Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Oral Maxillofac Surg ; 23(3): 307-310, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31286292

ABSTRACT

INTRODUCTION: Oncological head and neck operations as well as carotid endarterectomy are common surgical procedures. In some occasions, both procedures have occurred in the past, leading to possible diagnostic and therapeutic challenges when follow-up operations seem indicated. CASE REPORT: We report of a patient presenting with carotid endarterectomy including patch operation 8 years ago and neck dissection due to a squamous cell cancer of the tongue 3 months ago, now showing up with a suspected metastatic tumor of the neck during routine follow-up. Intraoperatively, nearly fatal bleeding occurs due to a partial release of the carotid patch and needs to be managed immediately. DISCUSSION: The primarily pre-operated neck remains challenging for the radiologist in terms of differentiating between chronic lymphadenitis and metastasis. Furthermore, it remains challenging for the oncological surgeon in case these entities are in the near proximity of the previously operated carotid artery. The operative treatment according to the guidelines can lead to major bleeding during the second surgery. During the diagnostic process, metastases and chronic lymphadenitis after alloplastic carotid operations must be differentiated remaining however difficult, due to only scarce data in the literature. CONCLUSION: In the case of previous neck surgery, the decision to operate must be chosen individually regarding the specific conditions and their sometimes vital risks. In case an operation is indicated, the team must be trained to treat life-threatening intraoperative bleeding. In reviewing the literature, we were unable to find published recommendations on how to tackle these challenges.


Subject(s)
Endarterectomy, Carotid , Lymphadenitis , Tongue Neoplasms , Angioplasty , Epithelial Cells , Humans
3.
Front Mol Biosci ; 4: 62, 2017.
Article in English | MEDLINE | ID: mdl-28894739

ABSTRACT

We recently reported that the open-mesh (0.7 µ) polyacrylamide microparticles (MPs) with internally-coupled Cibacron affinity dye demonstrate protective effect in mice challenged into footpads with high doses (200 LD50) of anthrax (Sterne) spores. A single injection of MPs before spore challenge reduces inflammatory response, delays onset of mortality and promotes survival. In this study, we show that the effect of MPs was substantially increased at the lower spore dose (7 LD50). The inflammation of footpads was reduced to the background level, and 60% of animals survived for 16 days while all untreated infected animals died within 6 days with strong inflammation. The effects of MPs were promoted when the MPs were loaded with a combination of neutrophil-attracting chemokines IL-8 and MIP-1α which delayed the onset of mortality in comparison with untreated mice for additional 8 days. The MPs were not inherently cytotoxic against the bacteria or cultured murine Raw 264.7 cells, but stimulated these cells to release G-CSF, MCP-1, MIP-1α, and TNF-α. Consistent with this finding the injection of MPs induced neutrophil influx into footpads, stimulated production of TNF-α associated with migration of pERK1/2-positive cells with the Langerhans phenotype from epidermis to regional lymph nodes. Our data support the mechanism of protection in which the immune defense induced by MPs along with the exogenous chemokines counterbalances the suppressive effect caused by anthrax infection.

4.
PLoS One ; 11(9): e0163163, 2016.
Article in English | MEDLINE | ID: mdl-27632537

ABSTRACT

In this study the hydrogel microparticles (MPs) were used to enhance migration of neutrophils in order to improve outcome of anthrax infection in a mouse model. Two MP formulations were tested. In the first one the polyacrylamide gel MPs were chemically coupled with Cibacron Blue (CB) affinity bait. In the second one the bait molecules within the MPs were additionally loaded with neutrophil-attracting chemokines (CKs), human CXCL8 and mouse CCL3. A non-covalent interaction of the bait with the CKs provided their gradual release after administration of the MPs to the host. Mice were challenged into footpads with Bacillus anthracis Sterne spores and given a dose of MPs a few hours before and/or after the spores. Pre-treatment with a single dose of CK-releasing MPs without any additional intervention was able to induce influx of neutrophils to the site of spore inoculation and regional lymph nodes correlating with reduced bacterial burden and decreased inflammatory response in footpads. On average, in two independent experiments, up to 53% of mice survived over 13 days. All control spore-challenged but MP-untreated mice died. The CB-coupled particles were also found to improve survival likely due to the capacity to stimulate release of endogenous CKs, but were less potent at decreasing the inflammatory host response than the CK-releasing MPs. The CK post-treatment did not improve survival compared to the untreated mice which died within 4 to 6 days with a strong inflammation of footpads, indicating quick dissemination of spores though the lymphatics after challenge. This is the first report on the enhanced innate host resistance to anthrax in response to CKs delivered and/or endogenously induced by the MPs.


