51
|
Mwakapeje ER, Høgset S, Softic A, Mghamba J, Nonga HE, Mdegela RH, Skjerve E. Risk factors for human cutaneous anthrax outbreaks in the hotspot districts of Northern Tanzania: an unmatched case-control study. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180479. [PMID: 30839712 PMCID: PMC6170534 DOI: 10.1098/rsos.180479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/16/2018] [Indexed: 06/09/2023]
Abstract
Bacillus anthracis is an aerobic, Gram-positive and spore-forming bacterium, which causes anthrax in herbivores. Humans get infected after coming into contact with infected animals' products. An unmatched case-control study was conducted to identify the importance of demographic, biological and/or behavioural factors associated with human cutaneous anthrax outbreaks in the hotspot areas of Northern Tanzania. A semi-structured questionnaire was administered to both cases and controls. The age range of participants was 1-80 years with a median age of 32 years. In the younger group (1-20 years), the odds of being infected were 25 times higher in the exposed group compared to the unexposed group (OR= 25, 95% CI = 1.5-410). By contrast, the odds of exposure in the old group (≥20 years) were three times lower in the exposed group compared to the unexposed group (OR = 3.2, 95% CI = 1.28-8.00). Demographic characteristics, sleeping on animal's skins, contacting with infected carcasses through skinning and butchering, and not having formal education were linked to exposure for anthrax infection. Hence, a One Health approach is inevitable for the prevention and control of anthrax outbreaks in the hotspot areas of Northern Tanzania.
Collapse
Affiliation(s)
- Elibariki R. Mwakapeje
- Epidemiology and Diseases Control Section, Ministry of Health, Community Development, Gender, Elderly and Children, PO Box 9083, Dar es Salaam, Tanzania
- Department of Veterinary Medicine and Public Health, Sokoine University of Agriculture, PO Box 3021, Chuo Kikuu Morogoro, Tanzania
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, PO Box 8146 Dep., 0033 Oslo, Norway
| | - Sol Høgset
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, PO Box 8146 Dep., 0033 Oslo, Norway
| | - Adis Softic
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, PO Box 8146 Dep., 0033 Oslo, Norway
| | - Janneth Mghamba
- Epidemiology and Diseases Control Section, Ministry of Health, Community Development, Gender, Elderly and Children, PO Box 9083, Dar es Salaam, Tanzania
| | - Hezron E. Nonga
- Department of Veterinary Medicine and Public Health, Sokoine University of Agriculture, PO Box 3021, Chuo Kikuu Morogoro, Tanzania
| | - Robinson H. Mdegela
- Department of Veterinary Medicine and Public Health, Sokoine University of Agriculture, PO Box 3021, Chuo Kikuu Morogoro, Tanzania
| | - Eystein Skjerve
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, PO Box 8146 Dep., 0033 Oslo, Norway
| |
Collapse
|
52
|
Popescu NI, Silasi R, Keshari RS, Girton A, Burgett T, Zeerleder SS, Gailani D, Gruber A, Lupu F, Coggeshall KM. Peptidoglycan induces disseminated intravascular coagulation in baboons through activation of both coagulation pathways. Blood 2018; 132:849-860. [PMID: 29921614 PMCID: PMC6107880 DOI: 10.1182/blood-2017-10-813618] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
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.
Collapse
Affiliation(s)
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Ravi S Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Alanson Girton
- Department of Arthritis and Clinical Immunology and
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Sacha S Zeerleder
- Department of Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN; and
| | - Andras Gruber
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - K Mark Coggeshall
- Department of Arthritis and Clinical Immunology and
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
53
|
Complete Genome Sequences of Three Bacillus anthracis Bacteriophages. GENOME ANNOUNCEMENTS 2018; 6:6/1/e01164-17. [PMID: 29301897 PMCID: PMC5754477 DOI: 10.1128/genomea.01164-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The new highly effective Bacillus anthracis phages Negev_SA, Carmel_SA, and Tavor_SA were isolated from soil samples, and their complete genomes were sequenced and analyzed. The isolated phages have potential use in future phage therapy treatment against anthrax.
Collapse
|
54
|
Anthrax immune globulin improves hemodynamics and survival during B. anthracis toxin-induced shock in canines receiving titrated fluid and vasopressor support. Intensive Care Med Exp 2017; 5:48. [PMID: 29058092 PMCID: PMC5651533 DOI: 10.1186/s40635-017-0159-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/22/2017] [Indexed: 11/10/2022] Open
Abstract
Background Although anthrax immune globulin (AIG) improved survival in antibiotic-treated Bacillus anthracis-challenged animal models, whether it adds to the benefit of conventional hemodynamic support for B. anthracis toxin-associated shock is unknown. Methods We therefore tested AIG in sedated, mechanically ventilated canines challenged with 24-h B. anthracis lethal and edema toxin infusions and supported for 96 h with a previously demonstrated protective regimen of titrated normal saline and norepinephrine. Results Compared to controls, proportional survival (%) was increased with AIG treatment started 4 h before (33 vs. 100%, n = 6 each) or 2 h (17 vs. 86%, n = 6 and 7 respectively) or 5 h (0 vs. 67%, n = 3 each) after the start of toxin (p ≤ 0.05) and overall [3 survivors of 15 controls (20%) vs. 14 of 16 AIG animals (88%); p = 0.006]. Averaged across treatment times, AIG increased blood pressure at 48 h and decreased norepinephrine requirements at 72 h (p ≤ 0.02), increased left ventricular ejection fraction at 48 and 72 h (p ≤ 0.02), and increased urine output and decreased net fluid balance at 72 and 96 h (p ≤ 0.04). AIG also reduced acidosis and renal and hepatic injury markers between 24 and 96 h. Conclusions These findings further support AIG’s potential benefit for patients with B. anthracis infection and developing toxin-associated shock. Electronic supplementary material The online version of this article (10.1186/s40635-017-0159-9) contains supplementary material, which is available to authorized users.
Collapse
|
55
|
Purulent Appearing Material in an Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration of Mediastinal Lymph Node: A Diagnostic Challenge. Case Rep Pulmonol 2017; 2017:3851849. [PMID: 29201483 PMCID: PMC5671702 DOI: 10.1155/2017/3851849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/18/2017] [Indexed: 11/23/2022] Open
Abstract
Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) has increasingly been performed for the diagnosis and staging of thoracic malignancies. Findings of a necrotic lymph node raise concern for infectious process and malignancy. A hypoechoic area on ultrasound/EBUS within a lymph node without blood flow is suggestive of pathologies like infections or malignancy. Inspection of the fluid could suggest a diagnosis; clear aspirates usually suggest bronchogenic or mediastinal cysts and purulent material suggests abscesses or necrotic lymph nodes. Growing tumor cells require a blood supply; if the vascular stroma is insufficient due to rapidly growing malignant tumors this could lead to large central areas of ischemic necrosis. Necrotic aspiration of lymph nodes is not always of infectious etiology. Aspiration of fluid in EBUS-TBNA is a rare occurrence, and malignancy should be considered when purulent fluid material is obtained. We present an elderly woman who underwent bronchoscopy with EBUS-TBNA for evaluation of upper lung nodule and mediastinal lymphadenopathy. Pus-like material was obtained on needle aspiration and endobronchial biopsy and mediastinal core biopsy revealed squamous cell carcinoma.
Collapse
|
56
|
Ouyang W, Guo P, Fang H, Frucht DM. Anthrax lethal toxin rapidly reduces c-Jun levels by inhibiting c-Jun gene transcription and promoting c-Jun protein degradation. J Biol Chem 2017; 292:17919-17927. [PMID: 28893904 DOI: 10.1074/jbc.m117.805648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/30/2017] [Indexed: 01/01/2023] Open
Abstract
Anthrax is a life-threatening disease caused by infection with Bacillus anthracis, which expresses lethal factor and the receptor-binding protective antigen. These two proteins combine to form anthrax lethal toxin (LT), whose proximal targets are mitogen-activated kinase kinases (MKKs). However, the downstream mediators of LT toxicity remain elusive. Here we report that LT exposure rapidly reduces the levels of c-Jun, a key regulator of cell proliferation and survival. Blockade of proteasome-dependent protein degradation with the 26S proteasome inhibitor MG132 largely restored c-Jun protein levels, suggesting that LT promotes degradation of c-Jun protein. Using the MKK1/2 inhibitor U0126, we further show that MKK1/2-Erk1/2 pathway inactivation similarly reduces c-Jun protein, which was also restored by MG132 pre-exposure. Interestingly, c-Jun protein rebounded to normal levels 4 h following U0126 exposure but not after LT exposure. The restoration of c-Jun in U0126-exposed cells was associated with increased c-Jun mRNA levels and was blocked by inactivation of the JNK1/2 signaling pathway. These results indicate that LT reduces c-Jun both by promoting c-Jun protein degradation via inactivation of MKK1/2-Erk1/2 signaling and by blocking c-Jun gene transcription via inactivation of MKK4-JNK1/2 signaling. In line with the known functions of c-Jun, LT also inhibited cell proliferation. Ectopic expression of LT-resistant MKK2 and MKK4 variants partially restored Erk1/2 and JNK1/2 signaling in LT-exposed cells, enabling the cells to maintain relatively normal c-Jun protein levels and cell proliferation. Taken together, these findings indicate that LT reduces c-Jun protein levels via two distinct mechanisms, thereby inhibiting critical cell functions, including cellular proliferation.
Collapse
Affiliation(s)
- Weiming Ouyang
- From the Division of Biotechnology Review and Research II, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| | - Pengfei Guo
- From the Division of Biotechnology Review and Research II, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| | - Hui Fang
- From the Division of Biotechnology Review and Research II, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| | - David M Frucht
- From the Division of Biotechnology Review and Research II, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| |
Collapse
|
57
|
Bacillus anthracis Edema Toxin Increases Fractional Free Water and Sodium Reabsorption in an Isolated Perfused Rat Kidney Model. Infect Immun 2017; 85:IAI.00264-17. [PMID: 28438974 DOI: 10.1128/iai.00264-17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 04/13/2017] [Indexed: 11/20/2022] Open
Abstract
Bacillus anthracis edema toxin (ET) consists of protective antigen (PA), necessary for host cell toxin uptake, and edema factor (EF), the toxic moiety which increases host cell cyclic AMP (cAMP). Since vasopressin stimulates renal water and sodium reabsorption via increased tubular cell cAMP levels, we hypothesized the ET would also do so. To test this hypothesis, we employed an isolated perfused rat kidney model. Kidneys were isolated and perfused with modified Krebs-Henseleit buffer. Perfusate and urine samples were obtained at baseline and every 10 min over 150 min following the addition of challenges with or without treatments to the perfusate. In kidneys perfused under constant flow or constant pressure, compared to PA challenge (n = 14 or 15 kidneys, respectively), ET (13 or 15 kidneys, respectively) progressively increased urine cAMP levels, water and sodium reabsorption, and urine osmolality and decreased urine output (P ≤ 0.04, except for sodium reabsorption under constant pressure [P = 0.17]). In ET-challenged kidneys, compared to placebo treatment, adefovir, an EF inhibitor, decreased urine cAMP levels, water and sodium reabsorption, and urine osmolality and increased urine output, while raxibacumab, a PA-directed monoclonal antibody (MAb), decreased urine cAMP levels, free water reabsorption, and urine osmolality and increased urine output (P ≤ 0.03 except for urine output with raxibacumab [P = 0.17]). Upon immunohistochemistry, aquaporin 2 was concentrated along the apical membrane of tubular cells with ET but not PA, and urine aquaporin 2 levels were higher with ET (5.52 ± 1.06 ng/ml versus 1.51 ± 0.44 ng/ml [means ± standard errors of the means {SEM}; P = 0.0001). Edema toxin has renal effects that could contribute to extravascular fluid collection characterizing anthrax infection clinically.
