated against five bacterial biothreats, the etiologic agents of glanders (Burkholderia mallei), melioidosis (Burkholderia pseudomallei), plague (Yersinia pestis), tularemia (Francisella tularensis), and anthrax (Bacillus anthracis). The therapeutics included licensed early generation antibiotics which are now rarely used. Three antibiotics and one 24- amino acid AMP were selected based on MIC assay data. Combinations of the AMP and tigecycline, minocycline, or novobiocin were screened for synergistic activity by checkerboard MIC assay. The combinations each enhanced the susceptibility of several strains. The tetracycline-peptide combinations increased the sensitivities of Y. pestis, F. tularensis, B. anthracis and B. pseudomallei, and the novobiocin-AMP combination augmented the sensitivity of all five. In time-kill assays, down-selected combinations of the peptide and minocycline or tigecycline enhanced killing of B. anthracis, Y. pestis, F. tularensis, and Burkholderia mallei but not B. pseudomallei. The novobiocin-AMP pair significantly reduced viability of all strains except B. mallei, which was very sensitive to the antibiotic alone. The results suggested that antibiotic-AMP combinations are useful tools for combating diverse pathogens. Future studies employing cell culture and animal models will utilize virulent strains of the agents to investigate the in vivo availability, host cytotoxicity, and protective efficacy of these therapeutics.

7.
Emerg Infect Dis ; 25(5): 919-926, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30681072

RESUMEN

For safety, designated Select Agents in tissues must be inactivated and viability tested before the tissue undergoes further processing and analysis. In response to the shipping of samples of "inactivated" Bacillus anthracis that inadvertently contained live spores to nonregulated entities and partners worldwide, the Federal Register now mandates in-house validation of inactivation procedures and standardization of viability testing to detect live organisms in samples containing Select Agents that have undergone an inactivation process. We tested and validated formaldehyde and glutaraldehyde inactivation procedures for animal tissues infected with virulent B. anthracis, Burkholderia pseudomallei, Francisella tularensis, and Yersinia pestis. We confirmed that our fixation procedures for tissues containing these Tier 1 Select Agents resulted in complete inactivation and that our validated viability testing methods do not interfere with detection of live organisms. Institutions may use this work as a guide to develop and conduct their own testing to comply with the policy.


Asunto(s)
Bacterias/efectos de los fármacos , Desinfectantes/farmacología , Formaldehído/farmacología , Glutaral/farmacología , Viabilidad Microbiana/efectos de los fármacos , Animales , Cobayas , Especificidad de Órganos , Esporas Bacterianas/efectos de los fármacos , Factores de Tiempo
8.
Emerg Infect Dis ; 24(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29553922

RESUMEN

The revelation in May 2015 of the shipment of γ irradiation-inactivated wild-type Bacillus anthracis spore preparations containing a small number of live spores raised concern about the safety and security of these materials. The finding also raised doubts about the validity of the protocols and procedures used to prepare them. Such inactivated reference materials were used as positive controls in assays to detect suspected B. anthracis in samples because live agent cannot be shipped for use in field settings, in improvement of currently deployed detection methods or development of new methods, or for quality assurance and training activities. Hence, risk-mitigated B. anthracis strains are needed to fulfill these requirements. We constructed a genetically inactivated or attenuated strain containing relevant molecular assay targets and tested to compare assay performance using this strain to the historical data obtained using irradiation-inactivated virulent spores.