Subject(s)
Bacillus anthracis/physiology , Chemokine CCL3/administration & dosage , Interleukin-8/administration & dosage , Spores, Bacterial , Animals , Drug Carriers , Female , Mice , Mice, Inbred DBA
5.
Front Microbiol ; 7: 69, 2016.
Article in English | MEDLINE | ID: mdl-26870026

ABSTRACT

Bacillus cereus, a food poisoning bacterium closely related to Bacillus anthracis, secretes a multitude of virulence factors including enterotoxins, hemolysins, and phospholipases. However, the majority of the in vitro experiments evaluating the cytotoxic potential of B. cereus were carried out in the conditions of aeration, and the impact of the oxygen limitation in conditions encountered by the microbe in natural environment such as gastrointestinal tract remains poorly understood. This research reports comparative analysis of ATCC strains 11778 (BC1) and 14579 (BC2) in aerobic and microaerobic (static) cultures with regard to their toxicity for human lung epithelial cells. We showed that BC1 increased its toxicity upon oxygen limitation while BC2 was highly cytotoxic in both growth conditions. The combined effect of the pore-forming, cholesterol-dependent hemolysin, cereolysin O (CLO), and metabolic product(s) such as succinate produced in microaerobic conditions provided substantial contribution to the toxicity of BC1 but not BC2 which relied mainly on other toxins. This mechanism is shared between CB1 and B. anthracis. It involves the permeabilization of the cell membrane which facilitates transport of toxic bacterial metabolites into the cell. The toxicity of BC1 was potentiated in the presence of bovine serum albumin which appeared to serve as reservoir for bacteria-derived nitric oxide participating in the downstream production of reactive oxidizing species with the properties of peroxynitrite. In agreement with this the BC1 cultures demonstrated the increased oxidation of the indicator dye Amplex Red catalyzed by peroxidase as well as the increased toxicity in the presence of externally added ascorbic acid.

6.
Front Microbiol ; 6: 921, 2015.
Article in English | MEDLINE | ID: mdl-26388860

ABSTRACT

Nitric oxide (NO) is a key physiological regulator in eukaryotic and prokaryotic organisms. It can cause a variety of biological effects by reacting with its targets or/and indirectly inducing oxidative stress. NO can also be produced by bacteria including the pathogenic Bacillus anthracis; however, its role in the infectious process only begins to emerge. NO incapacitates macrophages by S-nitrosylating the intracellular proteins and protects B. anthracis from oxidative stress. It is also implicated in the formation of toxic peroxynitrite. In this study we further assessed the effects of B. anthracis NO produced by the NO synthase (bNOS) on bacterial metabolism and host cells in experiments with the bNOS knockout Sterne strain. The mutation abrogated accumulation of nitrite and nitrate as tracer products of NO in the culture medium and markedly attenuated growth in both aerobic and microaerobic conditions. The regulatory role of NO was also suggested by the abnormally high rate of nitrate denitrification by the mutant in the presence of oxygen. Anaerobic regulation mediated by NO was reflected in reduced fermentation of glucose by the mutant correlating with the reduced toxicity of bacteria toward host cells in culture. The toxic effect of NO required permeabilization of the target cells as well as the activity of fermentation-derived metabolite in the conditions of reduced pH. The host cells demonstrated increased phosphorylation of major survivor protein kinase AKT correlating with reduced toxicity of the mutant in comparison with Sterne. Our global proteomic analysis of lymph from the lymph nodes of infected mice harboring bacteria revealed numerous changes in the pattern and levels of proteins associated with the activity of bNOS influencing key cell physiological processes relevant to energy metabolism, growth, signal transduction, stress response, septic shock, and homeostasis. This is the first in vivo observation of the bacterial NO effect on the lymphatic system.