Collapse
|
58
|
Steenbergen J, Tanaka SK, Miller LL, Halasohoris SA, Hershfield JR. In Vitro and In Vivo Activity of Omadacycline against Two Biothreat Pathogens, Bacillus anthracis and Yersinia pestis. Antimicrob Agents Chemother 2017; 61:e02434-16. [PMID: 28223382 PMCID: PMC5404541 DOI: 10.1128/aac.02434-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/05/2017] [Indexed: 11/20/2022] Open
Abstract
The in vitro activity and in vivo efficacy of omadacycline (OMC) were evaluated against the causative pathogens of anthrax and plague, Bacillus anthracis and Yersinia pestis, respectively. MICs of OMC were determined by broth microdilution according to CLSI guidelines for 30 isolates each of Y. pestis and B. anthracis The in vivo efficacy of omadacycline was studied at a range of dosages in both a postexposure prophylaxis (PEP) murine model of anthrax and plague as well as in a delayed treatment model of inhalational anthrax. Omadacycline was active in vitro against Y. pestis (MIC90 of 1 μg/ml) and B. anthracis (MIC90 of 0.06 μg/ml). Omadacycline was less active in vitro than ciprofloxacin (CIP) against Y. pestis (CIP MIC90 of 0.03 μg/ml) but was more potent in vitro against B. anthracis (CIP MIC90 of 0.12 μg/ml). In the mouse model of infection, the survival curves for all treatment cohorts differed significantly from the vehicle control (P = 0.004). The median survival for the vehicle-treated controls was 6 days postchallenge, while all antibiotic-treated mice survived the entire study. Omadacycline treatment with 5, 10, or 20 mg/kg of body weight twice daily for 14 days had significant efficacy over the vehicle control in the treatment of aerosolized B. anthracis Additionally, for postexposure prophylaxis treatment of mice infected with Y. pestis, the survival curves for omadacycline (40 mg/kg twice daily), ciprofloxacin, and doxycycline cohorts differed significantly from the vehicle control (P < 0.0001). Omadacycline is potent and demonstrates efficacy against both B. anthracis and Y. pestis The well-characterized oral and intravenous pharmacokinetics, safety, and tolerability warrant further assessment of the potential utility of omadacycline in combating these serious biothreat organisms.
Collapse
Affiliation(s)
| | - S Ken Tanaka
- Paratek Pharmaceuticals, King of Prussia, Pennsylvania, USA
| | - Lynda L Miller
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Stephanie A Halasohoris
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Jeremy R Hershfield
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| |
Collapse
|
59
|
Gulseren D, Süzük-Yıldız S, Çelebi B, Kılıç S. Evaluation of clinical and serological findings for diagnosis of cutaneous anthrax infection after an outbreak. Cutan Ocul Toxicol 2017; 36:289-293. [PMID: 28076993 DOI: 10.1080/15569527.2017.1281288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Anthrax, caused by the bacterium Bacillus anthracis, is one of the oldest documented infectious diseases in both livestock and humans. We aimed to evaluate clinical findings and risk factors of patients with cutaneous anthrax infection and report anti-lethal factor (LF) IgG and anti-protective antigen (PA) IgG titers in the serologic diagnosis of disease. METHODS In this study, serum samples of 18 cutaneous anthrax patients were collected and anti-LF IgG and anti-PA IgG titers were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS Twelve (67%) males and 6 (33%) females, with a mean age of 36.06 ± 16.58 years were included in the study. Risk factors identified in the patient population studied were slaughtering (28%), flaying (56%), chopping meat (67%), burying diseased animal corpses (17%) and milking (6%) livestock. Black eschar formation (94%), pruritus (78%) and painful lymphadenopathy (61%) were first three common clinical signs and symptoms, respectively. Fourteen (78%) patients produced a positive IgG response against PA, 11 (61%) patients produced against LF. Three (17%) patients had no response to either antigen. CONCLUSIONS A detailed history of contact with sick animals or animal products along with clinical findings should be taken at the first step for the diagnosis of cutaneous anthrax infection. Serologic detection of anti-LF IgG and anti-PA IgG with ELISA may be useful auxillary method for establishing the diagnosis.
Collapse
Affiliation(s)
- Duygu Gulseren
- a Department of Dermatology , Ankara Polatlı State Hospital , Ankara , Turkey and
| | - Serap Süzük-Yıldız
- b Department of Microbiology Reference Laboratory , Public Health Institution of Turkey , Ankara , Turkey
| | - Bekir Çelebi
- b Department of Microbiology Reference Laboratory , Public Health Institution of Turkey , Ankara , Turkey
| | - Selçuk Kılıç
- b Department of Microbiology Reference Laboratory , Public Health Institution of Turkey , Ankara , Turkey
| |
Collapse
|
60
|
Karuppanan K, Duhra-Gill S, Kailemia MJ, Phu ML, Lebrilla CB, Dandekar AM, Rodriguez RL, Nandi S, McDonald KA. Expression, Purification, and Biophysical Characterization of a Secreted Anthrax Decoy Fusion Protein in Nicotiana benthamiana. Int J Mol Sci 2017; 18:E89. [PMID: 28054967 PMCID: PMC5297723 DOI: 10.3390/ijms18010089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/17/2016] [Accepted: 12/26/2016] [Indexed: 11/16/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Kalimuthu Karuppanan
- Department of Chemical Engineering, University of California, Davis, CA 95616, USA.
| | - Sifti Duhra-Gill
- Department of Chemical Engineering, University of California, Davis, CA 95616, USA.
| | - Muchena J Kailemia
- Department of Chemistry, University of California, Davis, CA 95616, USA.
| | - My L Phu
- Department of Plant Sciences, University of California, Davis, CA 95616, USA.
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, CA 95616, USA.
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA.
| | - Abhaya M Dandekar
- Department of Plant Sciences, University of California, Davis, CA 95616, USA.
| | - Raymond L Rodriguez
- Department of Molecular & Cellular Biology, University of California, Davis, CA 95616, USA.
| | - Somen Nandi
- Department of Molecular & Cellular Biology, University of California, Davis, CA 95616, USA.
| | - Karen A McDonald
- Department of Chemical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
61
|
Schwarz NG, Loderstaedt U, Hahn A, Hinz R, Zautner AE, Eibach D, Fischer M, Hagen RM, Frickmann H. Microbiological laboratory diagnostics of neglected zoonotic diseases (NZDs). Acta Trop 2017; 165:40-65. [PMID: 26391646 DOI: 10.1016/j.actatropica.2015.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 08/03/2015] [Accepted: 09/04/2015] [Indexed: 02/06/2023]
Abstract
This review reports on laboratory diagnostic approaches for selected, highly pathogenic neglected zoonotic diseases, i.e. anthrax, bovine tuberculosis, brucellosis, echinococcosis, leishmaniasis, rabies, Taenia solium-associated diseases (neuro-/cysticercosis & taeniasis) and trypanosomiasis. Diagnostic options, including microscopy, culture, matrix-assisted laser-desorption-ionisation time-of-flight mass spectrometry, molecular approaches and serology are introduced. These procedures are critically discussed regarding their diagnostic reliability and state of evaluation. For rare diseases reliable evaluation data are scarce due to the rarity of samples. If bio-safety level 3 is required for cultural growth, but such high standards of laboratory infrastructure are not available, serological and molecular approaches from inactivated sample material might be alternatives. Multiple subsequent testing using various test platforms in a stepwise approach may improve sensitivity and specificity. Cheap and easy to use tests, usually called "rapid diagnostic tests" (RDTs) may impact disease control measures, but should not preclude developing countries from state of the art diagnostics.
Collapse
|
62
|
HU JL, CUI LL, BAO CJ, TAN ZM, RUTHERFORD S, YING L, ZHANG ML, ZHU FC. Source and risk factors of a cutaneous anthrax outbreak, Jiangsu, Eastern China, 2012. Epidemiol Infect 2016; 144:2672-8. [PMID: 27277672 PMCID: PMC9150473 DOI: 10.1017/s0950268816001205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 05/15/2016] [Accepted: 05/25/2016] [Indexed: 11/07/2022] Open
Abstract
Anthrax is still a severe public health problem and threat to human health. A cutaneous anthrax outbreak occurred in Jiangsu Province, a non-endemic anthrax region of eastern China, from July to August 2012. Epidemiological and laboratory investigation were initiated to trace the source of infection and identify the risk factors of the outbreak. On 25 July 2012, 17 persons were exposed to a sick cow, which had been imported from northeast China a few days previously. Of the 17 exposed, eight developed symptoms between 1 and 8 days and were diagnosed as cutaneous anthrax cases. Three main genes of Bacillus anthracis were detected from both human and cow meat samples, indicating that the outbreak was associated with this infected cow. A retrospective cohort study showed that contact with blood and presence of skin damage contributed to the case infection with B. anthracis. The outbreak highlights the need to enhance quarantine for imported livestock, which should have been vaccinated prior to importation, the significance of education for high-risk individuals, and training for primary healthcare workers even in anthrax-free areas.