Asunto(s)
Carbunco/microbiología , Bacillus anthracis/fisiología , Bacillus anthracis/efectos de la radiación , Radiación , Esporas Bacterianas/efectos de la radiación , Animales , Bacillus anthracis/virología , Toxinas Bacterianas/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mutagénesis Insercional , Plásmidos/genética , Recombinación Genética , Reproducibilidad de los Resultados , Virulencia , Secuenciación Completa del Genoma
9.
Appl Environ Microbiol ; 84(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29654186

RESUMEN

In 2015, a laboratory of the United States Department of Defense (DoD) inadvertently shipped preparations of gamma-irradiated spores of Bacillus anthracis that contained live spores. In response, a systematic evidence-based method for preparing, concentrating, irradiating, and verifying the inactivation of spore materials was developed. We demonstrate the consistency of spore preparations across multiple biological replicates and show that two different DoD institutions independently obtained comparable dose-inactivation curves for a monodisperse suspension of B. anthracis spores containing 3 × 1010 CFU. Spore preparations from three different institutions and three strain backgrounds yielded similar decimal reduction (D10) values and irradiation doses required to ensure sterility (DSAL) to the point at which the probability of detecting a viable spore is 10-6 Furthermore, spores of a genetically tagged strain of B. anthracis strain Sterne were used to show that high densities of dead spores suppress the recovery of viable spores. Together, we present an integrated method for preparing, irradiating, and verifying the inactivation of spores of B. anthracis for use as standard reagents for testing and evaluating detection and diagnostic devices and techniques.IMPORTANCE The inadvertent shipment by a U.S. Department of Defense (DoD) laboratory of live Bacillus anthracis (anthrax) spores to U.S. and international destinations revealed the need to standardize inactivation methods for materials derived from biological select agents and toxins (BSAT) and for the development of evidence-based methods to prevent the recurrence of such an event. Following a retrospective analysis of the procedures previously employed to generate inactivated B. anthracis spores, a study was commissioned by the DoD to provide data required to support the production of inactivated spores for the biodefense community. The results of this work are presented in this publication, which details the method by which spores can be prepared, irradiated, and tested, such that the chance of finding residual living spores in any given preparation is 1/1,000,000. These irradiated spores are used to test equipment and methods for the detection of agents of biological warfare and bioterrorism.


Asunto(s)
Bacillus anthracis/efectos de la radiación , Rayos gamma , Viabilidad Microbiana/efectos de la radiación , Esporas Bacterianas/efectos de la radiación , Esterilización/métodos , Bacillus anthracis/fisiología , Técnicas Microbiológicas/métodos , Estudios Retrospectivos , Esporas Bacterianas/fisiología
10.
Microb Pathog ; 86: 53-63, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26162294

RESUMEN

Burkholderia pseudomallei is the etiologic agent of melioidosis, which is endemic in Southeast Asia and Northern Australia. We previously found by the intraperitoneal (IP) route that we could discern differences in virulence in mice amongst different strains of B. pseudomallei. We report an early immune response study comparing two strains in our collection which represent the least, B. pseudomallei 1106a, and one of the most, HBPUB10134a, virulent strains in BALB/c mice. B. pseudomallei HBPUB10134a infected mouse spleens contained a 2-3 log higher bacterial burden than mice infected with B. pseudomallei 1106a 3 days post-infection (PI). More and higher amounts of inflammatory cytokines/chemokines were detected in sera and spleen extracts from B. pseudomallei HBPUB10134a than B. pseudomallei 1106a infected mice. The most prominent were IFNγ, IL-1α, IL-1ß, IL-6, IL-10, IP-10, and MIG. After 7 days PI, there was a decrease in bacterial burden in spleens from 1106a infected mice and a decrease in cytokines/chemokines in sera and spleen extracts from both sets of mice. By day 14 PI we saw an increase in monocytes/macrophages, NK cells, and granulocytes in spleens from both sets of mice. No B. pseudomallei HBPUB10134a infected mice survived after this time. In summary, B. pseudomallei HBPUB10134a was more virulent and induced host innate immune responses typical of a more acute-type infection than did B. pseudomallei 1106a which produced a more chronic infection in mice.


