Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Immunohorizons ; 8(3): 269-280, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38517345

RESUMEN

Bacillus anthracis peptidoglycan (PGN) is a major component of the bacterial cell wall and a key pathogen-associated molecular pattern contributing to anthrax pathology, including organ dysfunction and coagulopathy. Increases in apoptotic leukocytes are a late-stage feature of anthrax and sepsis, suggesting there is a defect in apoptotic clearance. In this study, we tested the hypothesis that B. anthracis PGN inhibits the capacity of human monocyte-derived macrophages (MΦ) to efferocytose apoptotic cells. Exposure of CD163+CD206+ MΦ to PGN for 24 h impaired efferocytosis in a manner dependent on human serum opsonins but independent of complement component C3. PGN treatment reduced cell surface expression of the proefferocytic signaling receptors MERTK, TYRO3, AXL, integrin αVß5, CD36, and TIM-3, whereas TIM-1, αVß3, CD300b, CD300f, STABILIN-1, and STABILIN-2 were unaffected. ADAM17 is a major membrane-bound protease implicated in mediating efferocytotic receptor cleavage. We found multiple ADAM17-mediated substrates increased in PGN-treated supernatant, suggesting involvement of membrane-bound proteases. ADAM17 inhibitors TAPI-0 and Marimastat prevented TNF release, indicating effective protease inhibition, and modestly increased cell-surface levels of MerTK and TIM-3 but only partially restored efferocytic capacity by PGN-treated MΦ. We conclude that human serum factors are required for optimal recognition of PGN by human MΦ and that B. anthracis PGN inhibits efferocytosis in part by reducing cell surface expression of MERTK and TIM-3.


Asunto(s)
Carbunco , Bacillus anthracis , Humanos , Tirosina Quinasa c-Mer/metabolismo , Peptidoglicano/farmacología , Peptidoglicano/metabolismo , Carbunco/metabolismo , Carbunco/patología , Eferocitosis , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Macrófagos/metabolismo , Pared Celular/metabolismo , Pared Celular/patología
2.
Microb Pathog ; 183: 106305, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37586464

RESUMEN

Inhalation anthrax, the deadliest form of the disease, requires inhaled B. anthracis spores to escape from the alveolar space and travel to the mediastinal lymph nodes, from where the vegetative form of the pathogen disseminates, resulting in a rapidly fatal outcome. The role of epithelia in alveolar escape is unclear, but previous work suggests these epithelial cells are involved in this process. Using confocal microscopy, we found that B. anthracis spores are internalized more rapidly by A549 type II alveolar epithelial cells compared to hAELVi type I alveolar epithelial cells. Internalization of spores by alveolar epithelial cells requires cytoskeletal rearrangement evidenced by significant inhibition by cytochalasin D, an actin inhibitor. Chemical inhibitors of macropinocytosis significantly downregulated B. anthracis spore internalization in human alveolar cells, while inhibitors of other endocytosis pathways had minimal effects. Additional studies using a macropinosome marker and electron microscopy confirmed the role of macropinocytosis in spore uptake. By colocalization of B. anthracis spores with four endocytic Rab proteins, we demonstrated that Rab31 played a role in B. anthracis spore macropinocytosis. Finally, we confirmed that Rab31 is involved in B. anthracis spore internalization by enhanced spore uptake in Rab31-overexpressing A549 cells. This is the first report that shows B. anthracis spore internalization by macropinocytosis in human epithelial cells. Several Rab GTPases are involved in the process.


Asunto(s)
Carbunco , Bacillus anthracis , Humanos , Esporas Bacterianas/metabolismo , Células Epiteliales , Pulmón , Carbunco/metabolismo
3.
J Biol Chem ; 298(1): 101467, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34871548