7.
PLoS One ; 10(6): e0129860, 2015.
Article in English | MEDLINE | ID: mdl-26091359

ABSTRACT

Anthrax is a frequently fatal infection of many animal species and men. The causative agent Bacillus anthracis propagates through the lymphatic system of the infected host; however, the specific interactions of the host and microbe within the lymphatics are incompletely understood. We report the first description of the phosphoprotein signaling in the lymph nodes of DBA/2 mice using a novel technique combining the reverse-phase microarray with the laser capture microdissection. Mice were challenged into foot pads with spores of toxinogenic, unencapsulated Sterne strain. The spores quickly migrated to the regional popliteal lymph nodes and spread to the bloodstream as early as 3 h post challenge. All mice died before 72 h post challenge from the systemic disease accompanied by a widespread LN tissue damage by bacteria, including the hemorrhagic necrotizing lymphadenitis, infiltration of CD11b+ and CD3+ cells, and massive proliferation of bacteria in lymph nodes. A macrophage scavenger receptor CD68/macrosialin was upregulated and found in association with vegetative bacteria likely as a marker of their prior interaction with macrophages. The major signaling findings among the 65 tested proteins included the reduced MAPK signaling, upregulation of STAT transcriptional factors, and altered abundance of a number of pro- and anti-apoptotic proteins with signaling properties opposing each other. Downregulation of ERK1/2 was associated with the response of CD11b+ macrophages/dendritic cells, while upregulation of the pro-apoptotic Puma indicated a targeting of CD3+ T-cells. A robust upregulation of the anti-apoptotic survivin was unexpected because generally it is not observed in adult tissues. Taken together with the activation of STATs it may reflect a new pathogenic mechanism aimed to delay the onset of apoptosis. Our data emphasize a notion that the net biological outcome of disease is determined by a cumulative impact of factors representing the microbial insult and the protective capacity of the host.


Subject(s)
Anthrax/metabolism , Anthrax/microbiology , Bacillus anthracis/physiology , Lymph Nodes/metabolism , Protein Array Analysis , Animals , Anthrax/mortality , Biomarkers , Disease Models, Animal , Immunohistochemistry , Lymph Nodes/microbiology , Lymph Nodes/pathology , Mice , Phosphoproteins/metabolism , Proteome , Proteomics/methods , Signal Transduction , Spores, Bacterial
8.
Nanomaterials (Basel) ; 5(1): 298-320, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25878893

ABSTRACT

Chemokines (CKs) secreted by the host cells into surrounding tissue establish concentration gradients directing the migration of leukocytes. We propose an in vivo CK gradient remodeling approach based on sustained release of CKs by the crosslinked poly(N-isopropylacrylamide) hydrogel open meshwork nano-particles (NPs) containing internal crosslinked dye affinity baits for a reversible CK binding and release. The sustained release is based on a new principle of affinity off-rate tuning. The NPs with Cibacron Blue F3G-A and Reactive Blue-4 baits demonstrated a low-micromolar affinity binding to IL-8, MIP-2, and MCP-1 with a half-life of several hours at 37°C. The capacity of NPs loaded with IL-8 and MIP-1α to increase neutrophil recruitment to lymph nodes (LNs) was tested in mice after footpad injection. Fluorescently-labeled NPs used as tracers indicated the delivery into the sub-capsular compartment of draining LNs. The animals administered the CK-loaded NPs demonstrated a widening of the sub-capsular space and a strong lymph node influx of leukocytes, while mice injected with control NPs without CKs or bolus doses of soluble CKs alone showed only a marginal neutrophil response. This technology provides a new means therapeutically direct or restore immune cell traffic, and can also be employed for simultaneous therapy delivery.

9.
PLoS One ; 9(10): e110873, 2014.
Article in English | MEDLINE | ID: mdl-25329596

ABSTRACT

This study aimed to characterize a soluble proteome of popliteal lymph nodes during lymphadenitis induced by intradermal injection of Bacillus anthracis Sterne spores in mice using tandem LC-MS/MS and reverse-phase protein microarray with antibodies specific to epitopes of phosphorylated proteins. More than 380 proteins were detected in the normal intra-nodal lymph, while the infectious process resulted in the profound changes in the protein abundances and appearance of 297 unique proteins. These proteins belong to an array of processes reflecting response to wounding, inflammation and perturbations of hemostasis, innate immune response, coagulation and fibrinolysis, regulation of body fluid levels and vascular disturbance among others. Comparison of lymph and serum revealed 83 common proteins. Also, using 71 antibodies specific to total and phosphorylated forms of proteins we carried initial characterization of circulating lymph phosphoproteome which brought additional information regarding signaling pathways operating in the lymphatics. The results demonstrate that the proteome of intra-nodal lymph serves as a sensitive sentinel of the processes occurring within the lymph nodes during infection. The acute innate response of the lymph nodes to anthrax is accompanied by cellular damage and inflammation with a large number of up- and down-regulated proteins many of which are distinct from those detected in serum. MS data are available via ProteomeXchange with identifier PXD001342.