Collapse
Affiliation(s)
- J. L. HU
- Department of Acute Infectious Diseases Control and Prevention, Jiangsu Province Center for Disease Control and Prevention, Nanjing, PR China
| | - L. L. CUI
- Jinan Municipal Center for Disease Control and Prevention, Jinan, PR China
- Chinese Field Epidemiology Training Program, Beijing, PR China
| | - C. J. BAO
- Department of Acute Infectious Diseases Control and Prevention, Jiangsu Province Center for Disease Control and Prevention, Nanjing, PR China
| | - Z. M. TAN
- Department of Acute Infectious Diseases Control and Prevention, Jiangsu Province Center for Disease Control and Prevention, Nanjing, PR China
| | - S. RUTHERFORD
- Centre for Environment and Population Health, School of Environment, Griffith University, Brisbane, Australia
| | - L. YING
- Lianyungang Municipal Center for Disease Control and Prevention, Lianyungang, PR China
| | - M. L. ZHANG
- Ganyu County Center for Disease Control and Prevention, Ganyu, PR China
| | - F. C. ZHU
- Jiangsu Province Center for Disease Control and Prevention, Nanjing, PR China
| |
Collapse
|
63
|
Ca-asp bound X-ray structure and inhibition of Bacillus anthracis dihydroorotase (DHOase). Bioorg Med Chem 2016; 24:4536-4543. [PMID: 27499369 DOI: 10.1016/j.bmc.2016.07.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 01/01/2023]
Abstract
Dihydroorotase (DHOase) is the third enzyme in the de novo pyrimidine synthesis pathway and is responsible for the reversible cyclization of carbamyl-aspartate (Ca-asp) to dihydroorotate (DHO). DHOase is further divided into two classes based on several structural characteristics, one of which is the length of the flexible catalytic loop that interacts with the substrate, Ca-asp, regulating the enzyme activity. Here, we present the crystal structure of Class I Bacillus anthracis DHOase with Ca-asp in the active site, which shows the peptide backbone of glycine in the shorter loop forming the necessary hydrogen bonds with the substrate, in place of the two threonines found in Class II DHOases. Despite the differences in the catalytic loop, the structure confirms that the key interactions between the substrate and active site residues are similar between Class I and Class II DHOase enzymes, which we further validated by mutagenesis studies. B. anthracis DHOase is also a potential antibacterial drug target. In order to identify prospective inhibitors, we performed high-throughput screening against several libraries using a colorimetric enzymatic assay and an orthogonal fluorescence thermal binding assay. Surface plasmon resonance was used for determining binding affinity (KD) and competition analysis with Ca-asp. Our results highlight that the primary difference between Class I and Class II DHOase is the catalytic loop. We also identify several compounds that can potentially be further optimized as potential B. anthracis inhibitors.
Collapse
|
64
|
Li Y, Cui X, Xu W, Ohanjanian L, Sampath-Kumar H, Suffredini D, Moayeri M, Leppla S, Fitz Y, Eichacker PQ. Nitric oxide production contributes to Bacillus anthracis edema toxin-associated arterial hypotension and lethality: ex vivo and in vivo studies in the rat. Am J Physiol Heart Circ Physiol 2016; 311:H781-93. [PMID: 27448553 DOI: 10.1152/ajpheart.00163.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/19/2016] [Indexed: 01/26/2023]
Abstract
We showed previously that Bacillus anthracis edema toxin (ET), comprised of protective antigen (PA) and edema factor (EF), inhibits phenylephrine (PE)-induced contraction in rat aortic rings and these effects are diminished in endothelial-denuded rings. Therefore, employing rat aortic ring and in vivo models, we tested the hypothesis that nitric oxide (NO) contributes to ET's arterial effects. Compared with rings challenged with PA alone, ET (PA + EF) reduced PE-stimulated maximal contractile force (MCF) and increased the PE concentration producing 50% MCF (EC50) (P < 0.0001). Compared with placebo, l-nitro-arginine methyl-ester (l-NAME), an NO synthase (NOS) inhibitor, reduced ET's effects on MCF and EC50 in patterns that approached or were significant (P = 0.06 and 0.03, respectively). In animals challenged with 24-h ET infusions, l-NAME (0.5 or 1.0 mg·kg(-1)·h(-1)) coadministration increased survival to 17 of 28 animals (60.7%) compared with 4 of 27 (14.8%) given placebo (P = 0.01). Animals receiving l-NAME but no ET all survived. Compared with PBS challenge, ET increased NO levels at 24 h and l-NAME decreased these increases (P < 0.0001). ET infusion decreased mean arterial blood pressure (MAP) in placebo and l-NAME-treated animals (P < 0.0001) but l-NAME reduced decreases in MAP with ET from 9 to 24 h (P = 0.03 for the time interaction). S-methyl-l-thiocitrulline, a selective neuronal NOS inhibitor, had effects in rings and, at a high dose in vivo models, comparable to l-NAME, whereas N'-[3-(aminomethyl)benzyl]-acetimidamide, a selective inducible NOS inhibitor, did not. NO production contributes to ET's arterial relaxant, hypotensive, and lethal effects in the rat.
Collapse
Affiliation(s)
- Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Wanying Xu
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Lernik Ohanjanian
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Hanish Sampath-Kumar
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Dante Suffredini
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Mahtab Moayeri
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Stephen Leppla
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Yvonne Fitz
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Peter Q Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
65
|
Amador-Molina JC, Valerdi-Madrigal ED, Domínguez-Castillo RI, Sirota LA, Arciniega JL. Temperature-mediated recombinant anthrax protective antigen aggregate development: Implications for toxin formation and immunogenicity. Vaccine 2016; 34:4188-4195. [PMID: 27364097 DOI: 10.1016/j.vaccine.2016.06.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/04/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022]
Abstract
Anthrax vaccines containing recombinant PA (rPA) as the only antigen face a stability issue: rPA forms aggregates in solution after exposure to temperatures ⩾40°C, thus losing its ability to form lethal toxin (LeTx) with Lethal Factor. To study rPA aggregation's impact on immune response, we subjected rPA to several time and temperature combinations. rPA treated at 50°C for 30min formed high mass aggregates when analyzed by gel electrophoresis and failed to form LeTx as measured by a macrophage lysis assay (MLA). Aggregated rPA-formed LeTx was about 30 times less active than LeTx containing native rPA. Mice immunized with heat-treated rPA combined with Al(OH)3 developed antibody titers about 49 times lower than mice immunized with native rPA, as measured by a Toxicity Neutralization Assay (TNA). Enzyme Linked Immunosorbent Assay (ELISA) of the same immune sera showed anti-rPA titers only 2-7 times lower than titers elicited by native rPA. Thus, rPA's ability to form LeTx correlates with its production of neutralizing antibodies, and aggregation significantly impairs the protein's antibody response. However, while these findings suggest MLA has some value as an in-process quality test for rPA in new anthrax vaccines, they also confirm the superiority of TNA for use in vaccine potency.
Collapse
Affiliation(s)
- Juan C Amador-Molina
- Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States.
| | - Esther D Valerdi-Madrigal
- Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Rocío I Domínguez-Castillo
- Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Lev A Sirota
- Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Juan L Arciniega
- Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| |
Collapse
|
66
|
Abstract
Bacillus anthracis is killed by the interferon-inducible, ELR(−) CXC chemokine CXCL10. Previous studies showed that disruption of the gene encoding FtsX, a conserved membrane component of the ATP-binding cassette transporter-like complex FtsE/X, resulted in resistance to CXCL10. FtsX exhibits some sequence similarity to the mammalian CXCL10 receptor, CXCR3, suggesting that the CXCL10 N-terminal region that interacts with CXCR3 may also interact with FtsX. A C-terminal truncated CXCL10 was tested to determine if the FtsX-dependent antimicrobial activity is associated with the CXCR3-interacting N terminus. The truncated CXCL10 exhibited antimicrobial activity against the B. anthracis parent strain but not the ΔftsX mutant, which supports a key role for the CXCL10 N terminus. Mutations in FtsE, the conserved ATP-binding protein of the FtsE/X complex, resulted in resistance to both CXCL10 and truncated CXCL10, indicating that both FtsX and FtsE are important. Higher concentrations of CXCL10 overcame the resistance of the ΔftsX mutant to CXCL10, suggesting an FtsX-independent killing mechanism, likely involving its C-terminal α-helix, which resembles a cationic antimicrobial peptide. Membrane depolarization studies revealed that CXCL10 disrupted membranes of the B. anthracis parent strain and the ΔftsX mutant, but only the parent strain underwent depolarization with truncated CXCL10. These findings suggest that CXCL10 is a bifunctional molecule that kills B. anthracis by two mechanisms. FtsE/X-dependent killing is mediated through an N-terminal portion of CXCL10 and is not reliant upon the C-terminal α-helix. The FtsE/X-independent mechanism involves membrane depolarization by CXCL10, likely because of its α-helix. These findings present a new paradigm for understanding mechanisms by which CXCL10 and related chemokines kill bacteria. Chemokines are a class of molecules known for their chemoattractant properties but more recently have been shown to possess antimicrobial activity against a wide range of Gram-positive and Gram-negative bacterial pathogens. The mechanism(s) by which these chemokines kill bacteria is not well understood, but it is generally thought to be due to the conserved amphipathic C-terminal α-helix that resembles cationic antimicrobial peptides in charge and secondary structure. Our present study indicates that the interferon-inducible, ELR(−) chemokine CXCL10 kills the Gram-positive pathogen Bacillus anthracis through multiple molecular mechanisms. One mechanism is mediated by interaction of CXCL10 with the bacterial FtsE/X complex and does not require the presence of the CXCL10 C-terminal α-helix. The second mechanism is FtsE/X receptor independent and kills through membrane disruption due to the C-terminal α-helix. This study represents a new paradigm for understanding how chemokines exert an antimicrobial effect that may prove applicable to other bacterial species.
Collapse
|
67
|
Rapid Detection of Viable Bacillus anthracis Spores in Environmental Samples by Using Engineered Reporter Phages. Appl Environ Microbiol 2016; 82:2380-2387. [PMID: 26873316 DOI: 10.1128/aem.03772-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/04/2016] [Indexed: 12/26/2022] Open
Abstract
Bacillus anthracis, the causative agent of anthrax, was utilized as a bioterrorism agent in 2001 when spores were distributed via the U.S. postal system. In responding to this event, the Federal Bureau of Investigation used traditional bacterial culture viability assays to ascertain the extent of contamination of the postal facilities within 24 to 48 h of environmental sample acquisition. Here, we describe a low-complexity, second-generation reporter phage assay for the rapid detection of viableB. anthracis spores in environmental samples. The assay uses an engineered B. anthracis reporter phage (Wβ::luxAB-2) which transduces bioluminescence to infected cells. To facilitate low-level environmental detection and maximize the signal response, expression of luxABin an earlier version of the reporter phage (Wβ::luxAB-1) was optimized. These alterations prolonged signal kinetics, increased light output, and improved assay sensitivity. Using Wβ::luxAB-2, detection of B. anthracis spores was 1 CFU in 8 h from pure cultures and as low as 10 CFU/g in sterile soil but increased to 10(5)CFU/g in unprocessed soil due to an unstable signal and the presence of competing bacteria. Inclusion of semiselective medium, mediated by a phage-expressed antibiotic resistance gene, maintained signal stability and enabled the detection of 10(4)CFU/g in 6 h. The assay does not require spore extraction and relies on the phage infecting germinating cells directly in the soil sample. This reporter phage displays promise for the rapid detection of low levels of spores on clean surfaces and also in grossly contaminated environmental samples from complex matrices such as soils.