RESUMEN

Bacillus anthracis lethal toxin and edema toxin are binary toxins that consist of a common cell-binding moiety, protective antigen (PA), and the enzymatic moieties, lethal factor (LF) and edema factor (EF). PA binds to either of two receptors, capillary morphogenesis protein-2 (CMG-2) or tumor endothelial marker-8 (TEM-8), which triggers the binding and cytoplasmic translocation of LF and EF. However, the distribution of functional TEM-8 and CMG-2 receptors during anthrax toxin intoxication in animals has not been fully elucidated. Herein, we describe an assay to image anthrax toxin intoxication in animals, and we use it to visualize TEM-8- and CMG-2-dependent intoxication in mice. Specifically, we generated a chimeric protein consisting of the N-terminal domain of LF fused to a nuclear localization signal-tagged Cre recombinase (LFn-NLS-Cre). When PA and LFn-NLS-Cre were coadministered to transgenic mice expressing a red fluorescent protein in the absence of Cre and a green fluorescent protein in the presence of Cre, intoxication could be visualized at single-cell resolution by confocal microscopy or flow cytometry. Using this assay, we found that: (a) CMG-2 is critical for intoxication in the liver and heart, (b) TEM-8 is required for intoxication in the kidney and spleen, (c) CMG-2 and TEM-8 are redundant for intoxication of some organs, (d) combined loss of CMG-2 and TEM-8 completely abolishes intoxication, and (e) CMG-2 is the dominant receptor on leukocytes. The novel assay will be useful for basic and clinical/translational studies of Bacillus anthracis infection and for clinical development of reengineered toxin variants for cancer treatment.


Asunto(s)
Carbunco , Antígenos Bacterianos , Bacillus anthracis , Toxinas Bacterianas , Animales , Carbunco/diagnóstico por imagen , Carbunco/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/toxicidad , Bacillus anthracis/metabolismo , Toxinas Bacterianas/toxicidad , Citoplasma/metabolismo , Ratones , Ratones Transgénicos
4.
PLoS Biol ; 18(12): e3001052, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370274

RESUMEN

Bacillus anthracis, a spore-forming gram-positive bacterium, causes anthrax. The external surface of the exosporium is coated with glycosylated proteins. The sugar additions are capped with the unique monosaccharide anthrose. The West African Group (WAG) B. anthracis have mutations rendering them anthrose deficient. Through genome sequencing, we identified 2 different large chromosomal deletions within the anthrose biosynthetic operon of B. anthracis strains from Chile and Poland. In silico analysis identified an anthrose-deficient strain in the anthrax outbreak among European heroin users. Anthrose-deficient strains are no longer restricted to West Africa so the role of anthrose in physiology and pathogenesis was investigated in B. anthracis Sterne. Loss of anthrose delayed spore germination and enhanced sporulation. Spores without anthrose were phagocytized at higher rates than spores with anthrose, indicating that anthrose may serve an antiphagocytic function on the spore surface. The anthrose mutant had half the LD50 and decreased time to death (TTD) of wild type and complement B. anthracis Sterne in the A/J mouse model. Following infection, anthrose mutant bacteria were more abundant in the spleen, indicating enhanced dissemination of Sterne anthrose mutant. At low sample sizes in the A/J mouse model, the mortality of ΔantC-infected mice challenged by intranasal or subcutaneous routes was 20% greater than wild type. Competitive index (CI) studies indicated that spores without anthrose disseminated to organs more extensively than a complemented mutant. Death process modeling using mouse mortality dynamics suggested that larger sample sizes would lead to significantly higher deaths in anthrose-negative infected animals. The model was tested by infecting Galleria mellonella with spores and confirmed the anthrose mutant was significantly more lethal. Vaccination studies in the A/J mouse model showed that the human vaccine protected against high-dose challenges of the nonencapsulated Sterne-based anthrose mutant. This work begins to identify the physiologic and pathogenic consequences of convergent anthrose mutations in B. anthracis.


Asunto(s)
Amino Azúcares/genética , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Desoxiglucosa/análogos & derivados , Amino Azúcares/inmunología , Amino Azúcares/metabolismo , Animales , Carbunco/genética , Carbunco/inmunología , Carbunco/metabolismo , Bacillus anthracis/patogenicidad , Evolución Biológica , Desoxiglucosa/genética , Desoxiglucosa/inmunología , Desoxiglucosa/metabolismo , Modelos Animales de Enfermedad , Brotes de Enfermedades , Evolución Molecular , Femenino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos A , Mariposas Nocturnas/microbiología , Oligosacáridos/genética , Oligosacáridos/inmunología , Oligosacáridos/metabolismo , Esporas Bacterianas/genética , Esporas Bacterianas/inmunología , Esporas Bacterianas/metabolismo
5.
Nanotechnology ; 31(24): 245706, 2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-32126532