Subject(s)
Anthrax/metabolism , Bacillus anthracis , Gene Expression Regulation , Lymph Nodes/metabolism , Proteome/biosynthesis , Skin Diseases, Bacterial/metabolism , Animals , Anthrax/pathology , Lymph Nodes/pathology , Mice , Skin Diseases, Bacterial/pathology
10.
Article in English | MEDLINE | ID: mdl-23730627

ABSTRACT

Bacillus anthracis is a dangerous pathogen of humans and many animal species. Its virulence has been mainly attributed to the production of Lethal and Edema toxins as well as the antiphagocytic capsule. Recent data indicate that the nitric oxide (NO) synthase (baNOS) plays an important pathogenic role at the early stage of disease by protecting bacteria from the host reactive species and S-nytrosylating the mitochondrial proteins in macrophages. In this study we for the first time present evidence that bacteria-derived NO participates in the generation of highly reactive oxidizing species which could be abolished by the NOS inhibitor L - NAME, free thiols, and superoxide dismutase but not catalase. The formation of toxicants is likely a result of the simultaneous formation of NO and superoxide leading to a labile peroxynitrite and its stable decomposition product, nitrogen dioxide. The toxicity of bacteria could be potentiated in the presence of bovine serum albumin. This effect is consistent with the property of serum albumin to serves as a trap of a volatile NO accelerating its reactions. Our data suggest that during infection in the hypoxic environment of pre-mortal host the accumulated NO is expected to have a broad toxic impact on host cell functions.


Subject(s)
Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Nitric Oxide/metabolism , Serum Albumin, Bovine/metabolism , Animals , Cattle , Humans , Hypoxia , Nitric Oxide/toxicity , Superoxides/metabolism , Superoxides/toxicity
11.
Microbes Infect ; 15(1): 37-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23108317

ABSTRACT

Bacillus anthracis is the causative agent of anthrax and is acquired by three routes of infection: inhalational, gastrointestinal and cutaneous. Gastrointestinal (GI) anthrax is rare, but can rapidly result in severe, systemic disease that is fatal in 25%-60% of cases. Disease mechanisms of GI anthrax remain unclear due to limited numbers of clinical cases and the lack of experimental animal models. Here, we developed an in vivo murine model of GI anthrax where spore survival was maximized through the neutralization of stomach acid followed by an intragastric administration of a thiabendazole paste spore formulation. Infected mice showed a dose-dependent mortality rate and pathological features closely mimicking human GI anthrax. Since Peyer's patches in the murine intestine are the primary sites of B. anthracis growth, we developed a human M (microfold)-like-cell model using a Caco-2/Raji B-cell co-culturing system to study invasive mechanisms of GI anthrax across the intestinal epithelium. Translocation of B. anthracis spores was higher in M-like cells than Caco-2 monolayers, suggesting that M-like cells may serve as an initial entry site for spores. Here, we developed an in vivo murine model of GI anthrax and an in vitro M-like cell model that could be used to further our knowledge of GI anthrax pathogenesis.


Subject(s)
Anthrax/microbiology , Bacillus anthracis/physiology , Disease Models, Animal , Gastrointestinal Diseases/microbiology , Animals , Anthrax/pathology , Bacillus anthracis/pathogenicity , Bacterial Adhesion/physiology , Bacterial Load , Bacterial Translocation , Caco-2 Cells , Female , Gastrointestinal Diseases/pathology , Host-Pathogen Interactions , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Kaplan-Meier Estimate , Mice , Mice, Inbred DBA , Spores, Bacterial
12.
Biochem Biophys Res Commun ; 430(1): 125-30, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23178574