Collapse
|
68
|
Parlak E, Parlak M. Human Cutaneous Anthrax, the East Anatolian Region of Turkey 2008-2014. Vector Borne Zoonotic Dis 2015; 16:42-7. [PMID: 26720232 DOI: 10.1089/vbz.2015.1835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Anthrax is a zoonotic infectious disease caused by Bacillus anthracis. While anthrax is rare in developed countries, it is endemic in Turkey. The names of the different forms of the disease refer to the manner of entry of the spores into the body-cutaneous, gastrointestinal, inhalation, and injection. The purpose of this study was to evaluate the clinical characteristics, epidemiological history, treatment, and outcomes of patients with anthrax. Eighty-two cases of anthrax hospitalized at Atatürk University Faculty of Medicine Department of Infectious Diseases and Clinical Microbiology in 2008-2014 were examined retrospectively. Gender, age, occupation, year, history, clinical characteristics, character of lesions, length of hospitalization, and outcomes were recorded. Thirty (36.6%) patients were female and 52 (63.4%) patients were male; ages were 18-69 and mean age was 43.77 ± 13.05. The mean incubation period was 4.79 ± 3.76 days. Cases were largely identified in August (41.5%) and September (25.6%). Sixty-nine (84.1%) of the 82 patients had been given antibiotics before presentation. Lesions were most common on the fingers and arms. The most common occupational groups were housewives (36.6%) and people working in animal husbandry (31.7%). All patients had histories of contact with diseased animals and animal products. Penicillin-group antibiotics (78%) were most commonly used in treatment. One patient (1.2%) died from anthrax meningitis. The mean length of hospitalization was 8.30 ± 5.36 days. Anthrax is an endemic disease of economic and social significance for the region. Effective public health control measures, risk group education, vaccination of animals, and decontamination procedures will reduce the number of cases.
Collapse
Affiliation(s)
- Emine Parlak
- Department of Infectious Diseases and Clinical Microbiology, Atatürk University Faculty of Medicine , Erzurum, Turkey
| | - Mehmet Parlak
- Department of Infectious Diseases and Clinical Microbiology, Atatürk University Faculty of Medicine , Erzurum, Turkey
| |
Collapse
|
69
|
D'Amelio E, Gentile B, Lista F, D'Amelio R. Historical evolution of human anthrax from occupational disease to potentially global threat as bioweapon. ENVIRONMENT INTERNATIONAL 2015; 85:133-146. [PMID: 26386727 DOI: 10.1016/j.envint.2015.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 09/03/2015] [Accepted: 09/04/2015] [Indexed: 06/05/2023]
Abstract
PURPOSE Anthrax is caused by Bacillus anthracis, which can naturally infect livestock, wildlife and occupationally exposed humans. However, for its resistance due to spore formation, ease of dissemination, persistence in the environment and high virulence, B. anthracis has been considered the most serious bioterrorism agent for a long time. During the last century anthrax evolved from limited natural disease to potentially global threat if used as bioweapon. Several factors may mitigate the consequences of an anthrax attack, including 1. the capability to promptly recognize and manage the illness and its public health consequences; 2. the limitation of secondary contamination risk through an appropriate decontamination; and 3. the evolution of genotyping methods (for microbes characterization at high resolution level) that can influence the course and/or focus of investigations, impacting the response of the government to an attack. METHODS A PubMed search has been done using the key words “bioterrorism anthrax”. RESULTS Over one thousand papers have been screened and the most significant examined to present a comprehensive literature review in order to discuss the current knowledge and strategies in preparedness for a possible deliberate release of B. anthracis spores and to indicate the most current and complete documents in which to deepen. CONCLUSIONS The comprehensive analysis of the two most relevant unnatural anthrax release events, Sverdlovsk in the former Soviet Union (1979) and the contaminated letters in the USA (2001), shows that inhalational anthrax may easily and cheaply be spread resulting in serious consequences. The damage caused by an anthrax attack can be limited if public health organization, first responders, researchers and investigators will be able to promptly manage anthrax cases and use new technologies for decontamination methods and in forensic microbiology.
Collapse
Affiliation(s)
| | - Bernardina Gentile
- Histology and Molecular Biology Section, Army Medical Research Center, Via Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Florigio Lista
- Histology and Molecular Biology Section, Army Medical Research Center, Via Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Raffaele D'Amelio
- Sapienza University of Rome, Department of Clinical and Molecular Medicine, S. Andrea University Hospital, Via di Grottarossa 1039, 00189 Rome, Italy.
| |
Collapse
|
70
|
Abstract
Após 2001, a utilização de patógenos reforçou seu emprego como arma de guerra. Este estudo descritivo tem por objetivo discutir estratégias de contingenciamento em ataque por anthrax, auxiliando no reconhecimento precoce e estabelecimento de medidas de contenção. Profissionais de saúde necessitam reconhecer a infecção, pois em atentados, o elemento-chave é médico e não militar. O anthrax por inalação é a forma de atentado mais provável com 100% de mortalidade, caso não haja tratamento imediato. É altamente resistente; tem período de incubação de um a seis dias; seus sintomas iniciais são similares à influenza; só permite diagnóstico em NB3; a vacina é aquela recomendada para ocupações de risco, de disponibilidade restrita; e requer profilaxia antibiótica longa.
Collapse
|
71
|
Pontier-Bres R, Rampal P, Peyron JF, Munro P, Lemichez E, Czerucka D. The Saccharomyces boulardii CNCM I-745 strain shows protective effects against the B. anthracis LT toxin. Toxins (Basel) 2015; 7:4455-67. [PMID: 26529015 PMCID: PMC4663514 DOI: 10.3390/toxins7114455] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/15/2015] [Accepted: 10/26/2015] [Indexed: 12/11/2022] Open
Abstract
The probiotic yeast Saccharomyces boulardii (S. boulardii) has been prescribed for the prophylaxis and treatment of several infectious diarrheal diseases. Gastrointestinal anthrax causes fatal systemic disease. In the present study, we investigated the protective effects conferred by Saccharomyces boulardii CNCM I-745 strain on polarized T84 columnar epithelial cells intoxicated by the lethal toxin (LT) of Bacillus anthracis. Exposure of polarized T84 cells to LT affected cell monolayer integrity, modified the morphology of tight junctions and induced the formation of actin stress fibers. Overnight treatment of cells with S. boulardii before incubation with LT maintained the integrity of the monolayers, prevented morphological modification of tight junctions, restricted the effects of LT on actin remodeling and delayed LT-induced MEK-2 cleavage. Mechanistically, we demonstrated that in the presence of S. boulardii, the medium is depleted of both LF and PA sub-units of LT and the appearance of a cleaved form of PA. Our study highlights the potential of the S. boulardii CNCM I-745 strain as a prophylactic agent against the gastrointestinal form of anthrax.
Collapse
Affiliation(s)
| | - Patrick Rampal
- Centre Scientifique de Monaco, Monaco 98000, Monaco; E-Mails: (R.P.-B.); (P.R.)
| | - Jean-François Peyron
- Team Inflammation, Cancer, Cancer Stem Cells, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, U1065, Nice 06204, France; E-Mail:
- Faculté de Médecine, UFR Médecine, IFR50, Université de Nice-Sophia Antipolis, UNSA, Nice 06204, France; E-Mails: (P.M.); (E.L.)
| | - Patrick Munro
- Faculté de Médecine, UFR Médecine, IFR50, Université de Nice-Sophia Antipolis, UNSA, Nice 06204, France; E-Mails: (P.M.); (E.L.)
- Team Microbial Toxins in Host Pathogen Interactions, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, U1065, Nice 06204, France
| | - Emmanuel Lemichez
- Faculté de Médecine, UFR Médecine, IFR50, Université de Nice-Sophia Antipolis, UNSA, Nice 06204, France; E-Mails: (P.M.); (E.L.)
- Team Microbial Toxins in Host Pathogen Interactions, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, U1065, Nice 06204, France
| | - Dorota Czerucka
- Centre Scientifique de Monaco, Monaco 98000, Monaco; E-Mails: (R.P.-B.); (P.R.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +377-97-77-44-35
| |
Collapse
|
72
|
Enhanced Immune Response to DNA Vaccine Encoding Bacillus anthracis PA-D4 Protects Mice against Anthrax Spore Challenge. PLoS One 2015; 10:e0139671. [PMID: 26430894 PMCID: PMC4591996 DOI: 10.1371/journal.pone.0139671] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 09/16/2015] [Indexed: 11/19/2022] Open
Abstract
Anthrax has long been considered the most probable bioweapon-induced disease. The protective antigen (PA) of Bacillus anthracis plays a crucial role in the pathogenesis of anthrax. In the current study, we evaluated the efficiency of a genetic vaccination with the fourth domain (D4) of PA, which is responsible for initial binding of the anthrax toxin to the cellular receptor. The eukaryotic expression vector was designed with the immunoglobulin M (IgM) signal sequence encoding for PA-D4, which contains codon-optimized genes. The expression and secretion of recombinant protein was confirmed in vitro in 293T cells transfected with plasmid and detected by western blotting, confocal microscopy, and enzyme-linked immunosorbent assay (ELISA). The results revealed that PA-D4 protein can be efficiently expressed and secreted at high levels into the culture medium. When plasmid DNA was given intramuscularly to mice, a significant PA-D4-specific antibody response was induced. Importantly, high titers of antibodies were maintained for nearly 1 year. Furthermore, incorporation of the SV40 enhancer in the plasmid DNA resulted in approximately a 15-fold increase in serum antibody levels in comparison with the plasmid without enhancer. The antibodies produced were predominantly the immunoglobulin G2 (IgG2) type, indicating the predominance of the Th1 response. In addition, splenocytes collected from immunized mice produced PA-D4-specific interferon gamma (IFN-γ). The biodistribution study showed that plasmid DNA was detected in most organs and it rapidly cleared from the injection site. Finally, DNA vaccination with electroporation induced a significant increase in immunogenicity and successfully protected the mice against anthrax spore challenge. Our approach to enhancing the immune response contributes to the development of DNA vaccines against anthrax and other biothreats.