RESUMEN

The development of novel 2D materials, due to the promising applications they have enabled through their unique properties, has attracted increasingly more research interest. In this regard, novel dual-emitting coordination polymer nanosheets were developed by doping Eu3+ and Tb3+ ions into the nanostructures of the [Ba(DPA)2(H2O)2] n (DPA = dipicolinic acid) coordination polymer (BCP). Single crystal x-ray crystallography revealed that BCP is a 1D coordination polymer and its three-dimensional supramolecular architecture is constructed with a relatively strong hydrogen bonding in the ac crystallographic plane and weak non-covalent interactions along the b axis. Using energetic ultrasound irradiations, synthesis of nanoscale BCP along with the unzipping of the weak interactions between the ac layers was accomplished. The resulting BCP nanosheets was used as the host lattice and was doped with Eu3+ and Tb3+ ions. Remarkably, the sensing ability of both Eu3+ and Tb3+ doped coordination polymer (Ln@BCP) nanosheets towards temperature and the DPA anthrax biomarker were investigate. The high relative sensitivity value of 2.42% K-1 and their reusability, makes Ln@BCP nanosheets an ideal candidate for the nanothermometry. They also exhibited high selective detection characteristics towards the DPA anthrax biomarker with a 0.03 nM detection limit. Therefore, Ln@BCP nanosheets can also be considered as an efficient multi-responsive optical sensor.


Asunto(s)
Bario/química , Técnicas Biosensibles/instrumentación , Complejos de Coordinación/química , Ácidos Picolínicos/análisis , Carbunco/diagnóstico , Carbunco/metabolismo , Cristalografía por Rayos X , Europio/química , Peróxido de Hidrógeno/química , Límite de Detección , Estructura Molecular , Nanoestructuras , Temperatura , Terbio/química
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 229: 117999, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-31935655

RESUMEN

Bacillus anthracis spores have a unique biomarker of calcium dipicolinate (CaDPA). In this work, we reported a composite nanostructure for the optical sensing of DPA, with Eu (III)-doped metal-organic framework (MOF) as supporting lattice, a rhodamine-derived dye as sensing probe, respectively. By means of XRD, IR, TGA and photophysical analysis, this composite structure was carefully discussed. It was found that rhodamine absorption and emission were enhanced by DPA, while Eu emission was quenched by DPA. As a consequence, two sensing skills were observed from this composite structure, which are colorimetric sensing based on absorption spectra and ratiometric fluorescent sensing based on emission spectra. Linear sensing response was observed for both sensing channels with a warning signal at DPA concentration higher than 140 µM. Good selectivity was confirmed with a low LOD value of 0.52 µM. The sensing mechanism was revealed as the combination of emission turn-on effect triggered by DPA-released protons and emission turn-off effect originated from electron-transfer from EuBTC to DPA. This composite structure showed its advantage of naked eye detection and two sensing skills with linear response.


Asunto(s)
Carbunco/diagnóstico , Colorimetría/métodos , Europio/química , Colorantes Fluorescentes/química , Estructuras Metalorgánicas/química , Ácidos Picolínicos/análisis , Rodaminas/química , Carbunco/metabolismo , Carbunco/microbiología , Bacillus anthracis/aislamiento & purificación , Técnicas Biosensibles , Humanos , Espectrometría de Fluorescencia
7.
J Infect Dis ; 221(4): 660-667, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31574153

RESUMEN

BACKGROUND: Lethal and edema toxins are critical virulence factors of Bacillus anthracis. Few data are available on their presence in the early stage of intranasal infection. METHODS: To investigate the diffusion of edema factor (EF) and lethal factor (LF), we use sensitive quantitative methods to measure their enzymatic activities in mice intranasally challenged with a wild-type B anthracis strain or with an isogenic mutant deficient for the protective antigen. RESULTS: One hour after mouse challenge, although only 7% of mice presented bacteremia, LF and EF were detected in the blood of 100% and 42% of mice, respectively. Protective antigen facilitated the diffusion of LF and EF into the blood compartment. Toxins played a significant role in the systemic dissemination of B anthracis in the blood, spleen, and liver. A mouse model of intoxination further confirmed that LT and ET could diffuse rapidly in the circulation, independently of bacteria. CONCLUSIONS: In this inhalational model, toxins have disseminated rapidly in the blood, playing a significant and novel role in the early systemic diffusion of bacteria, demonstrating that they may represent a very early target for the diagnosis and the treatment of anthrax.