ABSTRACT

Bacillus anthracis, a causative agent of anthrax, is able to germinate and survive within macrophages. A recent study suggested that B. anthracis-derived nitric oxide (bNO) is a key aspect of bacterial defense that protects bacterial DNA from oxidative burst in the macrophages. However, the virulent effect of bNO in host cells has not been investigated. Here, we report that bNO contributes macrophage killing by S-nitrosylation of bioenergetic-relating proteins within mitochondria. Toxigenic Sterne induces expression of the bnos gene and produces bNO during early stage of infection. Nitroso-proteomic analysis coupled with a biotin-switch technique demonstrated that toxigenic infection induces protein S-nitrosylation in B. anthracis-susceptible RAW264.7. For each target enzyme tested (complex I, complex III and complex IV), infection by B. anthracis Sterne caused enzyme inhibition. Nω-nitro-L-arginine methyl ester, a NO synthase inhibitor, reduced S-nitrosylation and partially restored cell viability evaluated by intracellular ATP levels in macrophages. Our data suggest that bNO leads to energy depletion driven by impaired mitochondrial bioenergetic machinery that ultimately contributes to macrophage death. This novel mechanism of anthrax pathogenesis may offer specific approach to the development of therapeutics.


Subject(s)
Anthrax/immunology , Apoptosis/immunology , Bacillus anthracis/pathogenicity , Macrophages/microbiology , Mitochondria/microbiology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Animals , Anthrax/enzymology , Anthrax/microbiology , Bacillus anthracis/metabolism , Cell Line , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Gene Expression , Macrophages/enzymology , Mice , Mitochondria/enzymology , Nitric Oxide Synthase Type I/genetics
13.
Cell Microbiol ; 14(8): 1219-30, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22452315

ABSTRACT

To achieve widespread dissemination in the host, Bacillus anthracis cells regulate their attachment to host endothelium during infection. Previous studies identified BslA (Bacillus anthracis S-layer Protein A), a virulence factor of B. anthracis, as necessary and sufficient for adhesion of vegetative cells to human endothelial cells. While some factors have been identified, bacteria-specific contributions to BslA mediated adhesion remain unclear. Using the attenuated vaccine Sterne 7702 strain of B. anthracis, we tested the hypothesis that InhA (immune inhibitor A), a B. anthracis protease, regulates BslA levels affecting the bacteria's ability to bind to endothelium. To test this, a combination of inhA mutant and complementation analysis in adhesion and invasion assays, Western blot and InhA inhibitor assays were employed. Results show InhA downregulates BslA activity reducing B. anthracis adhesion and invasion in human brain endothelial cells. BslA protein levels in ΔinhA bacteria were significantly higher than wild-type and complemented strains showing InhA levels and BslA expression are inversely related. BslA was sensitive to purified InhA degradation in a concentration- and time-dependent manner. Taken together these data support the role of InhA regulation of BslA-mediated vegetative cell adhesion and invasion.


Subject(s)
Adhesins, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Adhesion , Endothelial Cells/microbiology , Metalloproteases/metabolism , Animals , Bacillus anthracis/genetics , Bacillus anthracis/physiology , Bicarbonates/chemistry , Brain/blood supply , Culture Media/chemistry , Gene Expression , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Host-Pathogen Interactions , Humans , Macrophages/microbiology , Metalloproteases/genetics , Mice , Microbial Viability , Microvessels/cytology , Phenanthrolines/pharmacology , Protease Inhibitors/pharmacology , Proteolysis , Virulence Factors/metabolism
14.
Microb Pathog ; 52(1): 1-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22001909