Collapse
|
73
|
Wycoff K, Maclean J, Belle A, Yu L, Tran Y, Roy C, Hayden F. Anti-infective immunoadhesins from plants. PLANT BIOTECHNOLOGY JOURNAL 2015; 13:1078-93. [PMID: 26242703 PMCID: PMC4749143 DOI: 10.1111/pbi.12441] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/24/2015] [Accepted: 06/27/2015] [Indexed: 05/22/2023]
Abstract
Immunoadhesins are recombinant proteins that combine the ligand-binding region of a receptor or adhesion molecule with immunoglobulin constant domains. All FDA-approved immunoadhesins are designed to modulate the interaction of a human receptor with its normal ligand, such as Etanercept (Enbrel(®) ), which interferes with the binding of tumour necrosis factor (TNF) to the TNF-alpha receptor and is used to treat inflammatory diseases such as rheumatoid arthritis. Like antibodies, immunoadhesins have long circulating half-lives, are readily purified by affinity-based methods and have the avidity advantages conferred by bivalency. Immunoadhesins that incorporate normal cellular receptors for viruses or bacterial toxins hold great, but as yet unrealized, potential for treating infectious disease. As decoy receptors, immunoadhesins have potential advantages over pathogen-targeted monoclonal antibodies. Planet Biotechnology has specialized in developing anti-infective immunoadhesins using plant expression systems. An immunoadhesin incorporating the cellular receptor for anthrax toxin, CMG2, potently blocks toxin activity in vitro and protects animals against inhalational anthrax. An immunoadhesin based on the receptor for human rhinovirus, ICAM-1, potently blocks infection of human cells by one of the major causes of the common cold. An immunoadhesin targeting the MERS coronavirus is in an early stage of development. We describe here the unique challenges involved in designing and developing immunoadhesins targeting infectious diseases in the hope of inspiring further research into this promising class of drugs.
Collapse
Affiliation(s)
| | | | | | - Lloyd Yu
- Planet Biotechnology Inc., Hayward, CA, USA
| | - Y Tran
- Planet Biotechnology Inc., Hayward, CA, USA
| | - Chad Roy
- Tulane National Primate Research Center, Covington, LA, USA
| | - Frederick Hayden
- University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
74
|
Ramachandran G, Gade P, Tsai P, Lu W, Kalvakolanu DV, Rosen GM, Cross AS. Potential role of autophagy in the bactericidal activity of human PMNs for Bacillus anthracis. Pathog Dis 2015; 73:ftv080. [PMID: 26424808 DOI: 10.1093/femspd/ftv080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2015] [Indexed: 12/19/2022] Open
Abstract
Bacillus anthracis, the causative agent of anthrax, is acquired by mammalian hosts from the environment, as quiescent endospores. These endospores must germinate inside host cells, forming vegetative bacilli, before they can express the virulence factors that enable them to evade host defenses and disseminate throughout the body. While the role of macrophages and dendritic cells in this initial interaction has been established, the role of polymorphonuclear leukocytes (PMNs) has not been adequately defined. We discovered that while B. anthracis 34F2 Sterne endospores germinate poorly within non-activated human PMNs, these phagocytes exhibit rapid microbicidal activity toward the outgrown vegetative bacilli, independent of superoxide and nitric oxide. These findings suggest that a non-free radical pathway kills B. anthracis bacilli. We also find in PMNs an autophagic mechanism of bacterial killing based on the rapid induction of LC-3 conversion, beclin-1 expression, sequestosome 1 (SQSTM1) degradation and inhibition of bactericidal activity by the inhibitor, 3-methyladenine. These findings extend to PMNs an autophagic bactericidal mechanism previously described for other phagocytes.
Collapse
Affiliation(s)
- Girish Ramachandran
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Padmaja Gade
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pei Tsai
- Department of Pharmaceutical Sciences, and the Center for EPR Imaging In Vivo Physiology, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Wuyuan Lu
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dhananjaya V Kalvakolanu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Gerald M Rosen
- Department of Pharmaceutical Sciences, and the Center for EPR Imaging In Vivo Physiology, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Alan S Cross
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
75
|
Fronczek CF, Yoon JY. Biosensors for Monitoring Airborne Pathogens. ACTA ACUST UNITED AC 2015; 20:390-410. [DOI: 10.1177/2211068215580935] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Indexed: 01/15/2023]
|
76
|
Powell JD, Hutchison JR, Hess BM, Straub TM. Bacillus anthracis spores germinate extracellularly at air-liquid interface in an in vitro lung model under serum-free conditions. J Appl Microbiol 2015; 119:711-23. [PMID: 26075586 PMCID: PMC4745038 DOI: 10.1111/jam.12872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/02/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022]
Abstract
Aims To better understand the parameters that govern spore dissemination after lung exposure using in vitro cell systems. Methods and Results We evaluated the kinetics of uptake, germination and proliferation of Bacillus anthracis Sterne spores in association with human primary lung epithelial cells, Calu‐3 and A549 cell lines. We also analysed the influence of various cell culture medium formulations related to spore germination. Conclusions We found negligible spore uptake by epithelial cells, but germination and proliferation of spores in the serum‐free extracellular environment was evident. Spore germination was appreciably higher in immortalized cell cultures than in primary epithelial cells. Additionally, spores still germinated apically at a mucus‐secreting air–liquid interface lung barrier that was devoid of cell culture medium much earlier than medium‐only controls. Significance and Impact of the Study The role of lung epithelial cells in B. anthracis spore dissemination after inhalation remains poorly defined and rather controversial. These results are novel as they show spore germination is appreciably enhanced in the presence of lung cells in vitro, however, the cell line and cell state (air–liquid interface vs submerged in medium) dictates the extent of germination and in some cases proliferation.
Collapse
Affiliation(s)
- J D Powell
- Chemical and Biological Signature Sciences Group, Pacific Northwest National Laboratory, Richland, WA, USA
| | - J R Hutchison
- Chemical and Biological Signature Sciences Group, Pacific Northwest National Laboratory, Richland, WA, USA
| | - B M Hess
- Chemical and Biological Signature Sciences Group, Pacific Northwest National Laboratory, Richland, WA, USA
| | - T M Straub
- Chemical and Biological Signature Sciences Group, Pacific Northwest National Laboratory, Richland, WA, USA
| |
Collapse
|
77
|
Immunization with a Recombinant, Pseudomonas fluorescens-Expressed, Mutant Form of Bacillus anthracis-Derived Protective Antigen Protects Rabbits from Anthrax Infection. PLoS One 2015. [PMID: 26207820 PMCID: PMC4514824 DOI: 10.1371/journal.pone.0130952] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Protective antigen (PA), one of the components of the anthrax toxin, is the major component of human anthrax vaccine (Biothrax). Human anthrax vaccines approved in the United States and Europe consist of an alum-adsorbed or precipitated (respectively) supernatant material derived from cultures of toxigenic, non-encapsulated strains of Bacillus anthracis. Approved vaccination schedules in humans with either of these vaccines requires several booster shots and occasionally causes adverse injection site reactions. Mutant derivatives of the protective antigen that will not form the anthrax toxins have been described. We have cloned and expressed both mutant (PA SNKE167-ΔFF-315-E308D) and native PA molecules recombinantly and purified them. In this study, both the mutant and native PA molecules, formulated with alum (Alhydrogel), elicited high titers of anthrax toxin neutralizing anti-PA antibodies in New Zealand White rabbits. Both mutant and native PA vaccine preparations protected rabbits from lethal, aerosolized, B. anthracis spore challenge subsequent to two immunizations at doses of less than 1 μg.
Collapse
|
78
|
Booth M, Donaldson L, Cui X, Sun J, Cole S, Dailsey S, Hart A, Johns N, McConnell P, McLennan T, Parcell B, Robb H, Shippey B, Sim M, Wallis C, Eichacker PQ. Confirmed Bacillus anthracis infection among persons who inject drugs, Scotland, 2009-2010. Emerg Infect Dis 2015; 20:1452-63. [PMID: 25148307 PMCID: PMC4178387 DOI: 10.3201/eid2009.131481] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Patients who died had an increased sequential organ failure assessment score and need for vasopressors.
Collapse
|
79
|
Tuntland ML, Wolf NM, Fung LWM. Differences in the purification and solution properties of PurC gene products from Streptococcus pneumoniae and Bacillus anthracis. Protein Expr Purif 2015; 114:143-8. [PMID: 26118696 DOI: 10.1016/j.pep.2015.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/02/2015] [Accepted: 05/14/2015] [Indexed: 11/24/2022]
Abstract
4-(N-succino)-5-aminoimidazole-4-carboxamide ribonucleotide synthetase (PurC) is a key enzyme in the de novo purine biosynthetic pathway of bacteria and an ideal target pathway for the discovery of antimicrobials. Bacillus anthracis (Ba) and Streptococcus pneumoniae (Sp) are two of the bacteria shown to be severe detriments to public health. To be able to carry out the experimentation that leads to drug discovery, high yields of pure soluble recombinant protein must first be obtained. We studied two recombinant PurC proteins from B. anthracis and S. pneumoniae, using Escherichia coli as the host cells. These two proteins, with very similar amino acid sequences, exhibit very different solution properties, leading to a large difference in yields during protein purification under the same conditions. The yield for SpPurC (>50mG per gram of cells) is ten times greater than that for BaPurC (<5mG per gram of cells). The BaPurC samples in solution consisted of oligomers and dimers, with dimers as its functional form. Comparing the yields of dimers, SpPurC is 25 times greater than that for BaPurC (∼2mG per gram of cell). Our studies suggest that the difference in exposed hydrophobic surface area is responsible for the difference in yields under the same conditions.
Collapse
Affiliation(s)
- Micheal L Tuntland
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nina M Wolf
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Leslie W-M Fung
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
80
|
Sharp NJ, Vandamm JP, Molineux IJ, Schofield DA. Rapid Detection of Bacillus anthracis in Complex Food Matrices Using Phage-Mediated Bioluminescence. J Food Prot 2015; 78:963-8. [PMID: 25951391 DOI: 10.4315/0362-028x.jfp-14-534] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bacillus anthracis, the causative agent of anthrax, is considered a high-priority agent that may be used in a food-related terrorist attack because it can be contracted by ingestion and it also forms spores with heat and chemical resistance. Thus, novel surveillance methodologies to detect B. anthracis on adulterated foods are important for bioterrorism preparedness. We describe the development of a phage-based bioluminescence assay for the detection of B. anthracis on deliberately contaminated foods. We previously engineered the B. anthracis phage Wβ with genes encoding bacterial luciferase (luxA and luxB) to create a "light-tagged" reporter (Wβ::luxAB) that is able to rapidly detect B. anthracis by transducing a bioluminescent signal response. Here, we investigate the ability of Wβ::luxAB to detect B. anthracis Sterne, an attenuated select agent strain, in inoculated food (ground beef) and milk (2%, baby formula, and half and half) matrices after incubation with spores for 72 h at 4°C as per AOAC testing guidelines. The majority of B. anthracis bacilli remained in spore form, and thus were potentially infectious, within each of the liquid matrices for 14 days. Detection limits were 80 CFU/ml after 7 h of enrichment; sensitivity of detection increased to 8 CFU/ml when enrichment was extended to 16 h. The limit of detection in ground beef was 3.2 × 10(3) CFU/g after 7 h of enrichment, improving to 3.2 × 10(2) CFU/g after 16 h. Because the time to result is rapid and minimal processing is required, and because gastrointestinal anthrax can be fatal, the reporter technology displays promise for the protection of our food supply following a deliberate release of this priority pathogen.