Asunto(s)
Carbunco/metabolismo , Antígenos Bacterianos/sangre , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/sangre , Absorción Nasal , Factores de Virulencia/sangre , Animales , Animales no Consanguíneos , Carbunco/microbiología , Bacillus anthracis/enzimología , Bacteriemia , Biomarcadores/sangre , Modelos Animales de Enfermedad , Activación Enzimática , Pruebas de Enzimas , Femenino , Ratones , Virulencia
8.
Artículo en Inglés | MEDLINE | ID: mdl-31681636

RESUMEN

Understanding bacterial virulence provides insight into the molecular basis behind infection and could identify new drug targets. However, assessing potential virulence determinants relies on testing in an animal model. The mouse is a well-validated model but it is constrained by the ethical and logistical challenges of using vertebrate animals. Recently the larva of the greater wax moth Galleria mellonella has been explored as a possible infection model for a number of pathogens. In this study, we developed G. mellonella as an infection model for Bacillus anthracis Sterne. We first validated two different infection assays, a survival assay and a competition assay, using mutants containing disruptions in known B. anthracis virulence genes. We next tested the utility of G. mellonella to assess the virulence of transposon mutants with unknown mutations that had increased susceptibility to hydrogen peroxide in in vitro assays. One of these transposon mutants also displayed significantly decreased virulence in G. mellonella. Further investigation revealed that this mutant had a disruption in the petrobactin biosynthesis operon (asbABCDEF), which has been previously implicated in both virulence and defense against oxidative stress. We conclude that G. mellonella can detect attenuated virulence of B. anthracis Sterne in a manner consistent with that of mammalian infection models. Therefore, G. mellonella could serve as a useful alternative to vertebrate testing, especially for early assessments of potential virulence genes when use of a mammalian model may not be ethical or practical.


Asunto(s)
Carbunco/microbiología , Bacillus anthracis , Animales , Carbunco/metabolismo , Bacillus anthracis/genética , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Genes Bacterianos , Peróxido de Hidrógeno/metabolismo , Ratones , Viabilidad Microbiana/genética , Mutación , Operón , Virulencia/genética , Factores de Virulencia/genética
10.
Nat Microbiol ; 4(11): 1805-1814, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31308522

RESUMEN

Anthrax is an ancient and deadly disease caused by the spore-forming bacterial pathogen Bacillus anthracis. At present, anthrax mostly affects wildlife and livestock, although it remains a concern for human public health-primarily for people who handle contaminated animal products and as a bioterrorism threat due to the high resilience of spores, a high fatality rate of cases and the lack of a civilian vaccination programme1,2. The cell surface of B. anthracis is covered by a protective paracrystalline monolayer-known as surface layer or S-layer-that is composed of the S-layer proteins Sap or EA1. Here, we generate nanobodies to inhibit the self-assembly of Sap, determine the structure of the Sap S-layer assembly domain (SapAD) and show that the disintegration of the S-layer attenuates the growth of B. anthracis and the pathology of anthrax in vivo. SapAD comprises six ß-sandwich domains that fold and support the formation of S-layers independently of calcium. Sap-inhibitory nanobodies prevented the assembly of Sap and depolymerized existing Sap S-layers in vitro. In vivo, nanobody-mediated disruption of the Sap S-layer resulted in severe morphological defects and attenuated bacterial growth. Subcutaneous delivery of Sap inhibitory nanobodies cleared B. anthracis infection and prevented lethality in a mouse model of anthrax disease. These findings highlight disruption of S-layer integrity as a mechanism that has therapeutic potential in S-layer-carrying pathogens.


Asunto(s)
Carbunco/tratamiento farmacológico , Bacillus anthracis/efectos de los fármacos , Glicoproteínas de Membrana/química , Anticuerpos de Dominio Único/administración & dosificación , Animales , Carbunco/metabolismo , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidad , Modelos Animales de Enfermedad , Inyecciones Subcutáneas , Glicoproteínas de Membrana/metabolismo , Ratones , Viabilidad Microbiana/efectos de los fármacos , Modelos Moleculares , Conformación Proteica en Lámina beta/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Anticuerpos de Dominio Único/farmacología
11.
Biochemistry ; 58(27): 2996-3004, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31243996