ABSTRACT

UNLABELLED: Vascular dysfunction and thrombosis have been described in association with anthrax infection in humans and animals but the mechanisms of these dysfunctions, as well as the components involved in thrombi formation are poorly understood. Immunofluorescent microscopy was used to define the composition of thrombi in the liver of mice challenged with the Bacillus anthracis Sterne spores. Lethal infection with the toxigenic Sterne strain, in contrast to the non-lethal, non-toxigenic delta-Sterne strain, demonstrated time-dependent increase in the number of vegetative bacteria inside the liver sinusoids and central vein. Massive appearance of thrombi typically occluding the lumen of the vessels coincided with the sudden death of infected animals. Bacterial chains in the thrombi were stained positive for syndecan-1 (SDC-1), fibronectin, and were surrounded by fibrin polymers, GPIIb-positive platelets, von Willebrand Factor (vWF), CD45-positive leukocytes, and massive amount of shed SDC-1. Experiments with human umbilical vein endothelial cells (HUVECs) demonstrated the active role of the host response to the secreted pathogenic factors of bacteria during the onset of the pro-thrombotic condition. The bacterial culture supernatants, as well as the isolated proteins (the pore-forming toxin anthrolysin O and phospholipase C) induced release of vWF, while anthrolysin O, sphingomyelinase and edema toxin induced release of thrombin from HUVECs and polymerization of fibrin in the presence of human plasma. CONCLUSION: Our findings suggest that activation of endothelium in response to infection can contribute to the formation of occlusive thrombi consisting of aggregated bacteria, vWF, shed SDC-1, fibrin, activated platelets, fibronectin and leukocytes.


Subject(s)
Anthrax/metabolism , Bacillus anthracis/physiology , Blood Platelets/metabolism , Fibrin/metabolism , Liver/microbiology , Syndecan-1/metabolism , Thrombosis/metabolism , von Willebrand Factor/metabolism , Animals , Anthrax/immunology , Anthrax/microbiology , Bacillus anthracis/genetics , Blood Coagulation , Fibronectins/metabolism , Humans , Leukocytes/immunology , Liver/pathology , Mice , Mice, Inbred DBA , Thrombosis/immunology , Thrombosis/microbiology
15.
PLoS One ; 6(5): e20354, 2011.
Article in English | MEDLINE | ID: mdl-21655261

ABSTRACT

Rift Valley fever (RVF) is a zoonotic disease caused by Rift Valley fever virus (RVFV). RVFV is a category A pathogen that belongs to the genus Phlebovirus, family Bunyaviridae. Understanding early host events to an infectious exposure to RVFV will be of significant use in the development of effective therapeutics that not only control pathogen multiplication, but also contribute to cell survival. In this study, we have carried out infections of human cells with a vaccine strain (MP12) and virulent strain (ZH501) of RVFV and determined host responses to viral infection. We demonstrate that the cellular antioxidant enzyme superoxide dismutase 1 (SOD1) displays altered abundances at early time points following exposure to the virus. We show that the enzyme is down regulated in cases of both a virulent (ZH501) and a vaccine strain (MP12) exposure. Our data demonstrates that the down regulation of SOD1 is likely to be due to post transcriptional processes and may be related to up regulation of TNFα following infection. We also provide evidence for extensive oxidative stress in the MP12 infected cells. Concomitantly, there is an increase in the activation of the p38 MAPK stress response, which our earlier published study demonstrated to be an essential cell survival strategy. Our data suggests that the viral anti-apoptotic protein NSm may play a role in the regulation of the cellular p38 MAPK response. Alterations in the host protein SOD1 following RVFV infection appears to be an early event that occurs in multiple cell types. Activation of the cellular stress response p38 MAPK pathway can be observed in all cell types tested. Our data implies that maintaining oxidative homeostasis in the infected cells may play an important role in improving survival of infected cells.


Subject(s)
Rift Valley fever virus/physiology , Superoxide Dismutase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Line , Chlorocebus aethiops , Flow Cytometry , Hep G2 Cells , Humans , Oxidative Stress/genetics , Oxidative Stress/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Tumor Necrosis Factor-alpha/metabolism , Vero Cells , p38 Mitogen-Activated Protein Kinases/genetics
16.
World J Biol Chem ; 2(5): 98-104, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21666812