Collapse
Affiliation(s)
- Natasha J Sharp
- Guild BioSciences, 1313B Ashley River Road, Charleston, South Carolina 29407, USA
| | - Joshua P Vandamm
- Guild BioSciences, 1313B Ashley River Road, Charleston, South Carolina 29407, USA
| | - Ian J Molineux
- Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas 78712, USA
| | - David A Schofield
- Guild BioSciences, 1313B Ashley River Road, Charleston, South Carolina 29407, USA.
| |
Collapse
|
81
|
Ohanjanian L, Remy KE, Li Y, Cui X, Eichacker PQ. An overview of investigational toxin-directed therapies for the adjunctive management of Bacillus anthracis infection and sepsis. Expert Opin Investig Drugs 2015; 24:851-65. [PMID: 25920540 DOI: 10.1517/13543784.2015.1041587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sepsis with Bacillus anthracis infection has a very high mortality rate despite appropriate antibiotic and supportive therapies. Over the past 15 years, recent outbreaks in the US and in Europe, coupled with anthrax's bioterrorism weapon potential, have stimulated efforts to develop adjunctive therapies to improve clinical outcomes. Since lethal toxin and edema toxin (LT and ET) make central contributions to the pathogenesis of B. anthracis, these have been major targets in this effort. AREAS COVERED Here, the authors review different investigative biopharmaceuticals that have been recently identified for their therapeutic potential as inhibitors of LT or ET. Among these inhibitors are two antibody preparations that have been included in the Strategic National Stockpile (SNS) and several more that have reached Phase I testing. Presently, however, many of these candidate agents have only been studied in vitro and very few tested in bacteria-challenged models. EXPERT OPINION Although a large number of drugs have been identified as potential therapeutic inhibitors of LT and ET, in most cases their testing has been limited. The use of the two SNS antibody therapies during a large-scale exposure to B. anthracis will be difficult. Further testing and development of agents with oral bioavailability and relatively long shelf lives should be a focus for future research.
Collapse
Affiliation(s)
- Lernik Ohanjanian
- National Institutes of Health, Clinical Center, Critical Care Medicine Department , Building 10, Room 2C145, Bethesda, MD 20892 , USA +1 301 402 2914 ; +1 301 402 1213 ;
| | | | | | | | | |
Collapse
|
82
|
Abstract
The agents most likely to be used in bioterrorism attacks are reviewed, along with the clinical syndromes they produce and their treatment.
Collapse
|
83
|
Remy KE, Cui X, Li Y, Sun J, Solomon SB, Fitz Y, Barochia AV, Al-Hamad M, Moayeri M, Leppla SH, Eichacker PQ. Raxibacumab augments hemodynamic support and improves outcomes during shock with B. anthracis edema toxin alone or together with lethal toxin in canines. Intensive Care Med Exp 2015; 3:9. [PMID: 26097803 PMCID: PMC4473792 DOI: 10.1186/s40635-015-0043-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Lethal and edema toxin contribute to shock and lethality with Bacillus anthracis. We showed previously in a 96-h sedated canine model that raxibacumab, a monoclonal antibody against protective antigen, augmented hemodynamic support (HS) and improved survival with lethal toxin challenge. Here we study raxibacumab further. Using this model, we have now studied raxibacumab with 24 h edema toxin challenges (Study 1), and lethal and edema toxin challenges together (Study 2). METHODS Using our canine model, we have now studied raxibacumab with 24h edema toxin challenges (Study-1), and lethal and edema toxin challenges together (Study-2). RESULTS In Study 1, compared to no treatment, HS (titrated fluid and norepinephrine) increased mean arterial blood pressure (MAP, p ≤ 0.05) but not survival [0 of 10 (0/10) animals survived in each group] or median survival time [43.8 h (range 16.8 to 80.3) vs. 45.2 h (21.0 to 57.1)]. Compared to HS, HS with raxibacumab treatment at or 6 h after the beginning of edema toxin increased MAP and survival rate (6/7 and 7/8, respectively) and time [96.0 h (39.5 to 96.0) and 96.0 h (89.5 to 96.0), respectively]; (p ≤ 0.05). HS with raxibacumab at 12 h increased MAP (p ≤ 0.05) but not survival [1/5; 55.3 h (12.6 to 96.0)]. In Study-2, survival rate and time increased with HS and raxibacumab at 0 h (4/4) or 6 h after (3/3) beginning lethal and edema toxin compared to HS [0/5; 71.5 h (65 to 93)] (p = 0.01 averaged over raxibacumab groups). CONCLUSIONS Raxibacumab augments HS and improves survival during shock with lethal and edema toxin.
Collapse
Affiliation(s)
- Kenneth E Remy
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Steven B Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Yvonne Fitz
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Amisha V Barochia
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Mariam Al-Hamad
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Mahtab Moayeri
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| | - Stephen H Leppla
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| | - Peter Q Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| |
Collapse
|
84
|
Arapović J, Skočibusić S, Jelavić B, Ivanković HB, Jurić M, Mamić D, Grgić S, Lesko J, Leventić M, Soldo I, Ravlija J, Nikolić J. Two cases of human cutaneous anthrax in Bosnia and Herzegovina, September 2014. ACTA ACUST UNITED AC 2015; 20:2-4. [PMID: 25719961 DOI: 10.2807/1560-7917.es2015.20.7.21039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Two cases of human cutaneous anthrax were reported in September 2014 in south-western Bosnia and Herzegovina. The two men were involved in slaughtering a cow and handled its Bacillus anthracis-infected meat. Anthrax has been sporadically observed in livestock in Bosnia and Herzegovina, but no confirmed human cases had been reported in the country in the last two decades. Clinicians in the country should be aware that anthrax may occur in humans, arising from exposure to infected animals.
Collapse
Affiliation(s)
- J Arapović
- Department of Infectious Diseases, University Hospital Mostar, Mostar, Bosnia and Herzegovina
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Hassett KJ, Vance DJ, Jain NK, Sahni N, Rabia LA, Cousins MC, Joshi S, Volkin DB, Middaugh CR, Mantis NJ, Carpenter JF, Randolph TW. Glassy-state stabilization of a dominant negative inhibitor anthrax vaccine containing aluminum hydroxide and glycopyranoside lipid A adjuvants. J Pharm Sci 2015; 104:627-39. [PMID: 25581103 DOI: 10.1002/jps.24295] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/11/2014] [Accepted: 10/28/2014] [Indexed: 01/14/2023]
Abstract
During transport and storage, vaccines may be exposed to temperatures outside of the range recommended for storage, potentially causing efficacy losses. To better understand and prevent such losses, dominant negative inhibitor (DNI), a recombinant protein antigen for a candidate vaccine against anthrax, was formulated as a liquid and as a glassy lyophilized powder with the adjuvants aluminum hydroxide and glycopyranoside lipid A (GLA). Freeze-thawing of the liquid vaccine caused the adjuvants to aggregate and decreased its immunogenicity in mice. Immunogenicity of liquid vaccines also decreased when stored at 40°C for 8 weeks, as measured by decreases in neutralizing antibody titers in vaccinated mice. Concomitant with efficacy losses at elevated temperatures, changes in DNI structure were detected by fluorescence spectroscopy and increased deamidation was observed by capillary isoelectric focusing (cIEF) after only 1 week of storage of the liquid formulation at 40°C. In contrast, upon lyophilization, no additional deamidation after 4 weeks at 40°C and no detectable changes in DNI structure or reduction in immunogenicity after 16 weeks at 40°C were observed. Vaccines containing aluminum hydroxide and GLA elicited higher immune responses than vaccines adjuvanted with only aluminum hydroxide, with more mice responding to a single dose.
Collapse
Affiliation(s)
- Kimberly J Hassett
- Department of Chemical and Biological Engineering, Center for Pharmaceutical Biotechnology, University of Colorado, Boulder, Boulder, Colorado, 80303
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Jeon JH, Kim YH, Choi MK, Kim KA, Lee HR, Jang J, Kim YR, Chun JH, Eo SK, Kim TS, Rhie GE. Bacillus anthracis genomic DNA enhances lethal toxin-induced cytotoxicity through TNF-α production. BMC Microbiol 2014; 14:300. [PMID: 25472474 PMCID: PMC4267052 DOI: 10.1186/s12866-014-0300-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/18/2014] [Indexed: 11/17/2022] Open
Abstract
Background Bacillus anthracis is the etiological agent of anthrax. Lethal toxin (LT) produced by B. anthracis is a well-known key virulence factor for anthrax because of its strong cytotoxic activity. However, little is known about the role of B. anthracis genomic DNA (BAG) in anthrax pathogenesis. Results We examined the effect of BAG on TNF-α production and LT-mediated cytotoxicity during B. anthracis spore infection in mouse macrophage cell lines (RAW264.7 cells and J774A.1) and BALB/c mice. Infection of RAW264.7 cells with B. anthracis spores induced TNF-α expression in a multiplicity of infection (MOI)-dependent manner, and this enhancement was attenuated by the toll-like receptor (TLR) 9 inhibitor oligodeoxynucleotide (ODN)2088. BAG led to TNF-α expression in a dose- and time-dependent manner when applied to RAW264.7 cells. TNF-α expression induced by BAG was reduced by either pretreatment with TLR9 inhibitors (ODN2088 and chloroquine (CQ)) or transfection with TLR9 siRNA. Furthermore, BAG-induced TNF-α production in TLR9+/+ macrophages was completely abrogated in TLR9−/− macrophages. BAG enhanced the phosphorylation of mitogen-activated protein kinases (MAPK), and BAG-induced TNF-α expression was attenuated by pretreatment with MAPK inhibitors. A reporter gene assay and confocal microscopy demonstrated that BAG increased NF-κB activation, which is responsible for TNF-α expression. Treatment with BAG alone showed no cytotoxic activity on the macrophage cell line J774A.1, whereas LT-mediated cytotoxicity was enhanced by treatment with BAG or TNF-α. Enhanced LT-induced lethality was also confirmed by BAG administration in mice. Furthermore, LT plus BAG-mediated lethality was significantly recovered by administration of Infliximab, an anti-TNF-α monoclonal antibody. Conclusions Our results suggest that B. anthracis DNA may contribute to anthrax pathogenesis by enhancing LT activity via TLR9-mediated TNF-α production. Electronic supplementary material The online version of this article (doi:10.1186/s12866-014-0300-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jun Ho Jeon
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Yeon Hee Kim
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea. .,School of Life Sciences and Biotechnology, Korea University, Seoul, 136-701, Republic of Korea.
| | - Min Kyung Choi
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Kyung Ae Kim
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Hae-Ri Lee
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Jeyoun Jang
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Yu-Ri Kim
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Jeong-Hoon Chun
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| | - Seong Kug Eo
- College of Veterinary medicine and Bio-Safety Research Institute, Chonbuk National University, Jeonju, 561-765, Republic of Korea.
| | - Tae Sung Kim
- School of Life Sciences and Biotechnology, Korea University, Seoul, 136-701, Republic of Korea.
| | - Gi-Eun Rhie
- Division of High-risk Pathogen Research, Center for Infectious Diseases, National Institute of Health, 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 361-951, Republic of Korea.
| |
Collapse
|
87
|
Rouli L, MBengue M, Robert C, Ndiaye M, La Scola B, Raoult D. Genomic analysis of three African strains of Bacillus anthracis demonstrates that they are part of the clonal expansion of an exclusively pathogenic bacterium. New Microbes New Infect 2014; 2:161-9. [PMID: 25566394 PMCID: PMC4265047 DOI: 10.1002/nmi2.62] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 07/04/2014] [Accepted: 08/08/2014] [Indexed: 01/31/2023] Open
Abstract
Bacillus anthracis is the causative agent of anthrax and is classified as a
‘Category A’ biological weapon. Six complete genomes of
B. anthracis (A0248, Ames, Ames Ancestor, CDC684, H0491, and Sterne) are
currently available. In this report, we add three African strain genomes: Sen2Col2, Sen3 and Gmb1.