Asunto(s)
Burkholderia pseudomallei/inmunología , Melioidosis/inmunología , Melioidosis/patología , Animales , Asia Sudoriental , Australia , Carga Bacteriana , Burkholderia pseudomallei/crecimiento & desarrollo , Enfermedad Crónica , Citocinas/análisis , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Leucocitos Mononucleares/inmunología , Ratones Endogámicos BALB C , Suero/química , Bazo/microbiología , Bazo/patología , Virulencia , Adulto Joven
11.
Front Immunol ; 15: 1397579, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38835755

RESUMEN

Background: Yersinia pestis is the etiological agent of plague, which can manifest as bubonic, septicemic, and/or pneumonic disease. Plague is a severe and rapidly progressing illness that can only be successfully treated with antibiotics initiated early after infection. There are no FDA-approved vaccines for plague, and some vaccine candidates may be less effective against pneumonic plague than bubonic plague. Y. pestis is not known to impact males and females differently in mechanisms of pathogenesis or severity of infection. However, one previous study reported sex-biased vaccine effectiveness after intranasal Y. pestis challenge. As part of developing a safe and effective vaccine, it is essential that potential sex differences are characterized. Methods: In this study we evaluated novel vaccines in male and female BALB/c mice using a heterologous prime-boost approach and monitored survival, bacterial load in organs, and immunological correlates. Our vaccine strategy consisted of two subcutaneous immunizations, followed by challenge with aerosolized virulent nonencapsulated Y. pestis. Mice were immunized with a combination of live Y. pestis pgm- pPst-Δcaf1, live Y. pestis pgm- pPst-Δcaf1/ΔyopD, or recombinant F1-V (rF1-V) combined with adjuvants. Results: The most effective vaccine regimen was initial priming with rF1-V, followed by boost with either of the live attenuated strains. However, this and other strategies were more protective in female mice. Males had higher bacterial burden and differing patterns of cytokine expression and serum antibody titers. Male mice did not demonstrate synergy between vaccination and antibiotic treatment as repeatedly observed in female mice. Conclusions: This study provides new knowledge about heterologous vaccine strategies, sex differences in plague-vaccine efficacy, and the immunological factors that differ between male and female mice.


Asunto(s)
Ratones Endogámicos BALB C , Vacuna contra la Peste , Peste , Yersinia pestis , Animales , Femenino , Peste/prevención & control , Peste/inmunología , Masculino , Yersinia pestis/inmunología , Vacuna contra la Peste/inmunología , Vacuna contra la Peste/administración & dosificación , Ratones , Anticuerpos Antibacterianos/sangre , Caracteres Sexuales , Factores Sexuales , Modelos Animales de Enfermedad , Eficacia de las Vacunas
12.
PLoS One ; 19(7): e0305034, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38954719

RESUMEN

Yersinia pestis, the causative agent of plague and a biological threat agent, presents an urgent need for novel medical countermeasures due to documented cases of naturally acquired antibiotic resistance and potential person-to-person spread during a pneumonic infection. Immunotherapy has been proposed as a way to circumvent current and future antibiotic resistance. Here, we describe the development and characterization of two affinity matured human antibodies (αF1Ig AM2 and αF1Ig AM8) that promote survival of mice after exposure to aerosolized Y. pestis. We share details of the error prone PCR and yeast display technology-based affinity maturation process that we used. The resultant matured antibodies have nanomolar affinity for Y. pestis F1 antigen, are produced in high yield, and are resilient to 37°C stress for up to 6 months. Importantly, in vitro assays using a murine macrophage cell line demonstrated that αF1Ig AM2 and αF1Ig AM8 are opsonic. Even more importantly, in vivo studies using pneumonic plague mouse models showed that 100% of the mice receiving 500 µg of IgGs αF1Ig AM2 and αF1Ig AM8 survived lethal challenge with aerosolized Y. pestis CO92. Combined, these results provide evidence of the quality and robustness of αF1Ig AM2 and αF1Ig AM8 and support their development as potential medical countermeasures against plague.