ABSTRACT

AIM: To establish whether activation of adenosine type-3 receptors (A3Rs) and inhibition of interleukin-1ß-induced inflammation is beneficial in combination with antibiotic therapy to increase survival of mice challenged with anthrax spores. METHODS: DBA/2 mice were challenged with Bacillus anthracis spores of the toxigenic Sterne strain 43F2. Survival of animals was monitored for 15 d. Ciprofloxacin treatment (50 mg/kg, once daily, intraperitoneally) was initiated at day +1 simultaneously with the administration of inhibitors, and continued for 10 d. Two doses (2.5 mg/kg and 12.5 mg/kg) of acetyl-tyrosyl-valyl-alanyl-aspartyl-chloromethylketone (YVAD) and three doses (0.05, 0.15 and 0.3 mg/kg) of 1-[2-Chloro-6-[[(3-iodophenyl) methyl]amino]-9H-purin-9-yl]-1-deoxy-N-methyl-ß-D- ribofuranuronamide (Cl-IB-MECA) were tested. Animals received YVAD on days 1-4, and Cl-IB-MECA on days 1-10 once daily, subcutaneously. Human lung epithelial cells in culture were challenged with spores or edema toxin and the effects of IB-MECA on phosphorylation of AKT and generation of cAMP were tested. RESULTS: We showed that the outcome of antibiotic treatment in a murine anthrax model could be substantially improved by co-administration of the caspase-1/4 inhibitor YVAD and the A3R agonist Cl-IB-MECA. Combination treatment with these substances and ciprofloxacin resulted in up to 90% synergistic protection. All untreated mice died, and antibiotic alone protected only 30% of animals. We conclude that both substances target the aberrant host signaling that underpins anthrax mortality. CONCLUSION: Our findings suggest new possibilities for combination therapy of anthrax with antibiotics, A3R agonists and caspase-1 inhibitors.

17.
PLoS One ; 6(3): e18119, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21464960

ABSTRACT

The causative agent of anthrax, Bacillus anthracis, is capable of circumventing the humoral and innate immune defense of the host and modulating the blood chemistry in circulation to initiate a productive infection. It has been shown that the pathogen employs a number of strategies against immune cells using secreted pathogenic factors such as toxins. However, interference of B. anthracis with the innate immune system through specific interaction of the spore surface with host proteins such as the complement system has heretofore attracted little attention. In order to assess the mechanisms by which B. anthracis evades the defense system, we employed a proteomic analysis to identify human serum proteins interacting with B. anthracis spores, and found that plasminogen (PLG) is a major surface-bound protein. PLG efficiently bound to spores in a lysine- and exosporium-dependent manner. We identified α-enolase and elongation factor tu as PLG receptors. PLG-bound spores were capable of exhibiting anti-opsonic properties by cleaving C3b molecules in vitro and in rabbit bronchoalveolar lavage fluid, resulting in a decrease in macrophage phagocytosis. Our findings represent a step forward in understanding the mechanisms involved in the evasion of innate immunity by B. anthracis through recruitment of PLG resulting in the enhancement of anti-complement and anti-opsonization properties of the pathogen.


Subject(s)
Bacillus anthracis/immunology , Complement C3b/immunology , Fibrinolysin/metabolism , Immunity, Innate/immunology , Plasminogen/metabolism , Animals , Bacillus anthracis/drug effects , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans , Immunity, Innate/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice , Opsonin Proteins/immunology , Phagocytosis/drug effects , Phagocytosis/immunology , Protein Binding/drug effects , Rabbits , Recombinant Proteins/metabolism , Spores, Bacterial/drug effects , Spores, Bacterial/metabolism , Urokinase-Type Plasminogen Activator/pharmacology
18.
FEMS Immunol Med Microbiol ; 62(2): 173-81, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21395696

ABSTRACT

The fibrinolytic system is often the target for pathogenic bacteria, resulting in increased fibrinolysis, bacterial dissemination, and inflammation. The purpose of this study was to explore whether proteases NprB and InhA secreted by Bacillus anthracis could activate the host's fibrinolytic system. NprB efficiently activated human pro-urokinase plasminogen activator (pro-uPA), a key protein in the fibrinolytic cascade. Conversely, InhA had little effect on pro-uPA. Plasminogen activator inhibitors (PAI)-1, 2 and the uPA receptor were also targets for NprB in vitro. InhA efficiently degraded the thrombin-activatable fibrinolysis inhibitor (TAFI) in vitro. Mice infected with B. anthracis showed a significant decrease in blood TAFI levels. In another mouse experiment, animals infected with isogenic inhA deletion mutants restored TAFI levels, while the levels in the parent strain decreased. We propose that NprB and InhA may contribute to the activation of the fibrinolytic system in anthrax infection.