RESUMEN

Anthrax, a lethal, weaponizable disease caused by Bacillus anthracis, acts through exotoxins that are primary mediators of systemic toxicity and also targets for neutralization by passive immunotherapy. The ease of engineering B. anthracis strains resistant to established therapy and the historic use of the microbe in bioterrorism present a compelling test case for platforms that permit the rapid and modular development of neutralizing agents. In vitro antigen-binding fragment (Fab) selection offers the advantages of speed, sequence level molecular control, and engineering flexibility compared to traditional monoclonal antibody pipelines. By screening an unbiased, chemically synthetic phage Fab library and characterizing hits in cell-based assays, we identified two high-affinity neutralizing Fabs, A4 and B7, against anthrax edema factor (EF), a key mediator of anthrax pathogenesis. Engineered homodimers of these Fabs exhibited potency comparable to that of the best reported neutralizing monoclonal antibody against EF at preventing EF-induced cyclic AMP production. Using internalization assays in COS cells, B7 was found to block steps prior to EF internalization. This work demonstrates the efficacy of synthetic alternatives to traditional antibody therapeutics against anthrax while also demonstrating a broadly generalizable, rapid, and modular screening pipeline for neutralizing antibody generation.


Asunto(s)
Carbunco/tratamiento farmacológico , Anticuerpos Neutralizantes/farmacología , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/antagonistas & inhibidores , Fragmentos Fab de Inmunoglobulinas/farmacología , Secuencia de Aminoácidos , Animales , Carbunco/metabolismo , Carbunco/microbiología , Anticuerpos Neutralizantes/química , Antígenos Bacterianos/metabolismo , Bacillus anthracis/fisiología , Toxinas Bacterianas/metabolismo , Células CHO , Células COS , Línea Celular , Chlorocebus aethiops , Cricetulus , AMP Cíclico/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Ratones , Multimerización de Proteína
12.
Mol Microbiol ; 112(2): 515-531, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31063630

RESUMEN

A challenge common to all bacterial pathogens is to acquire nutrients from hostile host environments. Iron is an important cofactor required for essential cellular processes such as DNA repair, energy production and redox balance. Within a mammalian host, most iron is sequestered within heme, which in turn is predominantly bound by hemoglobin. While little is understood about the mechanisms by which bacterial hemophores attain heme from host-hemoglobin, even less is known about intracellular heme processing. Bacillus anthracis, the causative agent of anthrax, displays a remarkable ability to grow in mammalian hosts. Hypothesizing this pathogen harbors robust ways to catabolize heme, we characterize two new intracellular heme-binding proteins that are distinct from the previously described IsdG heme monooxygenase. The first of these, HmoA, binds and degrades heme, is necessary for heme detoxification and facilitates growth on heme iron sources. The second protein, HmoB, binds and degrades heme too, but is not necessary for heme utilization or virulence. The loss of both HmoA and IsdG renders B. anthracis incapable of causing anthrax disease. The additional loss of HmoB in this background increases clearance of bacilli in lungs, which is consistent with this protein being important for survival in alveolar macrophages.


Asunto(s)
Carbunco/microbiología , Bacillus anthracis/metabolismo , Hemo/metabolismo , Carbunco/metabolismo , Bacillus anthracis/enzimología , Bacillus anthracis/genética , Bacillus anthracis/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Unión Proteica
13.
Biochem Biophys Res Commun ; 513(3): 651-656, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30982579

RESUMEN

Endospores are important for maintenance of the B. anthracis lifecycle and necessary for its effective spread between hosts. Our experiments with B. anthracis showed that disruption of SpoIIID results in a spore formation defect, as determined by heat resistance assays and microscopic assessment. We further found complete engulfment by the ΔspoIIID mutant strain by membrane morphology staining but no synthesis of the clarity coat and exosporium by transmission electron microscopy. Reduced transcription and expression of small acid-soluble spore protein(sasP-2) and the spore development associated genes (σK, gerE and cotE) in the mother cell were found in the ΔspoIIID strain, suggesting that the spore formation defect in B. anthracis A16R is related to decreased transcription and expression of these genes. Extracellular protease and virulence enhancement in the ΔspoIIID strain may be related to the elevation of metalloproteinases (TasA and Camelysin) levels. Our findings pave the way for further research on the regulation network of sporulation, survival and virulence in these two morphological forms of B. anthracis.