To study the pan-genome of B. anthracis, we used bioinformatics tools, such
as Cluster of Orthologous Groups, and performed phylogenetic analysis. We found that the three
African strains contained the pX01 and pX02 plasmids, the nonsense mutation in the
plcR gene and the four known prophages. These strains are most similar to the
CDC684 strain and belong to the A cluster. We estimated that the
B. anthracis pan-genome has 2893 core genes (99% of the genome size)
and 85 accessory genes. We validated the hypothesis that B. anthracis has a
closed pan-genome and found that the three African strains carry the two plasmids associated with
bacterial virulence. The pan-genome nature of B. anthracis confirms its lack
of exchange (similar to Clostridium tetani) and supports its exclusively pathogenic
role, despite its survival in the environment. Moreover, thanks to the study of the core content
single nucleotide polymorphisms, we can see that our three African strains diverged very recently
from the other B. anthracis strains.
Collapse
Affiliation(s)
- L Rouli
- Aix Marseille Université, URMITE, UM63, CNRS 7278, IRD 198, Inserm 1095 Marseille, France
| | - M MBengue
- Laboratoire National d'Elevage et des Recherches Vétérinaires (LNERV), Institut Sénégalais de Recherches Agricoles (ISRA) Hann, Dakar, Senegal
| | - C Robert
- Aix Marseille Université, URMITE, UM63, CNRS 7278, IRD 198, Inserm 1095 Marseille, France
| | - M Ndiaye
- Laboratoire de Biologie Cellulaire, Faculté des Sciences et Techniques de l'Université Cheikh Anta DIOP de Dakar (UCAD) Dakar, Senegal
| | - B La Scola
- Aix Marseille Université, URMITE, UM63, CNRS 7278, IRD 198, Inserm 1095 Marseille, France
| | - D Raoult
- Aix Marseille Université, URMITE, UM63, CNRS 7278, IRD 198, Inserm 1095 Marseille, France
| |
Collapse
|
88
|
Gurnev PA, Nestorovich EM. Channel-forming bacterial toxins in biosensing and macromolecule delivery. Toxins (Basel) 2014; 6:2483-540. [PMID: 25153255 PMCID: PMC4147595 DOI: 10.3390/toxins6082483] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/08/2014] [Accepted: 08/08/2014] [Indexed: 12/19/2022] Open
Abstract
To intoxicate cells, pore-forming bacterial toxins are evolved to allow for the transmembrane traffic of different substrates, ranging from small inorganic ions to cell-specific polypeptides. Recent developments in single-channel electrical recordings, X-ray crystallography, protein engineering, and computational methods have generated a large body of knowledge about the basic principles of channel-mediated molecular transport. These discoveries provide a robust framework for expansion of the described principles and methods toward use of biological nanopores in the growing field of nanobiotechnology. This article, written for a special volume on "Intracellular Traffic and Transport of Bacterial Protein Toxins", reviews the current state of applications of pore-forming bacterial toxins in small- and macromolecule-sensing, targeted cancer therapy, and drug delivery. We discuss the electrophysiological studies that explore molecular details of channel-facilitated protein and polymer transport across cellular membranes using both natural and foreign substrates. The review focuses on the structurally and functionally different bacterial toxins: gramicidin A of Bacillus brevis, α-hemolysin of Staphylococcus aureus, and binary toxin of Bacillus anthracis, which have found their "second life" in a variety of developing medical and technological applications.
Collapse
Affiliation(s)
- Philip A Gurnev
- Physics Department, University of Massachusetts, Amherst, MA 01003, USA.
| | | |
Collapse
|
89
|
Abstract
This retrospective, descriptive case-series reviews the clinical presentations and significant laboratory findings of patients diagnosed with and treated for injectional anthrax (IA) since December 2009 at Monklands Hospital in Central Scotland and represents the largest series of IA cases to be described from a single location. Twenty-one patients who fulfilled National Anthrax Control Team standardized case definitions of confirmed, probable or possible IA are reported. All cases survived and none required limb amputation in contrast to an overall mortality of 28% being experienced for this condition in Scotland. We document the spectrum of presentations of soft tissue infection ranging from mild cases which were managed predominantly with oral antibiotics to severe cases with significant oedema, organ failure and coagulopathy. We describe the surgical management, intensive care management and antibiotic management including the first description of daptomycin being used to treat human anthrax. It is noted that some people who had injected heroin infected with Bacillus anthracis did not develop evidence of IA. Also highlighted are biochemical and haematological parameters which proved useful in identifying deteriorating patients who required greater levels of support and surgical debridement.
Collapse
|
90
|
Role of YpeB in cortex hydrolysis during germination of Bacillus anthracis spores. J Bacteriol 2014; 196:3399-409. [PMID: 25022853 DOI: 10.1128/jb.01899-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The infectious agent of the disease anthrax is the spore of Bacillus anthracis. Bacterial spores are extremely resistant to environmental stresses, which greatly hinders spore decontamination efforts. The spore cortex, a thick layer of modified peptidoglycan, contributes to spore dormancy and resistance by maintaining the low water content of the spore core. The cortex is degraded by germination-specific lytic enzymes (GSLEs) during spore germination, rendering the cells vulnerable to common disinfection techniques. This study investigates the relationship between SleB, a GSLE in B. anthracis, and YpeB, a protein necessary for SleB stability and function. The results indicate that ΔsleB and ΔypeB spores exhibit similar germination phenotypes and that the two proteins have a strict codependency for their incorporation into the dormant spore. In the absence of its partner protein, SleB or YpeB is proteolytically degraded soon after expression during sporulation, rather than escaping the developing spore. The three PepSY domains of YpeB were examined for their roles in the interaction with SleB. YpeB truncation mutants illustrate the necessity of a region beyond the first PepSY domain for SleB stability. Furthermore, site-directed mutagenesis of highly conserved residues within the PepSY domains resulted in germination defects corresponding to reduced levels of both SleB and YpeB in the mutant spores. These results identify residues involved in the stability of both proteins and reiterate their codependent relationship. It is hoped that the study of GSLEs and interacting proteins will lead to the use of GSLEs as targets for efficient activation of spore germination and facilitation of spore cleanup.
Collapse
|
91
|
Abstract
Anthrax is a highly contagious and potentially fatal human disease caused by Bacillus anthracis, an aerobic, Gram-positive, spore-forming rod-shaped bacterium with worldwide distribution as a zoonotic infection in herbivore animals. Bioterrorist attacks with inhalational anthrax have prompted the development of more effective treatments. Antibodies against anthrax toxin have been shown to decrease mortality in animal studies. Raxibacumab is a recombinant human monoclonal antibody developed against inhalational anthrax. The drug received approval after human studies showed its safety and animal studies demonstrated its efficacy for treatment as well as prophylaxis against inhalational anthrax. It works by preventing binding of the protective antigen component of the anthrax toxin to its receptors in host cells, thereby blocking the toxin's deleterious effects. Recently updated therapy guidelines for Bacillus anthracis recommend the use of antitoxin treatment. Raxibacumab is the first monoclonal antitoxin antibody made available that can be used with the antibiotics recommended for treatment of the disease. When exposure is suspected, raxibacumab should be given with anthrax vaccination to augment immunity. Raxibacumab provides additional protection against inhalational anthrax via a mechanism different from that of either antibiotics or active immunization. In combination with currently available and recommended therapies, raxibacumab should reduce the morbidity and mortality of inhalational anthrax.
Collapse
Affiliation(s)
- Carlos E Kummerfeldt
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
92
|
Arévalo MT, Navarro A, Arico CD, Li J, Alkhatib O, Chen S, Diaz-Arévalo D, Zeng M. Targeted silencing of anthrax toxin receptors protects against anthrax toxins. J Biol Chem 2014; 289:15730-8. [PMID: 24742682 DOI: 10.1074/jbc.m113.538587] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Anthrax spores can be aerosolized and dispersed as a bioweapon. Current postexposure treatments are inadequate at later stages of infection, when high levels of anthrax toxins are present. Anthrax toxins enter cells via two identified anthrax toxin receptors: tumor endothelial marker 8 (TEM8) and capillary morphogenesis protein 2 (CMG2). We hypothesized that host cells would be protected from anthrax toxins if anthrax toxin receptor expression was effectively silenced using RNA interference (RNAi) technology. Thus, anthrax toxin receptors in mouse and human macrophages were silenced using targeted siRNAs or blocked with specific antibody prior to challenge with anthrax lethal toxin. Viability assays were used to assess protection in macrophages treated with specific siRNA or antibody as compared with untreated cells. Silencing CMG2 using targeted siRNAs provided almost complete protection against anthrax lethal toxin-induced cytotoxicity and death in murine and human macrophages. The same results were obtained by prebinding cells with specific antibody prior to treatment with anthrax lethal toxin. In addition, TEM8-targeted siRNAs also offered significant protection against lethal toxin in human macrophage-like cells. Furthermore, silencing CMG2, TEM8, or both receptors in combination was also protective against MEK2 cleavage by lethal toxin or adenylyl cyclase activity by edema toxin in human kidney cells. Thus, anthrax toxin receptor-targeted RNAi has the potential to be developed as a life-saving, postexposure therapy against anthrax.