Subject(s)
Bacillus anthracis/enzymology , Fibrinolysis , Peptide Hydrolases/metabolism , Animals , Anthrax/microbiology , Anthrax/pathology , Carboxypeptidase B2/blood , Carboxypeptidase B2/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred DBA , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Recombinant Proteins/metabolism , Rodent Diseases/microbiology , Rodent Diseases/pathology , Urokinase-Type Plasminogen Activator/metabolism
19.
PLoS One ; 6(3): e17921, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21437287

ABSTRACT

Hemorrhagic meningitis is a fatal complication of anthrax, but its pathogenesis remains poorly understood. The present study examined the role of B. anthracis-secreted metalloprotease InhA on monolayer integrity and permeability of human brain microvasculature endothelial cells (HBMECs) which constitute the blood-brain barrier (BBB). Treatment of HBMECs with purified InhA resulted in a time-dependent decrease in trans-endothelial electrical resistance (TEER) accompanied by zonula occluden-1 (ZO-1) degradation. An InhA-expressing B. subtilis exhibited increased permeability of HBMECs, which did not occur with the isogenic inhA deletion mutant (ΔinhA) of B. anthracis, compared with the corresponding wild-type strain. Mice intravenously administered with purified InhA or nanoparticles-conjugated to InhA demonstrated a time-dependent Evans Blue dye extravasation, leptomeningeal thickening, leukocyte infiltration, and brain parenchymal distribution of InhA indicating BBB leakage and cerebral hemorrhage. Mice challenged with vegetative bacteria of the ΔinhA strain of B. anthracis exhibited a significant decrease in leptomeningeal thickening compared to the wildtype strain. Cumulatively, these findings indicate that InhA contributes to BBB disruption associated with anthrax meningitis through proteolytic attack on the endothelial tight junctional protein zonula occluden (ZO)-1.


Subject(s)
Bacillus anthracis/enzymology , Bacterial Proteins/metabolism , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/pathology , Cerebral Hemorrhage/microbiology , Cerebral Hemorrhage/pathology , Metalloproteases/metabolism , Animals , Anthrax/microbiology , Anthrax/pathology , Blotting, Western , Cytoplasm/metabolism , Electric Impedance , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelial Cells/pathology , Female , Humans , Membrane Proteins/metabolism , Mice , Mutant Proteins/metabolism , Nanospheres , Permeability , Phosphoproteins/metabolism , Zonula Occludens-1 Protein
20.
FEMS Immunol Med Microbiol ; 61(1): 15-27, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20946354

ABSTRACT

Bacillus anthracis generates virulence factors such as lethal and edema toxins, capsule, and hemolytic proteins under conditions of reduced oxygenation. Here, we report on the acute cytotoxicity of culture supernatants (Sups) of six nonencapsulated B. anthracis strains grown till the stationary phase under static microaerobic conditions. Human small airway epithelial, umbilical vein endothelial, Caco-2, and Hep-G2 cells were found to be susceptible. Sups displayed a reduction of pH to 5.3-5.5, indicating the onset of acid anaerobic fermentation; however, low pH itself was not a major factor of toxicity. The pore-forming hemolysin, anthrolysin O (ALO), contributed to the toxicity in a concentration-dependent manner. Its effect was found to be synergistic with a metabolic product of B. anthracis, succinic acid. Cells exposed to Sups demonstrated cytoplasmic membrane blebbing, increased permeability, loss of ATP, mitochondrial membrane potential collapse, and arrest of cell respiration. The toxicity was reduced by inhibition of ALO by cholesterol, decomposition of reactive oxygen species, and inhibition of mitochondrial succinate dehydrogenase. Cell death appears to be caused by an acute primary membrane permeabilization by ALO, followed by a burst of reactive radicals from the mitochondria fuelled by the succinate, which is generated by bacteria in the hypoxic environment. This mechanism of metabolic toxicity is relevant to the late-stage conditions of hypoxia and acidosis found in anthrax patients and might operate at anatomical locations of the host deprived from oxygen supply.


Subject(s)
Bacillus anthracis/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Epithelial Cells/drug effects , Fermentation , Membrane Glycoproteins/toxicity , Aerobiosis , Bacillus anthracis/genetics , Bacillus anthracis/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Caco-2 Cells , Cell Survival/drug effects , Cholesterol/metabolism , Culture Media, Conditioned/chemistry , Epithelial Cells/metabolism , Gene Deletion , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Lung/cytology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/toxicity , Recombinant Proteins/toxicity , Succinic Acid/toxicity , Virulence Factors/metabolism , Virulence Factors/pharmacology , Virulence Factors/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...