Asunto(s)
Bacillus anthracis/crecimiento & desarrollo , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Esporas Bacterianas/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Animales , Carbunco/metabolismo , Carbunco/microbiología , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidad , Proteínas Bacterianas/genética , Proteínas de Unión al ADN/genética , Femenino , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Humanos , Ratones , Proteolisis , Esporas Bacterianas/genética , Esporas Bacterianas/metabolismo , Factores de Transcripción/genética
14.
ACS Infect Dis ; 4(12): 1746-1754, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30354048

RESUMEN

Inflammasomes activate caspase-1 in response to molecular signals from pathogens and other dangerous stimuli as a part of the innate immune response. A previous study discovered a small-molecule, 4-fluoro- N'-[1-(2-pyridinyl)ethylidene]benzohydrazide, which we named DN1, that reduces the cytotoxicity of anthrax lethal toxin (LT). We determined that DN1 protected cells irrespectively of LT concentration and reduced the pathogenicity of an additional bacterial exotoxin and several viruses. Using the LT cytotoxicity pathway, we show that DN1 does not prevent LT internalization and catalytic activity or caspase-1 activation. Moreover, DN1 does not affect the proteolytic activity of host cathepsin B, which facilitates the cytoplasmic entry of toxins. PubChem Bioactivities lists two G protein-coupled receptors (GPCR), type-1 angiotensin II receptor and apelin receptor, as targets of DN1. The inhibition of phosphatidylinositol 3-kinase, phospholipase C, and protein kinase B, which are downstream of GPCR signaling, synergized with DN1 in protecting cells from LT. We hypothesize that DN1-mediated antagonism of GPCRs modulates signal transduction pathways to induce a cellular state that reduces LT-induced pyroptosis downstream of caspase-1 activation. DN1 also reduced the susceptibility of Drosophila melanogaster to toxin-associated bacterial infections. Future experiments will aim to further characterize how DN1 modulates signal transduction pathways to inhibit pyroptotic cell death in LT-sensitive macrophages. DN1 represents a novel chemical probe to investigate host cellular mechanisms that mediate cell death in response to pathogenic agents.


Asunto(s)
Carbunco/fisiopatología , Antibacterianos/farmacología , Antígenos Bacterianos/toxicidad , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/toxicidad , Muerte Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Carbunco/tratamiento farmacológico , Carbunco/metabolismo , Carbunco/microbiología , Antibacterianos/química , Bacillus anthracis/genética , Bacillus anthracis/crecimiento & desarrollo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/antagonistas & inhibidores , Caspasa 1/genética , Caspasa 1/metabolismo , Catepsina B/genética , Catepsina B/metabolismo , Drosophila melanogaster , Femenino , Interacciones Huésped-Patógeno , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Bibliotecas de Moléculas Pequeñas/química
15.
Blood ; 132(8): 849-860, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-29921614

RESUMEN

Anthrax infections exhibit progressive coagulopathies that may contribute to the sepsis pathophysiology observed in fulminant disease. The hemostatic imbalance is recapitulated in primate models of late-stage disease but is uncommon in toxemic models, suggesting contribution of other bacterial pathogen-associated molecular patterns (PAMPs). Peptidoglycan (PGN) is a bacterial PAMP that engages cellular components at the cross talk between innate immunity and hemostasis. We hypothesized that PGN is critical for anthrax-induced coagulopathies and investigated the activation of blood coagulation in response to a sterile PGN infusion in primates. The PGN challenge, like the vegetative bacteria, induced a sepsis-like pathophysiology characterized by systemic inflammation, disseminated intravascular coagulation (DIC), organ dysfunction, and impaired survival. Importantly, the hemostatic impairment occurred early and in parallel with the inflammatory response, suggesting direct engagement of coagulation pathways. PGN infusion in baboons promoted early activation of contact factors evidenced by elevated protease-serpin complexes. Despite binding to contact factors, PGN did not directly activate either factor XII (FXII) or prekallikrein. PGN supported contact coagulation by enhancing enzymatic function of active FXII (FXIIa) and depressing its inhibition by antithrombin. In parallel, PGN induced de novo monocyte tissue factor expression in vitro and in vivo, promoting extrinsic clotting reactions at later stages. Activation of platelets further amplified the procoagulant state during PGN challenge, leading to DIC and subsequent ischemic damage of peripheral tissues. These data indicate that PGN may be a major cause for the pathophysiologic progression of Bacillus anthracis sepsis and is the primary PAMP behind the pathogen-induced coagulopathy in late-stage anthrax.