Asunto(s)
Anticuerpos Antibacterianos , Peste , Yersinia pestis , Animales , Humanos , Ratones , Yersinia pestis/inmunología , Peste/inmunología , Peste/prevención & control , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/inmunología , Femenino , Afinidad de Anticuerpos , Contramedidas Médicas , Antígenos Bacterianos/inmunología , Modelos Animales de Enfermedad
13.
PLoS Pathog ; 7(12): e1002469, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22241984

RESUMEN

Anthrax is a potentially fatal disease resulting from infection with Bacillus anthracis. The outcome of infection is influenced by pathogen-encoded virulence factors such as lethal toxin (LT), as well as by genetic variation within the host. To identify host genes controlling susceptibility to anthrax, a library of congenic mice consisting of strains with homozygous chromosomal segments from the LT-responsive CAST/Ei strain introgressed on a LT-resistant C57BL/6 (B6) background was screened for response to LT. Three congenic strains containing CAST/Ei regions of chromosome 11 were identified that displayed a rapid inflammatory response to LT similar to, but more severe than that driven by a LT-responsive allele of the inflammasome constituent NRLP1B. Importantly, increased response to LT in congenic mice correlated with greater resistance to infection by the Sterne strain of B. anthracis. The genomic region controlling the inflammatory response to LT was mapped to 66.36-74.67 Mb on chromosome 11, a region that encodes the LT-responsive CAST/Ei allele of Nlrp1b. However, known downstream effects of NLRP1B activation, including macrophage pyroptosis, cytokine release, and leukocyte infiltration could not fully explain the response to LT or the resistance to B. anthracis Sterne in congenic mice. Further, the exacerbated response in congenic mice is inherited in a recessive manner while the Nlrp1b-mediated response to LT is dominant. Finally, congenic mice displayed increased responsiveness in a model of sepsis compared with B6 mice. In total, these data suggest that allelic variation of one or more chromosome 11 genes in addition to Nlrp1b controls the severity of host response to multiple inflammatory stimuli and contributes to resistance to B. anthracis Sterne. Expression quantitative trait locus analysis revealed 25 genes within this region as high priority candidates for contributing to the host response to LT.


Asunto(s)
Alelos , Carbunco/genética , Bacillus anthracis , Cromosomas de los Mamíferos/genética , Variación Genética , Inmunidad Innata/genética , Animales , Carbunco/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Cromosomas de los Mamíferos/inmunología , Inflamación/genética , Inflamación/inmunología , Ratones , Sitios de Carácter Cuantitativo/inmunología
14.
Microb Pathog ; 57: 41-51, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23178382

RESUMEN

A transposon insertional mutagenesis spore library of the pathogen Bacillus anthracis was screened to identify mutants altered in germination kinetics. One mutant exhibited an accelerated rate of germination in association with disruption of benK. This gene encodes a putative protein with high homology to membrane transporters that facilitate benzoate transport. We hypothesized that BenK may not be only spore associated, but also have a vegetative cell role. A reporter strain with a translational fusion of benK to green fluorescent protein demonstrated that full-length BenK was present in vegetative cells and that a BenK degradation product was present in spores by detecting the reporter using fluorescence and Western blot analysis. A minimum inhibitory concentration assay indicated that vegetative cells of a benK::Kan mutant were more susceptible to the antimicrobial effects of Na-benzoate. The mutant spores germinated to a greater extent within 1 h than the wild type in an in vitro fluorescence assay. The disruption of benK also resulted in spores that were less readily phagocytosed in a macrophage assay. Despite these altered in vitro phenotypes, no apparent effect of the BenK protein on virulence in the intranasal mouse model or the guinea pig competitive assay was observed. This work shows that, although the BenK protein does not impact fitness or virulence in an infection model, it is involved in other aspects of both the spore and vegetative forms of the organism.