Collapse
Affiliation(s)
- Maria T Arévalo
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Ashley Navarro
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Chenoa D Arico
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Junwei Li
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Omar Alkhatib
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Shan Chen
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Diana Diaz-Arévalo
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| | - Mingtao Zeng
- From the Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905
| |
Collapse
|
93
|
Srinath R, Siva R, Babu S. B
acillus anthracis
-Like Strain-Carrying P
seudomonas
FPVA
Gene Occurs as Endophyte in Vegetables. J Food Saf 2014. [DOI: 10.1111/jfs.12095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- R. Srinath
- School of Biosciences and Technology; VIT University; Vellore 632014 India
| | - R. Siva
- School of Biosciences and Technology; VIT University; Vellore 632014 India
| | - S. Babu
- School of Biosciences and Technology; VIT University; Vellore 632014 India
| |
Collapse
|
94
|
Kim NY, Kang CI, Hur GH, Yang JM, Shin S. Bacillus anthracis lethal toxin induces cell-type-specific cytotoxicity in human lung cell lines. J Appl Microbiol 2014; 116:1334-43. [PMID: 24471528 DOI: 10.1111/jam.12457] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 01/13/2014] [Accepted: 01/23/2014] [Indexed: 12/20/2022]
Abstract
AIMS Inhalational anthrax is caused by the entry of Bacillus anthracis spores into the lung. Inhaled spores are phagocytosed by alveolar macrophages. Bacilli then escape from the macrophage and spread to other cells, initiating a systemic anthrax infection. Based on the pathological studies of primate and human inhalational anthrax cases, it appears that lung tissue injury is a lethal consequence of the disease. Although the cytotoxicity of anthrax lethal toxin to macrophages is well known, it is not clear how anthrax toxin affects the various lung cell types. METHODS AND RESULTS Using model cell lines representing different physiological compartments of the lung, we have investigated the cytotoxic effects of anthrax lethal toxin. The cell response was evaluated through MTT metabolism, neutral red uptake, initiation of apoptosis, and expression and binding activity of anthrax toxin receptors. We found that a human small airway epithelial cell line, HSAEC, was susceptible to anthrax lethal toxin. The other cell lines, A549, MRC-5, H358 and SKLU-1, displayed resistance to anthrax lethal toxin-mediated toxicity, although the expression of anthrax toxin receptors was detected in all the cell lines tested. CONCLUSIONS Our results indicate that cell-type-specific toxicity may be induced by anthrax lethal toxin in human lung tissues and does not correlate with anthrax toxin receptor expression levels. SIGNIFICANCE AND IMPACT OF THE STUDY This work suggests that cell-type-specific cytotoxicity of anthrax toxin in lung cells may cause subsequent lung disease progression. It may explain the initial pathogenic step of inhalational anthrax.
Collapse
Affiliation(s)
- N Y Kim
- Department of Life Science, Sogang University, Shinsu-Dong, Mapo, Seoul, Korea
| | | | | | | | | |
Collapse
|
95
|
Tripathi A, Schofield MM, Chlipala GE, Schultz PJ, Yim I, Newmister SA, Nusca TD, Scaglione JB, Hanna PC, Tamayo-Castillo G, Sherman DH. Baulamycins A and B, broad-spectrum antibiotics identified as inhibitors of siderophore biosynthesis in Staphylococcus aureus and Bacillus anthracis. J Am Chem Soc 2014; 136:1579-86. [PMID: 24401083 PMCID: PMC4028973 DOI: 10.1021/ja4115924] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Siderophores are high-affinity iron chelators produced by microorganisms and frequently contribute to the virulence of human pathogens. Targeted inhibition of the biosynthesis of siderophores staphyloferrin B of Staphylococcus aureus and petrobactin of Bacillus anthracis hold considerable potential as a single or combined treatment for methicillin-resistant S. aureus (MRSA) and anthrax infection, respectively. The biosynthetic pathways for both siderophores involve a nonribosomal peptide synthetase independent siderophore (NIS) synthetase, including SbnE in staphyloferrin B and AsbA in petrobactin. In this study, we developed a biochemical assay specific for NIS synthetases to screen for inhibitors of SbnE and AsbA against a library of marine microbial-derived natural product extracts (NPEs). Analysis of the NPE derived from Streptomyces tempisquensis led to the isolation of the novel antibiotics baulamycins A (BmcA, 6) and B (BmcB, 7). BmcA and BmcB displayed in vitro activity with IC50 values of 4.8 μM and 19 μM against SbnE and 180 μM and 200 μM against AsbA, respectively. Kinetic analysis showed that the compounds function as reversible competitive enzyme inhibitors. Liquid culture studies with S. aureus , B. anthracis , E. coli , and several other bacterial pathogens demonstrated the capacity of these natural products to penetrate bacterial barriers and inhibit growth of both Gram-positive and Gram-negative species. These studies provide proof-of-concept that natural product inhibitors targeting siderophore virulence factors can provide access to novel broad-spectrum antibiotics, which may serve as important leads for the development of potent anti-infective agents.
Collapse
Affiliation(s)
- Ashootosh Tripathi
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
| | - Michael M. Schofield
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - George E. Chlipala
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
| | - Pamela J. Schultz
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
| | - Isaiah Yim
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
| | - Sean A. Newmister
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
| | - Tyler D. Nusca
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jamie B. Scaglione
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
| | - Philip C. Hanna
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Giselle Tamayo-Castillo
- Unidad Estrategica de Bioprospección, Instituto Nacional de Biodiversidad (INBio), Santo Domingo de Heredia, Costa Rica & CIPRONA, Escuela de Química, Universidad de Costa Rica, 2060 San Pedro, Costa Rica
| | - David H. Sherman
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
- Departments of Medicinal Chemistry and Chemistry, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
96
|
Pneumonia. PRINCIPLES OF PULMONARY MEDICINE 2014. [PMCID: PMC7170200 DOI: 10.1016/b978-1-4557-2532-8.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
97
|
Ouyang W, Torigoe C, Fang H, Xie T, Frucht DM. Anthrax lethal toxin inhibits translation of hypoxia-inducible factor 1α and causes decreased tolerance to hypoxic stress. J Biol Chem 2013; 289:4180-90. [PMID: 24366872 DOI: 10.1074/jbc.m113.530006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hypoxia is considered to be a contributor to the pathology associated with administration of anthrax lethal toxin (LT). However, we report here that serum lactate levels in LT-treated mice are reduced, a finding inconsistent with the anaerobic metabolism expected to occur during hypoxia. Reduced lactate levels are also observed in the culture supernatants of LT-treated cells. LT inhibits the accumulation of hypoxia-inducible factor (HIF)-1α, a subunit of HIF-1, the master regulator directing cellular responses to hypoxia. The toxin has no effect on the transcription or protein turnover of HIF-1α, but instead it acts to inhibit HIF-1α translation. LT treatment diminishes phosphorylation of eIF4B, eIF4E, and rpS6, critical components of the intracellular machinery required for HIF-1α translation. Moreover, blockade of MKK1/2-ERK1/2, but not p38 or JNK signaling, lowers HIF-1α protein levels in both normoxic and hypoxic conditions, consistent with a role for MKK1 and MKK2 as the major targets of LT responsible for the inhibition of HIF-1α translation. The physiological importance of the LT-induced translation blockade is demonstrated by the finding that LT treatment decreases the survival of hepatocyte cell lines grown in hypoxic conditions, an effect that is overcome by preinduction of HIF-1α. Taken together, these data support a role for LT in dysregulating HIF-1α and thereby disrupting homeostatic responses to hypoxia, an environmental characteristic of certain tissues at baseline and/or during disseminated infection with Bacillus anthracis.
Collapse
Affiliation(s)
- Weiming Ouyang
- From the Division of Monoclonal Antibodies, Office of Biotechnology Products, Office of Pharmaceutical Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland 20892
| | | | | | | | | |
Collapse
|
98
|
Abstract
Bioterrorism is not only a reality of the times in which we live but bioweapons have been used for centuries. Critical care physicians play a major role in the recognition of and response to a bioterrorism attack. Critical care clinicians must be familiar with the diagnosis and management of the most likely bioterrorism agents, and also be adequately prepared to manage a mass casualty situation. This article reviews the epidemiology, diagnosis, and treatment of the most likely agents of biowarfare and bioterrorism.
Collapse
Affiliation(s)
- Michael D Christian
- Royal Canadian Air Force, Department of National Defence, 600 University Avenue, Room 18-232-1, Toronto, Ontario M5G 1X5, Canada.
| |
Collapse
|
99
|
Elad D. An unholy disease in the Holy Land: the history of anthrax between the Jordan River and the Mediterranean Sea (1909-2012). Vet J 2013; 199:319-23. [PMID: 24135549 DOI: 10.1016/j.tvjl.2013.08.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 08/26/2013] [Accepted: 08/28/2013] [Indexed: 11/27/2022]
Abstract
The history of recorded cases of anthrax in human beings and animals from 1909 to 2012 in the area between the Jordan River and the Mediterranean Sea is reviewed. The disease was endemic until the middle of the 20th century, but the incidence decreased thereafter, with only sporadic cases from the 1980s onwards. Human cases have not been diagnosed in the region since 1984 and the number of episodes of animal disease has reduced to less than one per year. This decline is mostly due to the disruption of the infective cycle by improved veterinary control, including vaccination, treatment and outbreak management. A policy of reactive vaccination for 10 years of affected herds and herds grazing in their proximity has been applied. No new outbreaks have been observed in such herds after the cessation of vaccination, despite continued grazing on the same sites, so it is assumed that spore survival in such areas is shorter than 10 years. This is independent of the soil composition, which is calcareous throughout most of the relevant area. However, reemergence of anthrax, even after decades, has occurred following disturbance and heavy rainfall.
Collapse
Affiliation(s)
- Daniel Elad
- Department of Clinical Bacteriology and Mycology, Kimron Veterinary Institute, Bet Dagan 50250, Israel.
| |
Collapse
|
100
|
Remy KE, Qiu P, Li Y, Cui X, Eichacker PQ. B. anthracis associated cardiovascular dysfunction and shock: the potential contribution of both non-toxin and toxin components. BMC Med 2013; 11:217. [PMID: 24107194 PMCID: PMC3851549 DOI: 10.1186/1741-7015-11-217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 09/13/2013] [Indexed: 01/31/2023] Open
Abstract
The development of cardiovascular dysfunction and shock in patients with invasive Bacillus anthracis infection has a particularly poor prognosis. Growing evidence indicates that several bacterial components likely play important pathogenic roles in this injury. As with other pathogenic Gram-positive bacteria, the B. anthracis cell wall and its peptidoglycan constituent produce a robust inflammatory response with its attendant tissue injury, disseminated intravascular coagulation and shock. However, B. anthracis also produces lethal and edema toxins that both contribute to shock. Growing evidence suggests that lethal toxin, a metalloprotease, can interfere with endothelial barrier function as well as produce myocardial dysfunction. Edema toxin has potent adenyl cyclase activity and may alter endothelial function, as well as produce direct arterial and venous relaxation. Furthermore, both toxins can weaken host defense and promote infection. Finally, B. anthracis produces non-toxin metalloproteases which new studies show can contribute to tissue injury, coagulopathy and shock. In the future, an understanding of the individual pathogenic effects of these different components and their interactions will be important for improving the management of B. anthracis infection and shock.
Collapse
Affiliation(s)
- Kenneth E Remy
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|