Asunto(s)
Carbunco/metabolismo , Bacillus anthracis , Coagulación Sanguínea/efectos de los fármacos , Coagulación Intravascular Diseminada/sangre , Monocitos/metabolismo , Animales , Carbunco/patología , Coagulación Intravascular Diseminada/inducido químicamente , Coagulación Intravascular Diseminada/patología , Factor XIIa/metabolismo , Femenino , Masculino , Monocitos/patología , Papio , Papio anubis , Precalicreína/metabolismo
16.
Anal Chem ; 90(11): 7004-7011, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29701058

RESUMEN

Lanthanide-based luminescent sensors have been widely used for the detection of the anthrax biomarker dipicolinic acid (DPA). However, mainly based on DPA sensitization to the lanthanide core, most of them failed to realize robust detection of DPA in bacterial spores. We proposed a new strategy for reliable detection of DPA by perturbing a tandem energy transfer in heterobinuclear lanthanide coordination polymer nanoparticles simply constructed by two kinds of lanthanide ions, Tb3+ and Eu3+, and guanosine 5'-monophosphate. This smart luminescent probe was demonstrated to exhibit highly sensitive and selective visual luminescence color change upon exposure to DPA, enabling accurate detection of DPA in complex biosystems such as bacterial spores. DPA release from bacterial spores on physiological germination was also successfully monitored in real time by confocal imaging. This probe is thus expected to be a powerful tool for efficient detection of bacterial spores in responding to anthrax threats.


Asunto(s)
Carbunco/metabolismo , Elementos de la Serie de los Lantanoides/química , Nanopartículas/química , Ácidos Picolínicos/análisis , Polímeros/química , Esporas Bacterianas/química , Bacillus anthracis/química , Biomarcadores/análisis , Biomarcadores/metabolismo , Transferencia de Energía , Luminiscencia , Mediciones Luminiscentes , Ácidos Picolínicos/metabolismo , Factores de Tiempo
17.
Microb Pathog ; 121: 9-21, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29704667

RESUMEN

The lung is the entry site for Bacillus anthracis in inhalation anthrax, the most deadly form of the disease. Spores must escape through the alveolar epithelial cell (AEC) barrier and migrate to regional lymph nodes, germinate and enter the circulatory system to cause disease. Several mechanisms to explain alveolar escape have been postulated, and all these tacitly involve the AEC barrier. In this study, we incorporate our primary human type I AEC model, microarray and gene enrichment analysis, qRT-PCR, multiplex ELISA, and neutrophil and monocyte chemotaxis assays to study the response of AEC to B. anthracis, (Sterne) spores at 4 and 24 h post-exposure. Spore exposure altered gene expression in AEC after 4 and 24 h and differentially expressed genes (±1.3 fold, p ≤ 0.05) included CCL4/MIP-1ß (4 h), CXCL8/IL-8 (4 and 24 h) and CXCL5/ENA-78 (24 h). Gene enrichment analysis revealed that pathways involving cytokine or chemokine activity, receptor binding, and innate immune responses to infection were prominent. Microarray results were confirmed by qRT-PCR and multiplex ELISA assays. Chemotaxis assays demonstrated that spores induced the release of biologically active neutrophil and monocyte chemokines, and that CXCL8/IL-8 was the major neutrophil chemokine. The small or sub-chemotactic doses of CXCL5/ENA-78, CXCL2/GROß and CCL20/MIP-3α may contribute to chemotaxis by priming effects. These data provide the first whole transcriptomic description of the human type I AEC initial response to B. anthracis spore exposure. Taken together, our findings contribute to an increased understanding of the role of AEC in the pathogenesis of inhalational anthrax.


Asunto(s)
Células Epiteliales Alveolares/microbiología , Bacillus anthracis/patogenicidad , Quimiocinas/metabolismo , Perfilación de la Expresión Génica , Esporas Bacterianas/patogenicidad , Carbunco/genética , Carbunco/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Quimiocinas/genética , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Monocitos/metabolismo , Monocitos/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/metabolismo , Regulación hacia Arriba
18.
Artículo en Inglés | MEDLINE | ID: mdl-29404280

RESUMEN

Bacillus anthracis, the causative agent of anthrax, has been a focus of study in host-pathogen dynamics since the nineteenth century. While the interaction between anthrax and host macrophages has been extensively modeled, comparatively little is known about the effect of anthrax on the immune function of neutrophils, a key frontline effector of innate immune defense. Here we showed that depletion of neutrophils significantly enhanced mortality in a systemic model of anthrax infection in mice. Ex vivo, we found that freshly isolated human neutrophils can rapidly kill anthrax, with specific inhibitor studies showing that phagocytosis and reactive oxygen species (ROS) generation contribute to this efficient bacterial clearance. Anthrax toxins, comprising lethal toxin (LT) and edema toxin (ET), are known to have major roles in B. anthracis macrophage resistance and systemic toxicity. Employing isogenic wild-type and mutant toxin-deficient B. anthracis strains, we show that despite previous studies that reported inhibition of neutrophil function by purified LT or ET, endogenous production of these toxins by live vegetative B. anthracis failed to alter key neutrophil functions. The lack of alteration in neutrophil function is accompanied by rapid killing of B. anthracis by neutrophils, regardless of the bacteria's expression of anthrax toxins. Lastly, our study demonstrates for the first time that anthrax induced neutrophil extracellular trap (NET) formation.