Asunto(s)
Bacillus anthracis/fisiología , Proteínas Bacterianas/genética , Fenotipo , Esporas Bacterianas , Antibacterianos/farmacología , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/patogenicidad , Aptitud Genética , Macrófagos/inmunología , Macrófagos/microbiología , Pruebas de Sensibilidad Microbiana , Mutación , Fagocitosis/genética , Fagocitosis/inmunología , Virulencia
15.
Antibiotics (Basel) ; 12(3)2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36978372

RESUMEN

Burkholderia pseudomallei, the causative agent of the disease melioidosis, has been isolated from the environment in 45 countries. The treatment of melioidosis is complex, requiring lengthy antibiotic regimens, which can result in the relapse of the disease following treatment cessation. It is important that novel therapies to treat infections with B. pseudomallei be assessed in appropriate animal models, and discussions regarding the different protocols used between laboratories are critical. A 'deep dive' was held in October 2020 focusing on the use of the BALB/c mouse model and the inhalational route of infection to evaluate new antibiotic therapies.

16.
Hum Vaccin Immunother ; 19(2): 2216085, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37289480

RESUMEN

Yersinia pestis is a gram-negative bacterium that causes plague in animals and humans. Depending on the route of disease transmission, the bacterium can cause an acute, often fatal disease that has a narrow window for treatment with antibiotics. Additionally, antibiotic resistant strains have been identified, emphasizing the need for novel treatments. Antibody therapy is an appealing option that can direct the immune system to clear bacterial infections. Advances in biotechnology have made both engineering and producing antibodies easier and more affordable. In this study, two screening assays were optimized to evaluate the ability of antibodies to promote phagocytosis of Y. pestis by macrophages and to induce a cytokine signature in vitro that may be predictive of protection in vivo. We evaluated a panel of 21 mouse monoclonal antibodies targeting either the anti-phagocytic capsule F1 protein or the LcrV antigen, which is part of the type 3 secretion system that facilitates translocation of virulence factors into the host cell, using two functional assays. Anti-F1 and anti-LcrV monoclonal antibodies both increased bacterial uptake by macrophages, with greater uptake observed in the presence of antibodies that were protective in the mouse pneumonic plague model. In addition, the protective anti-F1 and anti-LcrV antibodies produced unique cytokine signatures that were also associated with in vivo protection. These antibody-dependent characteristics from in vitro functional assays will be useful in down-selecting efficacious novel antibodies that can be used for treatment of plague.


Asunto(s)
Vacuna contra la Peste , Peste , Yersinia pestis , Ratones , Humanos , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos Bacterianos , Anticuerpos Antibacterianos , Citocinas , Proteínas Citotóxicas Formadoras de Poros
17.
Antibodies (Basel) ; 12(2)2023 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-37218899

RESUMEN

Plague is an ancient disease that continues to be of concern to both the public health and biodefense research communities. Pneumonic plague is caused by hematogenous spread of Yersinia pestis bacteria from a ruptured bubo to the lungs or by directly inhaling aerosolized bacteria. The fatality rate associated with pneumonic plague is significant unless effective antibiotic therapy is initiated soon after an early and accurate diagnosis is made. As with all bacterial pathogens, drug resistance is a primary concern when developing strategies to combat these Yersinia pestis infections in the future. While there has been significant progress in vaccine development, no FDA-approved vaccine strategy exists; thus, other medical countermeasures are needed. Antibody treatment has been shown to be effective in animal models of plague. We produced fully human polyclonal antibodies in transchromosomic bovines vaccinated with the recombinant F1-V plague vaccine. The resulting human antibodies opsonized Y. pestis bacteria in the presence of RAW264.7 cells and afforded significant protection to BALB/c mice after exposure to aerosolized Y. pestis. These data demonstrate the utility of this technology to produce large quantities of non-immunogenic anti-plague human antibodies to prevent or possibly treat pneumonic plague in human.