Asunto(s)
Carbunco/inmunología , Carbunco/microbiología , Bacillus anthracis/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Neutrófilos/inmunología , Neutrófilos/microbiología , Animales , Carbunco/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Trampas Extracelulares/microbiología , Femenino , Regulación Bacteriana de la Expresión Génica , Ratones , Neutrófilos/metabolismo , Fagocitosis/inmunología
19.
J Infect Dis ; 216(11): 1471-1475, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-28968672

RESUMEN

Gastrointestinal (GI) anthrax is the most prevalent form of naturally acquired Bacillus anthracis infection, which is associated with exposure to vegetative bacteria in infected meat (carnivores) or to fermented rumen contents (herbivores). We assessed whether key host and pathogen factors modulate infectivity and progression of infection using a mouse model of GI infection. Gastric acid neutralization increases infectivity, but 30%-40% of mice succumb to infection without neutralization. Mice either fed or fasted before exposure showed similar infectivity rates. Finally, the pathogen's anthrax lethal factor is required to establish lethal infection, whereas its edema factor modulates progression and dissemination of infection.


Asunto(s)
Carbunco/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/metabolismo , Progresión de la Enfermedad , Ácido Gástrico , Enfermedades Gastrointestinales/microbiología , Factores de Virulencia , Animales , Carbunco/microbiología , Carbunco/patología , Bacillus anthracis/fisiología , Modelos Animales de Enfermedad , Femenino , Enfermedades Gastrointestinales/patología , Corazón/microbiología , Concentración de Iones de Hidrógeno , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Pulmón/microbiología , Pulmón/patología , Ratones , Virulencia
20.
Int J Mol Sci ; 18(1)2017 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-28054967

RESUMEN

Anthrax toxin receptor-mediated drug development for blocking anthrax toxin action has received much attention in recent decades. In this study, we produced a secreted anthrax decoy fusion protein comprised of a portion of the human capillary morphogenesis gene-2 (CMG2) protein fused via a linker to the fragment crystallizable (Fc) domain of human immunoglobulin G1 in Nicotiana benthamiana plants using a transient expression system. Using the Cauliflower Mosaic Virus (CaMV) 35S promoter and co-expression with the p19 gene silencing suppressor, we were able to achieve a high level of recombinant CMG2-Fc-Apo (rCMG2-Fc-Apo) protein accumulation. Production kinetics were observed up to eight days post-infiltration, and maximum production of 826 mg/kg fresh leaf weight was observed on day six. Protein A affinity chromatography purification of the rCMG2-Fc-Apo protein from whole leaf extract and apoplast wash fluid showed the homodimeric form under non-reducing gel electrophoresis and mass spectrometry analysis confirmed the molecular integrity of the secreted protein. The N-glycosylation pattern of purified rCMG2-Fc-Apo protein was analysed; the major portion of N-glycans consists of complex type structures in both protein samples. The most abundant (>50%) N-glycan structure was GlcNAc2(Xyl)Man3(Fuc)GlcNAc2 in rCMG2-Fc-Apo recovered from whole leaf extract and apoplast wash fluid. High mannose N-glycan structures were not detected in the apoplast wash fluid preparation, which confirmed the protein secretion. Altogether, these findings demonstrate that high-level production of rCMG2-Fc-Apo can be achieved by transient production in Nicotiana benthamiana plants with apoplast targeting.


Asunto(s)
Inmunoglobulina G/genética , Plantas Modificadas Genéticamente/genética , Receptores de Péptidos/genética , Secuencia de Aminoácidos , Carbunco/metabolismo , Carbunco/microbiología , Bacillus anthracis/metabolismo , Biotecnología , Caulimovirus/genética , Clonación Molecular , Descubrimiento de Drogas , Glicosilación , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Regiones Promotoras Genéticas , Receptores de Péptidos/química , Receptores de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...