18.
Hum Vaccin Immunother ; 19(3): 2277083, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37975637

RESUMEN

Francisella tularensis is one of the several biothreat agents for which a licensed vaccine is needed. To ensure vaccine protection is achieved across a range of virulent F. tularensis strains, we assembled and characterized a panel of F. tularensis isolates to be utilized as challenge strains. A promising tularemia vaccine candidate is rLVS ΔcapB/iglABC (rLVS), in which the vector is the LVS strain with a deletion in the capB gene and which additionally expresses a fusion protein comprising immunodominant epitopes of proteins IglA, IglB, and IglC. Fischer rats were immunized subcutaneously 1-3 times at 3-week intervals with rLVS at various doses. The rats were exposed to a high dose of aerosolized Type A strain Schu S4 (FRAN244), a Type B strain (FRAN255), or a tick derived Type A strain (FRAN254) and monitored for survival. All rLVS vaccination regimens including a single dose of 107 CFU rLVS provided 100% protection against both Type A strains. Against the Type B strain, two doses of 107 CFU rLVS provided 100% protection, and a single dose of 107 CFU provided 87.5% protection. In contrast, all unvaccinated rats succumbed to aerosol challenge with all of the F. tularensis strains. A robust Th1-biased antibody response was induced in all vaccinated rats against all F. tularensis strains. These results demonstrate that rLVS ΔcapB/iglABC provides potent protection against inhalational challenge with either Type A or Type B F. tularensis strains and should be considered for further analysis as a future tularemia vaccine.


Asunto(s)
Francisella tularensis , Tularemia , Ratas , Animales , Ratones , Francisella tularensis/genética , Tularemia/prevención & control , Ratas Endogámicas F344 , Vacunas Bacterianas , Vacunas Atenuadas , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad
19.
J Immunol ; 184(1): 17-20, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19949100

RESUMEN

Pathogenesis of Bacillus anthracis is associated with the production of lethal toxin (LT), which activates the murine Nalp1b/Nlrp1b inflammasome and induces caspase-1-dependent pyroptotic death in macrophages and dendritic cells. In this study, we investigated the effect of allelic variation of Nlrp1b on the outcome of LT challenge and infection by B. anthracis spores. Nlrp1b allelic variation did not alter the kinetics or pathology of end-stage disease induced by purified LT, suggesting that, in contrast to previous reports, macrophage lysis does not contribute directly to LT-mediated pathology. However, animals expressing a LT-sensitive allele of Nlrp1b showed an early inflammatory response to LT and increased resistance to infection by B. anthracis. Data presented here support a model whereby LT-mediated activation of Nlrp1b and subsequent lysis of macrophages is not a mechanism used by B. anthracis to promote virulence, but rather a protective host-mediated innate immune response.


Asunto(s)
Carbunco/genética , Carbunco/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Predisposición Genética a la Enfermedad , Animales , Antígenos Bacterianos/toxicidad , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/toxicidad , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
Antibiotics (Basel) ; 11(12)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36551342

RESUMEN

The microbial pathogens Burkholderia pseudomallei and Bacillus anthracis are unrelated bacteria, yet both are the etiologic agents of naturally occurring diseases in animals and humans and are classified as Tier 1 potential biothreat agents. B. pseudomallei is the gram-negative bacterial agent of melioidosis, a major cause of sepsis and mortality globally in endemic tropical and subtropical regions. B. anthracis is the gram-positive spore-forming bacterium that causes anthrax. Infections acquired by inhalation of these pathogens are challenging to detect early while the prognosis is best; and they possess innate multiple antibiotic resistance or are amenable to engineered resistance. Previous studies showed that the early generation, rarely used aminocoumarin novobiocin was very effective in vitro against a range of highly disparate biothreat agents. The objective of the current research was to begin to characterize the therapeutic efficacy of novobiocin in mouse models of anthrax and melioidosis. The antibiotic was highly efficacious against infections by both pathogens, especially B. pseudomallei. Our results supported the concept that specific older generation antimicrobials can be effective countermeasures against infection by bacterial biothreat agents. Finally, novobiocin was shown to be a potential candidate for inclusion in a combined pre-exposure vaccination and post-exposure treatment strategy designed to target bacterial pathogens refractory to a single medical countermeasure.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA