1
|
Krantz BA. Anthrax Toxin: Model System for Studying Protein Translocation. J Mol Biol 2024; 436:168521. [PMID: 38458604 DOI: 10.1016/j.jmb.2024.168521] [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: 11/17/2023] [Revised: 02/08/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Dedicated translocase channels are nanomachines that often, but not always, unfold and translocate proteins through narrow pores across the membrane. Generally, these molecular machines utilize external sources of free energy to drive these reactions, since folded proteins are thermodynamically stable, and once unfolded they contain immense diffusive configurational entropy. To catalyze unfolding and translocate the unfolded state at appreciable timescales, translocase channels often utilize analogous peptide-clamp active sites. Here we describe how anthrax toxin has been used as a biophysical model system to study protein translocation. The tripartite bacterial toxin is composed of an oligomeric translocase channel, protective antigen (PA), and two enzymes, edema factor (EF) and lethal factor (LF), which are translocated by PA into mammalian host cells. Unfolding and translocation are powered by the endosomal proton gradient and are catalyzed by three peptide-clamp sites in the PA channel: the α clamp, the ϕ clamp, and the charge clamp. These clamp sites interact nonspecifically with the chemically complex translocating chain, serve to minimize unfolded state configurational entropy, and work cooperatively to promote translocation. Two models of proton gradient driven translocation have been proposed: (i) an extended-chain Brownian ratchet mechanism and (ii) a proton-driven helix-compression mechanism. These models are not mutually exclusive; instead the extended-chain Brownian ratchet likely operates on β-sheet sequences and the helix-compression mechanism likely operates on α-helical sequences. Finally, we compare and contrast anthrax toxin with other related and unrelated translocase channels.
Collapse
Affiliation(s)
- Bryan A Krantz
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|
2
|
Hilliard JJ, Jakielaszek C, Mannino F, Hossain M, Qian L, Fishman C, Demons S, Hershfield J, Soffler C, Russo R, Henning L, Novak J, O'Dwyer K. Efficacy of therapeutically administered gepotidacin in a rabbit model of inhalational anthrax. Antimicrob Agents Chemother 2024; 68:e0149723. [PMID: 38358266 PMCID: PMC10916377 DOI: 10.1128/aac.01497-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/17/2024] [Indexed: 02/16/2024] Open
Abstract
Bacillus anthracis is a Gram-positive Centers for Disease Control and Prevention category "A" biothreat pathogen. Without early treatment, inhalation of anthrax spores with progression to inhalational anthrax disease is associated with high fatality rates. Gepotidacin is a novel first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action and is being evaluated for use against biothreat and conventional pathogens. Gepotidacin selectively inhibits bacterial DNA replication via a unique binding mode and has in vitro activity against a collection of B. anthracis isolates including antibacterial-resistant strains, with the MIC90 ranging from 0.5 to 1 µg/mL. In vivo activity of gepotidacin was also evaluated in the New Zealand White rabbit model of inhalational anthrax. The primary endpoint was survival, with survival duration and bacterial clearance as secondary endpoints. The trigger for treatment was the presence of anthrax protective antigen in serum. New Zealand White rabbits were dosed intravenously for 5 days with saline or gepotidacin at 114 mg/kg/d to simulate a dosing regimen of 1,000 mg intravenous (i.v.) three times a day (TID) in humans. Gepotidacin provided a survival benefit compared to saline control, with 91% survival (P-value: 0.0001). All control animals succumbed to anthrax and were found to be blood- and organ culture-positive for B. anthracis. The novel mode of action, in vitro microbiology, preclinical safety, and animal model efficacy data, which were generated in line with Food and Drug Administration Animal Rule, support gepotidacin as a potential treatment for anthrax in an emergency biothreat situation.
Collapse
Affiliation(s)
| | | | | | | | - Lian Qian
- GSK, Collegeville, Pennsylvania, USA
| | | | - Samandra Demons
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Jeremy Hershfield
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Carl Soffler
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Riccardo Russo
- Rutgers University School of Medicine, Newark, New Jersey, USA
| | - Lisa Henning
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | - Joseph Novak
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | | |
Collapse
|
3
|
Beliveau M, Rubets I, Bojan D, Hall C, Toth D, Kodihalli S, Kammanadiminti S. Animal-to-Human Dose Translation of ANTHRASIL for Treatment of Inhalational Anthrax in Healthy Adults, Obese Adults, and Pediatric Subjects. Clin Pharmacol Ther 2024; 115:248-255. [PMID: 38082506 DOI: 10.1002/cpt.3097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/25/2023] [Indexed: 01/23/2024]
Abstract
Anthrax Immune Globulin Intravenous (AIGIV [ANTHRASIL]), was developed for the treatment of toxemia associated with inhalational anthrax. It is a plasma product collected from individuals vaccinated with anthrax vaccine and contains antitoxin IgG antibodies against Bacillus anthracis protective antigen. A pharmacokinetic (PK) and exposure-response model was constructed to assess the PKs of AIGIV in anthrax-free and anthrax-exposed rabbits, non-human primates and anthrax-free humans, as well as the relationship between AIGIV exposure and survival from anthrax, based on available preclinical/clinical studies. The potential effect of anthrax on the PKs of AIGIV was evaluated and estimates of survival odds following administration of AIGIV protective doses with and without antibiotic co-treatment were established. As the developed PK model can simulate exposure of AIGIV in any species for any dosing scenario, the relationship between the predicted area under the concentration curve of AIGIV in humans and the probability of survival observed in preclinical studies was explored. Based on the simulation results, the intravenous administration of 420 U (units of potency as measured by validated Toxin Neutralization Assay) of AIGIV is expected to result in a > 80% probability of survival in more than 90% of the human population. Additional simulations suggest that exposure levels were similar in healthy and obese humans, and exposure in pediatrics is expected to be up to approximately seven-fold higher than in healthy adults, allowing for doses in pediatric populations that ranged from one to seven vials. Overall, the optimal human dose was justified based on the PK/pharmacodynamic (PD) properties of AIGIV in animals and model-based translation of PK/PD to predict human exposure and efficacy.
Collapse
Affiliation(s)
- Martin Beliveau
- Integrated Drug Development, Certara, Montreal, Quebec, Canada
| | - Igor Rubets
- Integrated Drug Development, Certara, Montreal, Quebec, Canada
| | - Drobic Bojan
- Emergent BioSolutions Inc., Winnipeg, Manitoba, Canada
| | | | - Derek Toth
- Emergent BioSolutions Inc., Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
4
|
Tessier E, Cheutin L, Garnier A, Vigne C, Tournier JN, Rougeaux C. Early Circulating Edema Factor in Inhalational Anthrax Infection: Does It Matter? Microorganisms 2024; 12:308. [PMID: 38399712 PMCID: PMC10891819 DOI: 10.3390/microorganisms12020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Anthrax toxins are critical virulence factors of Bacillus anthracis and Bacillus cereus strains that cause anthrax-like disease, composed of a common binding factor, the protective antigen (PA), and two enzymatic proteins, lethal factor (LF) and edema factor (EF). While PA is required for endocytosis and activity of EF and LF, several studies showed that these enzymatic factors disseminate within the body in the absence of PA after intranasal infection. In an effort to understand the impact of EF in the absence of PA, we used a fluorescent EF chimera to facilitate the study of endocytosis in different cell lines. Unexpectedly, EF was found inside cells in the absence of PA and showed a pole-dependent endocytosis. However, looking at enzymatic activity, PA was still required for EF to induce an increase in intracellular cAMP levels. Interestingly, the sequential delivery of EF and then PA rescued the rise in cAMP levels, indicating that PA and EF may functionally associate during intracellular trafficking, as well as it did at the cell surface. Our data shed new light on EF trafficking and the potential location of PA and EF association for optimal cytosolic delivery.
Collapse
Affiliation(s)
- Emilie Tessier
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Laurence Cheutin
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Annabelle Garnier
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Clarisse Vigne
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Jean-Nicolas Tournier
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
- Institut Pasteur, 75015 Paris, France
| | - Clémence Rougeaux
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| |
Collapse
|
5
|
Goossens PL. Bacillus anthracis, "la maladie du charbon", Toxins, and Institut Pasteur. Toxins (Basel) 2024; 16:66. [PMID: 38393144 PMCID: PMC10891547 DOI: 10.3390/toxins16020066] [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: 11/08/2023] [Revised: 12/25/2023] [Accepted: 12/30/2023] [Indexed: 02/25/2024] Open
Abstract
Institut Pasteur and Bacillus anthracis have enjoyed a relationship lasting almost 120 years, starting from its foundation and the pioneering work of Louis Pasteur in the nascent fields of microbiology and vaccination, and blooming after 1986 following the molecular biology/genetic revolution. This contribution will give a historical overview of these two research eras, taking advantage of the archives conserved at Institut Pasteur. The first era mainly focused on the production, characterisation, surveillance and improvement of veterinary anthrax vaccines; the concepts and technologies with which to reach a deep understanding of this research field were not yet available. The second period saw a new era of B. anthracis research at Institut Pasteur, with the anthrax laboratory developing a multi-disciplinary approach, ranging from structural analysis, biochemistry, genetic expression, and regulation to bacterial-host cell interactions, in vivo pathogenicity, and therapy development; this led to the comprehensive unravelling of many facets of this toxi-infection. B. anthracis may exemplify some general points on how science is performed in a given society at a given time and how a scientific research domain evolves. A striking illustration can be seen in the additive layers of regulations that were implemented from the beginning of the 21st century and their impact on B. anthracis research. B. anthracis and anthrax are complex systems that raise many valuable questions regarding basic research. One may hope that B. anthracis research will be re-initiated under favourable circumstances later at Institut Pasteur.
Collapse
|
6
|
Kammanadiminti S, Comer J, Meister G, Carnelley T, Toth D, Kodihalli S. Efficacy of ANTHRASIL (Anthrax Immune Globulin Intravenous (Human)) in rabbit and nonhuman primate models of inhalational anthrax: Data supporting approval under animal rule. PLoS One 2023; 18:e0283164. [PMID: 36930692 PMCID: PMC10022752 DOI: 10.1371/journal.pone.0283164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
To meet the requirements of the Animal Rule, the efficacy of monotherapy with ANTHRASIL® (Anthrax Immune Globulin Intravenous (Human)) for inhalational anthrax was evaluated in blinded studies using rabbit and nonhuman primate models. Animals in both studies were randomized to treatment groups exposed to ~ 200 LD50 Bacillus anthracis (Ames strain) spores by the aerosol route to induce inhalational anthrax. Rabbits (N = 50/group) were treated with either 15 U/kg ANTHRASIL or a volume-matching dose of IGIV after disease onset as determined by the detection of bacterial toxin in the blood. At the end of the study, survival rates were 2% (1 of 48) in the IGIV control group, and 26% (13 of 50) in the ANTHRASIL-treated group (p = 0.0009). Similarly, ANTHRASIL was effective in cynomolgus monkeys (N = 16/group) when administered therapeutically after the onset of toxemia, with 6% survival in the IGIV control and a dose-related increase in survival of 36%, 43%, and 70% with 7.5, 15 or 30 U/kg doses of ANTHRASIL, respectively. These studies formed the basis for approval of ANTHRASIL by FDA under the Animal Rule.
Collapse
Affiliation(s)
| | - Jason Comer
- Battelle Biomedical Research Center, Columbus, Ohio, United States of America
| | - Gabriel Meister
- Battelle Biomedical Research Center, Columbus, Ohio, United States of America
| | - Trevor Carnelley
- Emergent BioSolutions Canada (Previously Cangene Corporation), Winnipeg, MB, Canada
| | - Derek Toth
- Emergent BioSolutions Canada (Previously Cangene Corporation), Winnipeg, MB, Canada
| | - Shantha Kodihalli
- Emergent BioSolutions Canada (Previously Cangene Corporation), Winnipeg, MB, Canada
- * E-mail:
| |
Collapse
|
7
|
Abstract
The biggest challenge to immune control of HIV infection is the rapid within-host viral evolution, which allows selection of viral variants that escape from T cell and antibody recognition. Thus, it is impossible to clear HIV infection without targeting "immutable" components of the virus. Unlike the adaptive immune system that recognizes cognate epitopes, the CARD8 inflammasome senses the essential enzymatic activity of the HIV-1 protease, which is immutable for the virus. Hence, all subtypes of HIV clinical isolates can be recognized by CARD8. In HIV-infected cells, the viral protease is expressed as a subunit of the viral Gag-Pol polyprotein and remains functionally inactive prior to viral budding. A class of anti-HIV drugs, the non-nucleoside reverse transcriptase inhibitors (NNRTIs), can promote Gag-pol dimerization and subsequent premature intracellular activation of the viral protease. NNRTI treatment triggers CARD8 inflammasome activation, which leads to pyroptosis of HIV-infected CD4+ T cells and macrophages. Targeting the CARD8 inflammasome can be a potent and broadly effective strategy for HIV eradication.
Collapse
Affiliation(s)
- Kolin M Clark
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Priya Pal
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Josh G Kim
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Qiankun Wang
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Liang Shan
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, United States.
| |
Collapse
|
8
|
Ochai SO, Crafford JE, Hassim A, Byaruhanga C, Huang YH, Hartmann A, Dekker EH, van Schalkwyk OL, Kamath PL, Turner WC, van Heerden H. Immunological Evidence of Variation in Exposure and Immune Response to Bacillus anthracis in Herbivores of Kruger and Etosha National Parks. Front Immunol 2022; 13:814031. [PMID: 35237267 PMCID: PMC8882864 DOI: 10.3389/fimmu.2022.814031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure and immunity to generalist pathogens differ among host species and vary across spatial scales. Anthrax, caused by a multi-host bacterial pathogen, Bacillus anthracis, is enzootic in Kruger National Park (KNP), South Africa and Etosha National Park (ENP), Namibia. These parks share many of the same potential host species, yet the main anthrax host in one (greater kudu (Tragelaphus strepsiceros) in KNP and plains zebra (Equus quagga) in ENP) is only a minor host in the other. We investigated species and spatial patterns in anthrax mortalities, B. anthracis exposure, and the ability to neutralize the anthrax lethal toxin to determine if observed host mortality differences between locations could be attributed to population-level variation in pathogen exposure and/or immune response. Using serum collected from zebra and kudu in high and low incidence areas of each park (18- 20 samples/species/area), we estimated pathogen exposure from anti-protective antigen (PA) antibody response using enzyme-linked immunosorbent assay (ELISA) and lethal toxin neutralization with a toxin neutralization assay (TNA). Serological evidence of pathogen exposure followed mortality patterns within each system (kudus: 95% positive in KNP versus 40% in ENP; zebras: 83% positive in ENP versus 63% in KNP). Animals in the high-incidence area of KNP had higher anti-PA responses than those in the low-incidence area, but there were no significant differences in exposure by area within ENP. Toxin neutralizing ability was higher for host populations with lower exposure prevalence, i.e., higher in ENP kudus and KNP zebras than their conspecifics in the other park. These results indicate that host species differ in their exposure to and adaptive immunity against B. anthracis in the two parks. These patterns may be due to environmental differences such as vegetation, rainfall patterns, landscape or forage availability between these systems and their interplay with host behavior (foraging or other risky behaviors), resulting in differences in exposure frequency and dose, and hence immune response.
Collapse
Affiliation(s)
- Sunday O. Ochai
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Jan E. Crafford
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Ayesha Hassim
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Charles Byaruhanga
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Yen-Hua Huang
- Wisconsin Cooperative Wildlife Research Unit, Department of Forest and Wildlife Ecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Axel Hartmann
- Etosha Ecological Institute, Ministry of Environment, Forestry and Tourism, Okaukuejo, Namibia
| | - Edgar H. Dekker
- Office of the State Veterinarian, Department of Agriculture, Forestry and Fisheries, Government of South Africa, Skukuza, South Africa
| | - O. Louis van Schalkwyk
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
- Office of the State Veterinarian, Department of Agriculture, Forestry and Fisheries, Government of South Africa, Skukuza, South Africa
- Department of Migration, Max Planck Institute of Animal Behavior, Radolfzell, Germany
| | - Pauline L. Kamath
- School of Food and Agriculture, University of Maine, Orono, ME, United States
| | - Wendy C. Turner
- U.S. Geological Survey, Wisconsin Cooperative Wildlife Research Unit, Department of Forest and Wildlife Ecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Henriette van Heerden
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
9
|
Hirschenberger M, Stadler N, Fellermann M, Sparrer KMJ, Kirchhoff F, Barth H, Papatheodorou P. CRISPA: A Non-viral, Transient Cas9 Delivery System Based on Reengineered Anthrax Toxin. Front Pharmacol 2021; 12:770283. [PMID: 34733166 PMCID: PMC8558532 DOI: 10.3389/fphar.2021.770283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
Translating the CRISPR/Cas9 genome editing technology into clinics is still hampered by rather unspecific, unsafe and/or inconvenient approaches for the delivery of its main components - the Cas9 endonuclease and a guide RNA - into cells. Here, we describe the development of a novel transient and non-viral Cas9 delivery strategy based on the translocation machinery of the Bacillus anthracis anthrax toxin, PA (protective antigen). We show that Cas9 variants fused to the N-terminus of the lethal factor or to a hexahistidine tag are shuttled through channels formed by PA into the cytosol of human cells. As proof-of-principle, we applied our new approach, denoted as CRISPA, to knock out lipolysis-stimulated lipoprotein receptor (LSR) in the human colon cancer cell line HCT116 and green-fluorescent protein (GFP) in human embryonic kidney 293T cells stably expressing GFP. Notably, we confirmed that the transporter PA can be adapted to recognize specific host cell-surface receptor proteins and may be optimized for cell type-selective delivery of Cas9. Altogether, CRISPA provides a novel, transient and non-viral way to deliver Cas9 into specific cells. Thus, this system is an additional step towards safe translation of the CRISPR/Cas9 technology into clinics.
Collapse
Affiliation(s)
- Maximilian Hirschenberger
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany.,Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Nicole Stadler
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Maximilian Fellermann
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | | |
Collapse
|
10
|
Complement C5 inhibition protects against hemolytic anemia and acute kidney injury in anthrax peptidoglycan-induced sepsis in baboons. Proc Natl Acad Sci U S A 2021; 118:2104347118. [PMID: 34507997 DOI: 10.1073/pnas.2104347118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 01/20/2023] Open
Abstract
Late-stage anthrax infections are characterized by dysregulated immune responses and hematogenous spread of Bacillus anthracis, leading to extreme bacteremia, sepsis, multiple organ failure, and, ultimately, death. Despite the bacterium being nonhemolytic, some fulminant anthrax patients develop a secondary atypical hemolytic uremic syndrome (aHUS) through unknown mechanisms. We recapitulated the pathology in baboons challenged with cell wall peptidoglycan (PGN), a polymeric, pathogen-associated molecular pattern responsible for the hemostatic dysregulation in anthrax sepsis. Similar to aHUS anthrax patients, PGN induces an initial hematocrit elevation followed by progressive hemolytic anemia and associated renal failure. Etiologically, PGN induces erythrolysis through direct excessive activation of all three complement pathways. Blunting terminal complement activation with a C5 neutralizing peptide prevented the progressive deposition of membrane attack complexes on red blood cells (RBC) and subsequent intravascular hemolysis, heme cytotoxicity, and acute kidney injury. Importantly, C5 neutralization did not prevent immune recognition of PGN and shifted the systemic inflammatory responses, consistent with improved survival in sepsis. Whereas PGN-induced hemostatic dysregulation was unchanged, C5 inhibition augmented fibrinolysis and improved the thromboischemic resolution. Overall, our study identifies PGN-driven complement activation as the pathologic mechanism underlying hemolytic anemia in anthrax and likely other gram-positive infections in which PGN is abundantly represented. Neutralization of terminal complement reactions reduces the hemolytic uremic pathology induced by PGN and could alleviate heme cytotoxicity and its associated kidney failure in gram-positive infections.
Collapse
|
11
|
Bacillus anthracis chain length, a virulence determinant, is regulated by membrane localized serine/threonine protein kinase PrkC. J Bacteriol 2021; 203:JB.00582-20. [PMID: 33753466 PMCID: PMC8117516 DOI: 10.1128/jb.00582-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anthrax is a zoonotic disease caused by Bacillus anthracis, a spore-forming pathogen that displays a chaining phenotype. It has been reported that the chaining phenotype acts as a virulence factor in B. anthracis In this study, we identify a serine/threonine protein kinase of B. anthracis, PrkC, the only kinase localized at the bacteria-host interface, as a determinant of B. anthracis chain length. In vitro, prkC disruption strain (BAS ΔprkC) grew as shorter chains throughout the bacterial growth cycle. A comparative analysis between the parent strain and BAS ΔprkC indicated that the levels of proteins, BslO and Sap, associated with the regulation of the bacterial chain length, were upregulated in BAS ΔprkC BslO is a septal murein hydrolase that catalyzes daughter cell separation and Sap is an S-layer structural protein required for the septal localization of BslO. PrkC disruption also has a significant effect on bacterial growth, cell wall thickness, and septa formation. Upregulation of ftsZ in BAS ΔprkC was also observed. Altogether, our results indicate that PrkC is required for maintaining optimum growth, cell wall homeostasis and most importantly - for the maintenance of the chaining phenotype.IMPORTANCEChaining phenotype acts as a virulence factor in Bacillus anthracis This is the first study that identifies a 'signal transduction protein' with an ability to regulate the chaining phenotype in Bacillus anthracis We show that the disruption of the lone surface-localized serine/threonine protein kinase, PrkC, leads to the shortening of the bacterial chains. We report upregulation of the de-chaining proteins in the PrkC disruption strain. Apart from this, we also report for the first time that PrkC disruption results in an attenuated cell growth, a decrease in the cell wall thickness and aberrant cell septa formation during the logarithmic phase of growth - a growth phase where PrkC is expressed maximally.
Collapse
|
12
|
Sittner A, Bar-David E, Glinert I, Ben-Shmuel A, Schlomovitz J, Levy H, Weiss S. Role of acpA and acpB in Bacillus anthracis capsule accumulation and toxin independent pathogenicity in rabbits. Microb Pathog 2021; 155:104904. [PMID: 33930422 DOI: 10.1016/j.micpath.2021.104904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
The poly- δ- d-glutamic acid capsule of Bacillus anthracis plays a major role in this bacterium pathogenicity. Capsule synthesis relies on a 5 gene operon; capB, C, A, D and E that are regulated by acpA and acpB, that respond to the major virulence regulator - atxA. We took a genetic approach to examine the involvement of acpA and acpB in capsule production in vitro and on B. anthracis virulence in vivo. To complement the effect of the mutations on capsule accumulation in vitro, we applied our toxin independent systemic infection method to study their effects in vivo. We found that though the roles of acpA and axpB are redundant in vitro, deleting acpA had a significant effect on pathogenicity, mainly on the time to death. As expected, deletion of both acpA and acpB resulted in loss of capsule accumulation in vitro and full attenuation in vivo, indicating that capsule production depends exclusively on acpA/B regulation. To identify additional effects of acpA and acpB on pathogenicity via non-capsule related virulence pathways, we bypassed acpA/B regulation by inserting the pagA promotor upstream to the cap operon, diverting regulation directly to atxA. This resulted in restoration of capsule accumulation in vitro and virulence (in intravenous or subcutaneous inoculation) in vivo. To test for additional pXO2-based genes involved in capsule production, we cloned the pagAprom-capA-E into the chromosome of VollumΔpXO2, which restored capsule accumulation. These results indicate that of the pXO2 genes, only capA-E and acpA are required for capsule production.
Collapse
Affiliation(s)
- Assa Sittner
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel
| | - Elad Bar-David
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel
| | - Itai Glinert
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel
| | - Amir Ben-Shmuel
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel
| | - Josef Schlomovitz
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel
| | - Haim Levy
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel
| |
Collapse
|
13
|
Abstract
Anthrax toxin is a major virulence factor of Bacillus anthracis, a Gram-positive bacterium which can form highly stable spores that are the causative agents of the disease, anthrax. While chiefly a disease of livestock, spores can be "weaponized" as a bio-terrorist agent, and can be deadly if not recognized and treated early with antibiotics. The intracellular pathways affected by the enzymes are broadly understood and are not discussed here. This chapter focuses on what is known about the assembly of secreted toxins on the host cell surface and how the toxin is delivered into the cytosol. The central component is the "Protective Antigen", which self-oligomerizes and forms complexes with its pay-load, either Lethal Factor or Edema Factor. It binds a host receptor, CMG2, or a close relative, triggering receptor-mediated endocytosis, and forms a remarkably elegant yet powerful machine that delivers toxic enzymes into the cytosol, powered only by the pH gradient across the membrane. We now have atomic structures of most of the starting, intermediate and final assemblies in the infectious process. Together with a major body of biophysical, mutational and biochemical work, these studies reveal a remarkable story of both how toxin assembly is choreographed in time and space.
Collapse
|
14
|
Mukarati NL, Ndumnego OC, Ochai SO, Jauro S, Loveridge A, van Heerden H, Matope G, Caron A, Hanyire TG, de Garine-Wichatitsky M, Pfukenyi DM. A serological survey of Bacillus anthracis reveals widespread exposure to the pathogen in free-range and captive lions in Zimbabwe. Transbound Emerg Dis 2020; 68:1676-1684. [PMID: 32964687 DOI: 10.1111/tbed.13842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 11/29/2022]
Abstract
Numerous unknown factors influence anthrax epidemiology in multi-host systems, especially at wildlife/livestock/human interfaces. Serology tests for anti-anthrax antibodies in carnivores are useful tools in identifying the presence or absence of Bacillus anthracis in a range. These were employed to ascertain whether the disease pattern followed the recognized high- and low-risk anthrax zonation in Zimbabwe and also to establish whether anthrax was absent from Hwange National Park in which there have been no reported outbreaks. African lions (Panthera leo) (n = 114) drawn from free-range protected areas and captive game parks located in recognized high- and low-risk zones across Zimbabwe were tested for antibodies to anthrax PA antigen using the ELISA immunoassay. A random selection of 27 lion sera samples comprising 17 seropositive and 10 seronegative sera was further tested in the species-independent toxin neutralization assay (TNA) in order to validate the former as a surveillance tool for anthrax in African lions. Using the ELISA-PA immunoassay, 21.9% (25/114) of the lions tested positive for antibodies to anthrax. Seropositivity was recorded in all study areas, and there was no significant difference (p = .852) in seropositivity between lions in high- and low-risk anthrax zones. Also, there was no significant difference (McNemar's chi-square test = 0.9, p = .343) in the proportion of lions testing positive to anti-PA anthrax antibodies on ELISA-PA immunoassay compared with the TNA, with fair agreement between the two tests [kappa (K) statistic = 0.30; 0.08 < K<0.613]. Results of this study indicate that anthrax could be more widespread than 42 currently realized in Zimbabwe, and present in recognized high- and low-risk zones, including 43 where it has not been reported in over 20 years such as Hwange National Park. This is also the 44 first report documenting the presence of anthrax lethal toxin-neutralizing antibodies in naturally 45 infected carnivores, further confirming exposure to B. anthracis. The research results point to a 46 need for revisiting the currently recognized anthrax risk zones in Zimbabwe. This should be based 47 on improved surveillance of the disease in both wild and domestic animals for better understanding and control of the disease.
Collapse
Affiliation(s)
- Norman L Mukarati
- Faculty of Veterinary Science, University of Zimbabwe, Harare, Zimbabwe
| | - Okechukwu C Ndumnego
- Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa.,Moredun Scientific, Edinburgh, UK
| | - Sunday O Ochai
- Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Solomon Jauro
- Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | | | | | - Gift Matope
- Faculty of Veterinary Science, University of Zimbabwe, Harare, Zimbabwe
| | - Alexandre Caron
- ASTRE, Univ. de Montpellier, CIRAD, INRA, Montpellier, France.,CIRAD, UMR ASTRE, RP-PCP, Maputo, Mozambique.,Faculdade de Veterinária, Universidade Eduardo Mondlane, Maputo, Mozambique
| | | | - Michel de Garine-Wichatitsky
- ASTRE, Univ. de Montpellier, CIRAD, INRA, Montpellier, France.,CIRAD, UMR ASTRE, Bangkok, Thailand.,Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Davies M Pfukenyi
- Faculty of Veterinary Science, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
15
|
Greaney AJ, Portley MK, O’Mard D, Crown D, Maier NK, Mendenhall MA, Mayer-Barber KD, Leppla SH, Moayeri M. Frontline Science: Anthrax lethal toxin-induced, NLRP1-mediated IL-1β release is a neutrophil and PAD4-dependent event. J Leukoc Biol 2020; 108:773-786. [PMID: 32421904 PMCID: PMC11062252 DOI: 10.1002/jlb.4hi0320-028r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/17/2020] [Accepted: 03/28/2020] [Indexed: 12/21/2022] Open
Abstract
Anthrax lethal toxin (LT) is a protease that activates the NLRP1b inflammasome sensor in certain rodent strains. Unlike better-studied sensors, relatively little is known about the priming requirements for NLRP1b. In this study, we investigate the rapid and striking priming-independent LT-induced release of IL-1β in mice within hours of toxin challenge. We find IL-1β release to be a NLRP1b- and caspase-1-dependent, NLRP3 and caspase-11-independent event that requires both neutrophils and peptidyl arginine deiminiase-4 (PAD4) activity. The simultaneous LT-induced IL-18 response is neutrophil-independent. Bone marrow reconstitution experiments in mice show toxin-induced IL-1β originates from hematopoietic cells. LT treatment of neutrophils in vitro did not induce IL-1β, neutrophil extracellular traps (NETs), or pyroptosis. Although platelets interact closely with neutrophils and are also a potential source of IL-1β, they were unable to bind or endocytose LT and did not secrete IL-1β in response to the toxin. LT-treated mice had higher levels of cell-free DNA and HMGB1 in circulation than PBS-treated controls, and treatment of mice with recombinant DNase reduced the neutrophil- and NLRP1-dependent IL-1β release. DNA sensor AIM2 deficiency, however, did not impact IL-1β release. These data, in combination with the findings on PAD4, suggest a possible role for in vivo NETs or cell-free DNA in cytokine induction in response to LT challenge. Our findings suggest a complex interaction of events and/or mediators in LT-treated mice with the neutrophil as a central player in induction of a profound and rapid inflammatory response to toxin.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/physiology
- Animals
- Anthrax/immunology
- Antigens, Bacterial/pharmacology
- Antigens, Bacterial/toxicity
- Apoptosis Regulatory Proteins/deficiency
- Apoptosis Regulatory Proteins/physiology
- Bacillus anthracis/pathogenicity
- Bacillus anthracis/physiology
- Bacterial Toxins/pharmacology
- Bacterial Toxins/toxicity
- Extracellular Traps/physiology
- Inflammasomes/physiology
- Interleukin-1beta/metabolism
- Mice
- Mice, 129 Strain
- Mice, Congenic
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Monocytes/drug effects
- Monocytes/physiology
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- Neutrophils/drug effects
- Neutrophils/metabolism
- Protein-Arginine Deiminase Type 4/deficiency
- Protein-Arginine Deiminase Type 4/physiology
- Pyroptosis/drug effects
- Radiation Chimera
- Species Specificity
- Spores, Bacterial
Collapse
Affiliation(s)
- Allison J. Greaney
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Makayla K. Portley
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Danielle O’Mard
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Devorah Crown
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nolan K. Maier
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Megan A. Mendenhall
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen H. Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mahtab Moayeri
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Baldwin VM. You Can't B. cereus - A Review of Bacillus cereus Strains That Cause Anthrax-Like Disease. Front Microbiol 2020; 11:1731. [PMID: 32973690 PMCID: PMC7468541 DOI: 10.3389/fmicb.2020.01731] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Emerging strains of Bacillus cereus, traditionally considered a self-limiting foodborne pathogen, have been associated with anthrax-like disease in mammals, including humans. The strains have emerged by divergent evolution and, as exchange of genetic material in the Bacillus genus occurs naturally, it is possible that further isolates will be identified in the future. The strains vary in their genotypes and phenotypes, combining traits of both B. cereus and B. anthracis species. Cases of anthrax-like disease associated with these strains result in similar symptoms and mortality rates as those caused by B. anthracis. The strains are susceptible to frontline antibiotics used in the treatment of anthrax and existing vaccines provide protection in animal models. The emergence of these strains has reignited the debate surrounding classification of the B. cereus sensu lato group and serves as a reminder that the field of medical microbiology is constantly changing and remains an important and ongoing area of research.
Collapse
|
17
|
Popescu NI, Keshari RS, Cochran J, Coggeshall KM, Lupu F. C3 Opsonization of Anthrax Bacterium and Peptidoglycan Supports Recognition and Activation of Neutrophils. Microorganisms 2020; 8:E1039. [PMID: 32668703 PMCID: PMC7409185 DOI: 10.3390/microorganisms8071039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/08/2020] [Accepted: 07/11/2020] [Indexed: 12/01/2022] Open
Abstract
Neutrophils are the most abundant innate cell population and a key immune player against invading pathogens. Neutrophils can kill both bacterium and spores of Bacillus anthracis, the causative anthrax pathogen. Unlike interactions with professional phagocytes, the molecular recognition of anthrax by neutrophils is largely unknown. In this study, we investigated the role of complement C3 deposition on anthrax particles for neutrophil recognition of bacterium and/or its cell wall peptidoglycan, an abundant pathogen-associated molecular pattern that supports anthrax sepsis. C3 opsonization and recognition by complement receptors accounted for 70-80% of the affinity interactions between neutrophils and anthrax particles at subphysiologic temperatures. In contrast, C3 supported up to 50% of the anthrax particle ingestion under thermophysiologic conditions. Opsonin-dependent low affinity interactions and, to a lower extent, opsonin-independent mechanisms, provide alternative entry routes. Similarly, C3 supported 58% of peptidoglycan-induced degranulation and, to a lower extent, 23% of bacterium-induced degranulation. Interestingly, an opsonin independent mechanism mediated by complement C5, likely through C5a anaphylatoxin, primes azurophilic granules in response to anthrax particles. Overall, we show that C3 deposition supports anthrax recognition by neutrophils but is dispensable for pathogen ingestion and neutrophil degranulation, highlighting immune recognition redundancies that minimize the risk of pathogen evasion.
Collapse
Affiliation(s)
- Narcis I. Popescu
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.C.); (K.M.C.)
| | - Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA;
| | - Jackie Cochran
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.C.); (K.M.C.)
| | - K. Mark Coggeshall
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.C.); (K.M.C.)
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA;
| |
Collapse
|
18
|
FINKE ERNSTJÜRGEN, BEYER WOLFGANG, LODERSTÄDT ULRIKE, FRICKMANN HAGEN. Review: The risk of contracting anthrax from spore-contaminated soil - A military medical perspective. Eur J Microbiol Immunol (Bp) 2020; 10:29-63. [PMID: 32590343 PMCID: PMC7391381 DOI: 10.1556/1886.2020.00008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/10/2020] [Indexed: 12/31/2022] Open
Abstract
Anthrax is an infectious disease of relevance for military forces. Although spores of Bacillus anthracis obiquitously occur in soil, reports on soil-borne transmission to humans are scarce. In this narrative review, the potential of soil-borne transmission of anthrax to humans is discussed based on pathogen-specific characteristics and reports on anthrax in the course of several centuries of warfare. In theory, anthrax foci can pose a potential risk of infection to animals and humans if sufficient amounts of virulent spores are present in the soil even after an extended period of time. In praxis, however, transmissions are usually due to contacts with animal products and reported events of soil-based transmissions are scarce. In the history of warfare, even in the trenches of World War I, reported anthrax cases due to soil-contaminated wounds are virtually absent. Both the perspectives and the experience of the Western hemisphere and of former Soviet Republics are presented. Based on the accessible data as provided in the review, the transmission risk of anthrax by infections of wounds due to spore-contaminated soil is considered as very low under the most circumstance. Active historic anthrax foci may, however, still pose a risk to the health of deployed soldiers.
Collapse
Affiliation(s)
| | - WOLFGANG BEYER
- Department of Infectiology and Animal Hygiene, University of Hohenheim, Institute of Animal Science, Stuttgart, Germany
| | - ULRIKE LODERSTÄDT
- Diagnostic Department, Bernhard-Nocht-Institute for Tropical Medicine Hamburg, Hamburg, Germany
| | - HAGEN FRICKMANN
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, Hamburg, Germany
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
19
|
Savransky V, Ionin B, Reece J. Current Status and Trends in Prophylaxis and Management of Anthrax Disease. Pathogens 2020; 9:E370. [PMID: 32408493 PMCID: PMC7281134 DOI: 10.3390/pathogens9050370] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/30/2022] Open
Abstract
Bacillus anthracis has been identified as a potential military and bioterror agent as it is relatively simple to produce, with spores that are highly resilient to degradation in the environment and easily dispersed. These characteristics are important in describing how anthrax could be used as a weapon, but they are also important in understanding and determining appropriate prevention and treatment of anthrax disease. Today, anthrax disease is primarily enzootic and found mostly in the developing world, where it is still associated with considerable mortality and morbidity in humans and livestock. This review article describes the spectrum of disease caused by anthrax and the various prevention and treatment options. Specifically we discuss the following; (1) clinical manifestations of anthrax disease (cutaneous, gastrointestinal, inhalational and intravenous-associated); (2) immunology of the disease; (3) an overview of animal models used in research; (4) the current World Health Organization and U.S. Government guidelines for investigation, management, and prophylaxis; (5) unique regulatory approaches to licensure and approval of anthrax medical countermeasures; (6) the history of vaccination and pre-exposure prophylaxis; (7) post-exposure prophylaxis and disease management; (8) treatment of symptomatic disease through the use of antibiotics and hyperimmune or monoclonal antibody-based antitoxin therapies; and (9) the current landscape of next-generation product candidates under development.
Collapse
Affiliation(s)
- Vladimir Savransky
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA; (B.I.); (J.R.)
| | | | | |
Collapse
|
20
|
Kondakova OA, Nikitin NA, Evtushenko EA, Ryabchevskaya EM, Atabekov JG, Karpova OV. Vaccines against anthrax based on recombinant protective antigen: problems and solutions. Expert Rev Vaccines 2019; 18:813-828. [PMID: 31298973 DOI: 10.1080/14760584.2019.1643242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Introduction: Anthrax is a dangerous bio-terror agent because Bacillus anthracis spores are highly resilient and can be easily aerosolized and disseminated. There is a threat of deliberate use of anthrax spores aerosol that could lead to serious fatal diseases outbreaks. Existing control measures against inhalation form of the disease are limited. All of this has provided an impetus to the development of new generation vaccines. Areas сovered: This review is devoted to challenges and achievements in the design of vaccines based on the anthrax recombinant protective antigen (rPA). Scientific databases have been searched, focusing on causes of PA instability and solutions to this problem, including new approaches of rPA expression, novel rPA-based vaccines formulations as well as the simultaneous usage of PA with other anthrax antigens. Expert opinion: PA is a central anthrax toxin component, playing a key role in the defense against encapsulated and unencapsulated strains. Subunit rPA-based vaccines have a good safety and protective profile. However, there are problems of PA instability that are greatly enhanced when using aluminum adjuvants. New adjuvant compositions, dry formulations and resistant to proteolysis and deamidation mutant PA forms can help to handle this issue. Devising a modern anthrax vaccine requires huge efforts.
Collapse
Affiliation(s)
- Olga A Kondakova
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Nikolai A Nikitin
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Ekaterina A Evtushenko
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Ekaterina M Ryabchevskaya
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Joseph G Atabekov
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Olga V Karpova
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| |
Collapse
|
21
|
Glinert I, Weiss S, Sittner A, Bar-David E, Ben-Shmuel A, Schlomovitz J, Kobiler D, Levy H. Infection with a Nonencapsulated Bacillus anthracis Strain in Rabbits-The Role of Bacterial Adhesion and the Potential for a Safe Live Attenuated Vaccine. Toxins (Basel) 2018; 10:toxins10120506. [PMID: 30513757 PMCID: PMC6316610 DOI: 10.3390/toxins10120506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/15/2018] [Accepted: 11/22/2018] [Indexed: 12/14/2022] Open
Abstract
Nonencapsulated (∆pXO2) Bacillus anthracis strains are commonly used as vaccines and for anthrax research, mainly in the mouse model. Previously, we demonstrated that the infection of rabbits, intranasally or subcutaneously, with the spores of a fully virulent strain results in the systemic dissemination of the bacteria, meningitis, and death, whereas ∆pXO2 strains are fully attenuated in this animal model. We used the intravenous inoculation of rabbits to study the pathogenicity of the ∆pXO2 strain infection. Bacteremia, brain bacterial burden, and pathology were used as criteria to compare the Vollum∆pXO2 disease to the wild type Vollum infection. To test the role of adhesion in the virulence of Vollum∆pXO2, we deleted the major adhesion protein BslA and tested the virulence and immunogenicity of this mutant. We found that 50% of the rabbits succumb to Vollum∆pXO2 strain following i.v. infection, a death that was accompanied with significant neurological symptoms. Pathology revealed severe brain infection coupled with an atypical massive bacterial growth into the parenchyma. Contrary to the Vollum strain, deletion of the bslA gene fully attenuated the ∆pXO2 strain. Though the Vollum∆pXO2 cannot serve as a model for B. anthracis pathogenicity in rabbits, deletion of the bslA gene prevents central nervous system (CNS) infections, possibly leading to the generation of a safer vaccine.
Collapse
Affiliation(s)
- Itai Glinert
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Assa Sittner
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Elad Bar-David
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Amir Ben-Shmuel
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Josef Schlomovitz
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - David Kobiler
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| | - Haim Levy
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness Ziona 74100, Israel.
| |
Collapse
|
22
|
Zhang D, Liu W, Wen Z, Li B, Liu S, Li J, Chen W. Establishment of a New Zealand White Rabbit Model for Lethal Toxin (LT) Challenge and Efficacy of Monoclonal Antibody 5E11 in the LT-Challenged Rabbit Model. Toxins (Basel) 2018; 10:E289. [PMID: 30002351 PMCID: PMC6071005 DOI: 10.3390/toxins10070289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/09/2018] [Indexed: 12/23/2022] Open
Abstract
Anthrax caused by Bacillus anthracis is a lethal infectious disease, especially when inhaled, and the mortality rate approaches 100% without treatment. The anthrax antitoxin monoclonal antibody (MAb) 5E11 is a humanized antibody that targets the anthrax protective antigen (PA). The efficacy of 5E11 needs proper animal models. However, anthrax spores are extremely dangerous, so experiments must be conducted under Biosafety Level 3 conditions. Considering the critical effects of lethal toxin (LT) on hosts during infection, we report the establishment of a LT-challenged rabbit model, which caused 100% mortality with a dose of 2 mg PA + 1 mg LF, while a 4 mg PA + 2 mg LF challenge could limit death to within three days. Then, we evaluated 5E11 efficacy against LT. A prophylactic study showed that the i.v. administration of 40 mg/kg 5E11 four days before lethal dose LT challenge could lead to 100% survival. In therapeutic studies, the i.v. administration of 40 mg/kg 5E11 10 min after lethal dose LT challenge could provide complete protection. Overall, we developed a new LT-challenged rabbit model, and our results indicate that 5E11 shows potential for the clinical application in anthrax treatment.
Collapse
Affiliation(s)
- Duanyang Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| | - Weicen Liu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| | - Zhonghua Wen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| | - Bing Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| | - Shuling Liu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| | - Jianmin Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, 20 Dongdajie Street, Fengtai District, Beijing 100071, China.
| |
Collapse
|
23
|
Ireton K, Van Ngo H, Bhalla M. Interaction of microbial pathogens with host exocytic pathways. Cell Microbiol 2018; 20:e12861. [PMID: 29797532 DOI: 10.1111/cmi.12861] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/13/2018] [Indexed: 02/06/2023]
Abstract
Many microbial pathogens co-opt or perturb host membrane trafficking pathways. This review covers recent examples in which microbes interact with host exocytosis, the fusion of intracellular vesicles with the plasma membrane. The bacterial pathogens Listeria monocytogenes and Staphylococcus aureus subvert recycling endosomal pathways of exocytosis in order to induce their entry into human cells. By contrast, entry of the protozoan pathogen Trypanosoma cruzi or the virus adenovirus into host cells involves exploitation of lysosomal exocytosis. Toxins produced by Bacillus anthracis or Vibrio cholerae interfere with exocytosis pathways mediated by the GTPase Rab11 and the exocyst complex. By doing so, anthrax or cholera toxins impair recycling of cadherins to cell-cell junctions and disrupt the barrier properties of endothelial cells or intestinal epithelial cells, respectively. Uropathogenic Escherichia coli (UPEC) is expelled from bladder epithelial cells through two different exocytic routes that involve sensing of bacteria in vacuoles by host Toll-like receptor 4 (TLR4) or monitoring of the pH of lysosomes harbouring UPEC. The TLR4 pathway is mediated by multiple Rab GTPases and the exocyst, whereas the other pathway involves exocytosis of lysosomes. Expulsion of UPEC through these pathways is thought to benefit the host.
Collapse
Affiliation(s)
- Keith Ireton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Hoan Van Ngo
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
24
|
Sécher T, Guilleminault L, Reckamp K, Amanam I, Plantier L, Heuzé-Vourc'h N. Therapeutic antibodies: A new era in the treatment of respiratory diseases? Pharmacol Ther 2018; 189:149-172. [PMID: 29730443 DOI: 10.1016/j.pharmthera.2018.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Respiratory diseases affect millions of people worldwide, and account for significant levels of disability and mortality. The treatment of lung cancer and asthma with therapeutic antibodies (Abs) is a breakthrough that opens up new paradigms for the management of respiratory diseases. Antibodies are becoming increasingly important in respiratory medicine; dozens of Abs have received marketing approval, and many more are currently in clinical development. Most of these Abs target asthma, lung cancer and respiratory infections, while very few target chronic obstructive pulmonary disease - one of the most common non-communicable causes of death - and idiopathic pulmonary fibrosis. Here, we review Abs approved for or in clinical development for the treatment of respiratory diseases. We notably highlight their molecular mechanisms, strengths, and likely future trends.
Collapse
Affiliation(s)
- T Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université François Rabelais de Tours, F-37032 Tours, France
| | - L Guilleminault
- Pôle des Voies respiratoires, Hôpital Larrey, CHU de Toulouse, F-31059 Toulouse, France; STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm, UPS, F-31013 Toulouse, France
| | - K Reckamp
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - I Amanam
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - L Plantier
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université François Rabelais de Tours, F-37032 Tours, France; CHRU de Tours, Service de Pneumologie, F-37000 Tours, France
| | - N Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université François Rabelais de Tours, F-37032 Tours, France.
| |
Collapse
|
25
|
Cui X, Nolen LD, Sun J, Booth M, Donaldson L, Quinn CP, Boyer AE, Hendricks K, Shadomy S, Bothma P, Judd O, McConnell P, Bower WA, Eichacker PQ. Analysis of Anthrax Immune Globulin Intravenous with Antimicrobial Treatment in Injection Drug Users, Scotland, 2009-2010. Emerg Infect Dis 2018; 23:56-65. [PMID: 27983504 PMCID: PMC5176236 DOI: 10.3201/eid2301.160608] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We studied anthrax immune globulin intravenous (AIG-IV) use from a 2009-2010 outbreak of Bacillus anthracis soft tissue infection in injection drug users in Scotland, UK, and we compared findings from 15 AIG-IV recipients with findings from 28 nonrecipients. Death rates did not differ significantly between recipients and nonrecipients (33% vs. 21%). However, whereas only 8 (27%) of 30 patients at low risk for death (admission sequential organ failure assessment score of 0-5) received AIG-IV, 7 (54%) of the 13 patients at high risk for death (sequential organ failure assessment score of 6-11) received treatment. AIG-IV recipients had surgery more often and, among survivors, had longer hospital stays than did nonrecipients. AIG-IV recipients were sicker than nonrecipients. This difference and the small number of higher risk patients confound assessment of AIG-IV effectiveness in this outbreak.
Collapse
|
26
|
Wallez Y, Bouillot S, Soleilhac E, Huber P, Attrée I, Faudry E. CLIQ-BID: A method to quantify bacteria-induced damage to eukaryotic cells by automated live-imaging of bright nuclei. Sci Rep 2018; 8:5. [PMID: 29311668 PMCID: PMC5758606 DOI: 10.1038/s41598-017-18501-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/07/2017] [Indexed: 11/09/2022] Open
Abstract
Pathogenic bacteria induce eukaryotic cell damage which range from discrete modifications of signalling pathways, to morphological alterations and even to cell death. Accurate quantitative detection of these events is necessary for studying host-pathogen interactions and for developing strategies to protect host organisms from bacterial infections. Investigation of morphological changes is cumbersome and not adapted to high-throughput and kinetics measurements. Here, we describe a simple and cost-effective method based on automated analysis of live cells with stained nuclei, which allows real-time quantification of bacteria-induced eukaryotic cell damage at single-cell resolution. We demonstrate that this automated high-throughput microscopy approach permits screening of libraries composed of interference-RNA, bacterial strains, antibodies and chemical compounds in ex vivo infection settings. The use of fluorescently-labelled bacteria enables the concomitant detection of changes in bacterial growth. Using this method named CLIQ-BID (Cell Live Imaging Quantification of Bacteria Induced Damage), we were able to distinguish the virulence profiles of different pathogenic bacterial species and clinical strains.
Collapse
Affiliation(s)
- Yann Wallez
- UGA, CEA, INSERM, CNRS, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Stéphanie Bouillot
- UGA, CEA, INSERM, CNRS, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Emmanuelle Soleilhac
- UGA, CEA, INSERM; Genetics & Chemogenomics, CMBA Platform, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Philippe Huber
- UGA, CEA, INSERM, CNRS, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Ina Attrée
- UGA, CEA, INSERM, CNRS, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Eric Faudry
- UGA, CEA, INSERM, CNRS, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France.
| |
Collapse
|
27
|
Turk BE. Exceptionally Selective Substrate Targeting by the Metalloprotease Anthrax Lethal Factor. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:189-203. [PMID: 30267305 DOI: 10.1007/5584_2018_273] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zinc-dependent metalloprotease anthrax lethal factor (LF) is the enzymatic component of a toxin thought to have a major role in Bacillus anthracis infections. Like many bacterial toxins, LF is a secreted protein that functions within host cells. LF is a highly selective protease that cleaves a limited number of substrates in a site-specific manner, thereby impacting host signal transduction pathways. The major substrates of LF are mitogen-activated protein kinase kinases (MKKs), which lie in the middle of three-component phosphorylation cascades mediating numerous functions in a variety of cells and tissues. How LF targets its limited substrate repertoire has been an active area of investigation. LF recognizes a specific sequence motif surrounding the scissile bonds of substrate proteins. X-ray crystallography of the protease in complex with peptide substrates has revealed the structural basis of selectivity for the LF cleavage site motif. In addition to having interactions proximal to the cleavage site, LF binds directly to a more distal region in its substrates through a so-called exosite interaction. This exosite has been mapped to a surface within a non-catalytic domain of LF with previously unknown function. A putative LF-binding site has likewise been identified on the catalytic domains of MKKs. Here we review our current state of understanding of LF-substrate interactions and discuss the implications for the design and discovery of inhibitors that may have utility as anthrax therapeutics.
Collapse
Affiliation(s)
- Benjamin E Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
28
|
Jiao GS, Kim S, Moayeri M, Thai A, Cregar-Hernandez L, McKasson L, O'Malley S, Leppla SH, Johnson AT. Small molecule inhibitors of anthrax edema factor. Bioorg Med Chem Lett 2017; 28:134-139. [PMID: 29198864 DOI: 10.1016/j.bmcl.2017.11.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/23/2017] [Accepted: 11/24/2017] [Indexed: 11/19/2022]
Abstract
Anthrax is a highly lethal disease caused by the Gram-(+) bacteria Bacillus anthracis. Edema toxin (ET) is a major contributor to the pathogenesis of disease in humans exposed to B. anthracis. ET is a bipartite toxin composed of two proteins secreted by the vegetative bacteria, edema factor (EF) and protective antigen (PA). Our work towards identifying a small molecule inhibitor of anthrax edema factor is the subject of this letter. First we demonstrate that the small molecule probe 5'-Fluorosulfonylbenzoyl 5'-adenosine (FSBA) reacts irreversibly with EF and blocks enzymatic activity. We then show that the adenosine portion of FSBA can be replaced to provide more drug-like molecules which are up to 1000-fold more potent against EF relative to FSBA, display low cross reactivity when tested against a panel of kinases, and are nanomolar inhibitors of EF in a cell-based assay of cAMP production.
Collapse
Affiliation(s)
- Guan-Sheng Jiao
- Hawaii Biotech, 650 Iwilei Road, Suite 204, Honolulu, HI 96817, USA
| | - Seongjin Kim
- Hawaii Biotech, 650 Iwilei Road, Suite 204, Honolulu, HI 96817, USA
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - April Thai
- Hawaii Biotech, 650 Iwilei Road, Suite 204, Honolulu, HI 96817, USA
| | | | - Linda McKasson
- Hawaii Biotech, 650 Iwilei Road, Suite 204, Honolulu, HI 96817, USA
| | - Sean O'Malley
- Hawaii Biotech, 650 Iwilei Road, Suite 204, Honolulu, HI 96817, USA
| | - Stephen H Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan T Johnson
- Hawaii Biotech, 650 Iwilei Road, Suite 204, Honolulu, HI 96817, USA.
| |
Collapse
|
29
|
Guichard A, Jain P, Moayeri M, Schwartz R, Chin S, Zhu L, Cruz-Moreno B, Liu JZ, Aguilar B, Hollands A, Leppla SH, Nizet V, Bier E. Anthrax edema toxin disrupts distinct steps in Rab11-dependent junctional transport. PLoS Pathog 2017; 13:e1006603. [PMID: 28945820 PMCID: PMC5612732 DOI: 10.1371/journal.ppat.1006603] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
Various bacterial toxins circumvent host defenses through overproduction of cAMP. In a previous study, we showed that edema factor (EF), an adenylate cyclase from Bacillus anthracis, disrupts endocytic recycling mediated by the small GTPase Rab11. As a result, cargo proteins such as cadherins fail to reach inter-cellular junctions. In the present study, we provide further mechanistic dissection of Rab11 inhibition by EF using a combination of Drosophila and mammalian systems. EF blocks Rab11 trafficking after the GTP-loading step, preventing a constitutively active form of Rab11 from delivering cargo vesicles to the plasma membrane. Both of the primary cAMP effector pathways -PKA and Epac/Rap1- contribute to inhibition of Rab11-mediated trafficking, but act at distinct steps of the delivery process. PKA acts early, preventing Rab11 from associating with its effectors Rip11 and Sec15. In contrast, Epac functions subsequently via the small GTPase Rap1 to block fusion of recycling endosomes with the plasma membrane, and appears to be the primary effector of EF toxicity in this process. Similarly, experiments conducted in mammalian systems reveal that Epac, but not PKA, mediates the activity of EF both in cell culture and in vivo. The small GTPase Arf6, which initiates endocytic retrieval of cell adhesion components, also contributes to junctional homeostasis by counteracting Rab11-dependent delivery of cargo proteins at sites of cell-cell contact. These studies have potentially significant practical implications, since chemical inhibition of either Arf6 or Epac blocks the effect of EF in cell culture and in vivo, opening new potential therapeutic avenues for treating symptoms caused by cAMP-inducing toxins or related barrier-disrupting pathologies. Recent anthrax outbreaks in Zambia and northern Russia and biodefense preparedness highlight the need for new therapies to counteract fatal late-stage pathologies in patients infected with Bacillus anthracis. Indeed, two toxins secreted by this pathogen—edema toxin (ET) and lethal toxin (LT)—can cause death in face of effective antibiotic treatment. ET, a potent adenylate cyclase, severely impacts host cells and tissues through an overproduction of the ubiquitous second messenger cAMP. Previously, we identified Rab11 as a key host factor inhibited by ET. Blockade of Rab11-dependent endocytic recycling resulted in the disruption of intercellular junctions, likely contributing to life threatening vascular effusion observed in anthrax patients. Here we present a multi-system analysis of the mechanism by which EF inhibits Rab11 and exocyst-dependent trafficking. Epistasis experiments in Drosophila reveal that over-activation of the cAMP effectors PKA and Epac/Rap1 interferes with Rab11-mediated trafficking at two distinct steps. We further describe conserved roles of Epac and the small GTPase Arf6 in ET-mediated disruption of vesicular trafficking and show how chemical inhibition of either pathway greatly alleviates ET-induced edema. Thus, our study defines Epac and Arf6 as promising drug targets for the treatment of infectious diseases and other pathologies involving cAMP overload or related barrier disruption.
Collapse
Affiliation(s)
- Annabel Guichard
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
| | - Prashant Jain
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States of America
| | - Ruth Schwartz
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
| | - Stephen Chin
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
| | - Lin Zhu
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
| | - Beatriz Cruz-Moreno
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
| | - Janet Z. Liu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States of America
| | - Bernice Aguilar
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States of America
- Division of Pediatric Infectious Diseases and the Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Andrew Hollands
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States of America
| | - Stephen H. Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States of America
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States of America
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - Ethan Bier
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, United States of America
- * E-mail:
| |
Collapse
|
30
|
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: 0.6] [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
|
31
|
Suffredini DA, Li Y, Xu W, Moayeri M, Leppla S, Fitz Y, Cui X, Eichacker PQ. Shock and lethality with anthrax edema toxin in rats are associated with reduced arterial responsiveness to phenylephrine and are reversed with adefovir. Am J Physiol Heart Circ Physiol 2017; 313:H946-H958. [PMID: 28887331 DOI: 10.1152/ajpheart.00285.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/18/2022]
Abstract
Although edema toxin (ETx) and lethal toxin (LTx) contribute to Bacillus anthracis shock and lethality, the mechanisms underlying their cardiovascular effects are unclear. We have previously shown that ETx but not LTx inhibited phenylephrine-stimulated contraction of aortic rings prepared from healthy rats and that adefovir, a selective inhibitor of ETx cAMP production, blocked this effect. Here, we examined arterial function in rats that received 24-h ETx or LTx infusions. Compared with control rats, ETx reduced mean arterial pressure (MAP) and survival over 48 h (P ≤ 0.0003) and increased plasma cAMP at 4, 24, and 48 h (P < 0.0001) and nitric oxide (NO) at 24 and 48 h (P ≤ 0.01). Compared with control animals, at 24- and 48-h phenylephrine stimulation of aortic rings from ETx animals produced decreased maximal contractile force (MCF; P = 0.05 and 0.006) and in vivo phenylephrine infusion in ETx animals produced decreased proportional increases in MAP (P < 0.0001 and P = 0.05). In ETx-treated animals, compared with placebo-treated animals, adefovir treatment prevented all lethality (P = 0.01), increased MAP (P ≤ 0.0001), decreased plasma and aortic tissue cAMP at 24 and 48 h, respectively (P ≤ 0.03), and plasma NO at both times (P ≤ 0.004), and increased phenylephrine-stimulated increases in MCF in aortic rings and MAP in vivo at 48 h (P = 0.02). LTx decreased MAP and survival also, but it did not alter the response to phenylephrine of MCF in aortic rings prepared from LTx animals or of MAP in vivo. In conclusion, in rats, hypotension and lethality are associated with reduced arterial contractile function with ETx but not LTx and adefovir improves ETx-induced hypotension and lethality.NEW & NOTEWORTHY The most important aspects of the present study are the findings that 1) in vivo challenge with anthrax edema but not lethal toxin depresses arterial contractile function measured both ex vivo and in vivo and 2) adefovir inhibits the effects of edema toxin on arterial hypotension and improves survival with lethal dose of edema toxin challenge.
Collapse
Affiliation(s)
- Dante A Suffredini
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Yan Li
- 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
| | - 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
| | - Xizhong Cui
- 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
|
32
|
Basso P, Wallet P, Elsen S, Soleilhac E, Henry T, Faudry E, Attrée I. Multiple Pseudomonas species secrete exolysin-like toxins and provoke Caspase-1-dependent macrophage death. Environ Microbiol 2017; 19:4045-4064. [PMID: 28654176 DOI: 10.1111/1462-2920.13841] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/02/2017] [Accepted: 06/19/2017] [Indexed: 12/19/2022]
Abstract
Pathogenic bacteria secrete protein toxins that provoke apoptosis or necrosis of eukaryotic cells. Here, we developed a live-imaging method, based on incorporation of a DNA-intercalating dye into membrane-damaged host cells, to study the kinetics of primary bone marrow-derived macrophages (BMDMs) mortality induced by opportunistic pathogen Pseudomonas aeruginosa expressing either Type III Secretion System (T3SS) toxins or the pore-forming toxin, Exolysin (ExlA). We found that ExlA promotes the activation of Caspase-1 and maturation of interleukin-1β. BMDMs deficient for Caspase-1 and Caspase-11 were resistant to ExlA-induced death. Furthermore, by using KO BMDMs, we determined that the upstream NLRP3/ASC complex leads to the Caspase-1 activation. We also demonstrated that Pseudomonas putida and Pseudomonas protegens and the Drosophila pathogen Pseudomonas entomophila, which naturally express ExlA-like toxins, are cytotoxic toward macrophages and provoke the same type of pro-inflammatory death as does ExlA+ P. aeruginosa. These results demonstrate that ExlA-like toxins of two-partner secretion systems from diverse Pseudomonas species activate the NLRP3 inflammasome and provoke inflammatory pyroptotic death of macrophages.
Collapse
Affiliation(s)
- Pauline Basso
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| | - Pierre Wallet
- CIRI, Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, F-69007, France
| | - Sylvie Elsen
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| | - Emmanuelle Soleilhac
- CMBA Platform, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, CEA, INSERM; Genetics & Chemogenomics, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, F-69007, France
| | - Eric Faudry
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| | - Ina Attrée
- CNRS-ERL5261, INSERM, U1036, CEA, Bacterial Pathogenesis and Cellular Responses, Biosciences and Biotechnology Institute of Grenoble, University Grenoble Alpes, France
| |
Collapse
|
33
|
Zheng S, Fan X, Wang J, Zhao J, Chen PR. Dissection of Kinase Isoforms through Orthogonal and Chemical Inducible Signaling Cascades. Chembiochem 2017; 18:1593-1598. [DOI: 10.1002/cbic.201700255] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Siqi Zheng
- Beijing National Laboratory for Molecular Sciences; Synthetic and Functional Biomolecules Center; Key Laboratory of Bioorganic Chemistry and; Molecular Engineering of the Ministry of Education; College of Chemistry and Molecular Engineering; Peking University; Haidian District Beijing 100871 China
| | - Xinyuan Fan
- Academy for Advanced Interdisciplinary Studies; Peking-Tsinghua Center for Life Sciences; Peking University; Haidian District Beijing 100871 China
| | - Jie Wang
- Academy for Advanced Interdisciplinary Studies; Peking-Tsinghua Center for Life Sciences; Peking University; Haidian District Beijing 100871 China
| | - Jingyi Zhao
- Academy for Advanced Interdisciplinary Studies; Peking-Tsinghua Center for Life Sciences; Peking University; Haidian District Beijing 100871 China
| | - Peng R. Chen
- Beijing National Laboratory for Molecular Sciences; Synthetic and Functional Biomolecules Center; Key Laboratory of Bioorganic Chemistry and; Molecular Engineering of the Ministry of Education; College of Chemistry and Molecular Engineering; Peking University; Haidian District Beijing 100871 China
- Academy for Advanced Interdisciplinary Studies; Peking-Tsinghua Center for Life Sciences; Peking University; Haidian District Beijing 100871 China
| |
Collapse
|
34
|
Li Y, Gould A, Aboye T, Bi T, Breindel L, Shekhtman A, Camarero JA. Full Sequence Amino Acid Scanning of θ-Defensin RTD-1 Yields a Potent Anthrax Lethal Factor Protease Inhibitor. J Med Chem 2017; 60:1916-1927. [PMID: 28151653 DOI: 10.1021/acs.jmedchem.6b01689] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
θ-Defensin RTD-1 is a noncompetitive inhibitor of anthrax lethal factor (LF) protease (IC50 = 390 ± 20 nM, Ki = 365 ± 20 nM) and a weak inhibitor of other mammalian metalloproteases such as TNFα converting enzyme (TACE) (Ki = 4.45 ± 0.48 μM). Using full sequence amino acid scanning in combination with a highly efficient "one-pot" cyclization-folding approach, we obtained an RTD-1-based peptide that was around 10 times more active than wild-type RTD-1 in inhibiting LF protease (IC50 = 43 ± 3 nM, Ki = 18 ± 1 nM). The most active peptide was completely symmetrical, rich in Arg and Trp residues, and able to adopt a native RTD-1-like structure. These results show the power of optimized chemical peptide synthesis approaches for the efficient production of libraries of disulfide-rich backbone-cyclized peptides to quickly perform structure-activity relationship studies for optimizing protease inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Leonard Breindel
- Department of Chemistry, State University of New York , Albany, New York 12222, United States
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York , Albany, New York 12222, United States
| | | |
Collapse
|
35
|
Functional and Evolutionary Analyses Identify Proteolysis as a General Mechanism for NLRP1 Inflammasome Activation. PLoS Pathog 2016; 12:e1006052. [PMID: 27926929 PMCID: PMC5142783 DOI: 10.1371/journal.ppat.1006052] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 11/09/2016] [Indexed: 12/19/2022] Open
Abstract
Inflammasomes are cytosolic multi-protein complexes that initiate immune responses to infection by recruiting and activating the Caspase-1 protease. Human NLRP1 was the first protein shown to form an inflammasome, but its physiological mechanism of activation remains unknown. Recently, specific variants of mouse and rat NLRP1 were found to be activated upon N-terminal cleavage by the anthrax lethal factor protease. However, agonists for other NLRP1 variants, including human NLRP1, are not known, and it remains unclear if they are also activated by proteolysis. Here we demonstrate that two mouse NLRP1 paralogs (NLRP1AB6 and NLRP1BB6) are also activated by N-terminal proteolytic cleavage. We also demonstrate that proteolysis within a specific N-terminal linker region is sufficient to activate human NLRP1. Evolutionary analysis of primate NLRP1 shows the linker/cleavage region has evolved under positive selection, indicative of pathogen-induced selective pressure. Collectively, these results identify proteolysis as a general mechanism of NLRP1 inflammasome activation that appears to be contributing to the rapid evolution of NLRP1 in rodents and primates.
Collapse
|
36
|
Head BM, Rubinstein E, Meyers AFA. Alternative pre-approved and novel therapies for the treatment of anthrax. BMC Infect Dis 2016; 16:621. [PMID: 27809794 PMCID: PMC5094018 DOI: 10.1186/s12879-016-1951-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 10/22/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bacillus anthracis, the causative agent of anthrax, is a spore forming and toxin producing rod-shaped bacterium that is classified as a category A bioterror agent. This pathogenic microbe can be transmitted to both animals and humans. Clinical presentation depends on the route of entry (direct contact, ingestion, injection or aerosolization) with symptoms ranging from isolated skin infections to more severe manifestations such as cardiac or pulmonary shock, meningitis, and death. To date, anthrax is treatable if antibiotics are administered promptly and continued for 60 days. However, if treatment is delayed or administered improperly, the patient's chances of survival are decreased drastically. In addition, antibiotics are ineffective against the harmful anthrax toxins and spores. Therefore, alternative therapeutics are essential. In this review article, we explore and discuss advances that have been made in anthrax therapy with a primary focus on alternative pre-approved and novel antibiotics as well as anti-toxin therapies. METHODS A literature search was conducted using the University of Manitoba search engine. Using this search engine allowed access to a greater variety of journals/articles that would have otherwise been restricted for general use. In order to be considered for discussion for this review, all articles must have been published later than 2009. RESULTS The alternative pre-approved antibiotics demonstrated high efficacy against B. anthracis both in vitro and in vivo. In addition, the safety profile and clinical pharmacology of these drugs were already known. Compounds that targeted underexploited bacterial processes (DNA replication, RNA synthesis, and cell division) were also very effective in combatting B. anthracis. In addition, these novel compounds prevented bacterial resistance. Targeting B. anthracis virulence, more specifically the anthrax toxins, increased the length of which treatment could be administered. CONCLUSIONS Several novel and pre-existing antibiotics, as well as toxin inhibitors, have shown increasing promise. A combination treatment that targets both bacterial growth and toxin production would be ideal and probably necessary for effectively combatting this armed bacterium.
Collapse
Affiliation(s)
- Breanne M. Head
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9 Canada
| | - Ethan Rubinstein
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9 Canada
| | - Adrienne F. A. Meyers
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9 Canada
- National Laboratory for HIV Immunology, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
37
|
Peptide- and proton-driven allosteric clamps catalyze anthrax toxin translocation across membranes. Proc Natl Acad Sci U S A 2016; 113:9611-6. [PMID: 27506790 DOI: 10.1073/pnas.1600624113] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Anthrax toxin is an intracellularly acting toxin in which sufficient information is available regarding the structure of its transmembrane channel, allowing for detailed investigation of models of translocation. Anthrax toxin, comprising three proteins-protective antigen (PA), lethal factor (LF), and edema factor-translocates large proteins across membranes. Here we show that the PA translocase channel has a transport function in which its catalytic active sites operate allosterically. We find that the phenylalanine clamp (ϕ-clamp), the known conductance bottleneck in the PA translocase, gates as either a more closed state or a more dilated state. Thermodynamically, the two channel states have >300-fold different binding affinities for an LF-derived peptide. The change in clamp thermodynamics requires distant α-clamp and ϕ-clamp sites. Clamp allostery and translocation are more optimal for LF peptides with uniform stereochemistry, where the least allosteric and least efficiently translocated peptide had a mixed stereochemistry. Overall, the kinetic results are in less agreement with an extended-chain Brownian ratchet model but, instead, are more consistent with an allosteric helix-compression model that is dependent also on substrate peptide coil-to-helix/helix-to-coil cooperativity.
Collapse
|
38
|
Revisiting the Concept of Targeting Only Bacillus anthracis Toxins as a Treatment for Anthrax. Antimicrob Agents Chemother 2016; 60:4878-85. [PMID: 27270276 DOI: 10.1128/aac.00546-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/26/2016] [Indexed: 02/05/2023] Open
Abstract
Protective antigen (PA)-based vaccines are effective in preventing the development of fatal anthrax disease both in humans and in relevant animal models. The Bacillus anthracis toxins lethal toxin (lethal factor [LF] plus PA) and edema toxin (edema factor [EF] plus PA) are essential for the establishment of the infection, as inactivation of these toxins results in attenuation of the pathogen. Since the toxins reach high toxemia levels at the bacteremic stages of the disease, the CDC's recommendations include combining antibiotic treatment with antitoxin (anti-PA) immunotherapy. We demonstrate here that while treatment with a highly potent neutralizing monoclonal antibody was highly efficient as postexposure prophylaxis treatment, it failed to protect rabbits with any detectable bacteremia (≥10 CFU/ml). In addition, we show that while PA vaccination was effective against a subcutaneous spore challenge, it failed to protect rabbits against systemic challenges (intravenous injection of vegetative bacteria) with the wild-type Vollum strain or a toxin-deficient mutant. To test the possibility that additional proteins, which are secreted by the bacteria under pathogenicity-stimulating conditions in vitro, may contribute to the vaccine's potency, we immunized rabbits with a secreted protein fraction from a toxin-null mutant. The antiserum raised against the secreted fraction reacts with the bacteria in an immunofluorescence assay. Immunization with the secreted protein fraction did not protect the rabbits against a systemic challenge with the fully pathogenic bacteria. Full protection was obtained only by a combined vaccination with PA and the secreted protein fraction. Therefore, these results indicate that an effective antiserum treatment in advanced stages of anthrax must include toxin-neutralizing antibodies in combination with antibodies against bacterial cell targets.
Collapse
|
39
|
Bachran C, Leppla SH. Tumor Targeting and Drug Delivery by Anthrax Toxin. Toxins (Basel) 2016; 8:toxins8070197. [PMID: 27376328 PMCID: PMC4963830 DOI: 10.3390/toxins8070197] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 12/17/2022] Open
Abstract
Anthrax toxin is a potent tripartite protein toxin from Bacillus anthracis. It is one of the two virulence factors and causes the disease anthrax. The receptor-binding component of the toxin, protective antigen, needs to be cleaved by furin-like proteases to be activated and to deliver the enzymatic moieties lethal factor and edema factor to the cytosol of cells. Alteration of the protease cleavage site allows the activation of the toxin selectively in response to the presence of tumor-associated proteases. This initial idea of re-targeting anthrax toxin to tumor cells was further elaborated in recent years and resulted in the design of many modifications of anthrax toxin, which resulted in successful tumor therapy in animal models. These modifications include the combination of different toxin variants that require activation by two different tumor-associated proteases for increased specificity of toxin activation. The anthrax toxin system has proved to be a versatile system for drug delivery of several enzymatic moieties into cells. This highly efficient delivery system has recently been further modified by introducing ubiquitin as a cytosolic cleavage site into lethal factor fusion proteins. This review article describes the latest developments in this field of tumor targeting and drug delivery.
Collapse
Affiliation(s)
| | - Stephen H Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Mechanistic Analysis of the Effect of Deamidation on the Immunogenicity of Anthrax Protective Antigen. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:396-402. [PMID: 26912784 DOI: 10.1128/cvi.00701-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/18/2016] [Indexed: 11/20/2022]
Abstract
The spontaneous modification of proteins, such as deamidation of asparagine residues, can significantly affect the immunogenicity of protein-based vaccines. Using a "genetically deamidated" form of recombinant protective antigen (rPA), we have previously shown that deamidation can decrease the immunogenicity of rPA, the primary component of new-generation anthrax vaccines. In this study, we investigated the biochemical and immunological mechanisms by which deamidation of rPA might decrease the immunogenicity of the protein. We found that loss of the immunogenicity of rPA vaccine was independent of the presence of adjuvant. We assessed the effect of deamidation on the immunodominant neutralizing B-cell epitopes of rPA and found that these epitopes were not significantly affected by deamidation. In order to assess the effect of deamidation on T-cell help for antibody production elicited by rPA vaccine, we examined the ability of the wild-type and genetically deamidated forms of rPA to serve as hapten carriers. We found that when wild-type and genetically deamidated rPA were modified to similar extents with 2,4-dinitrophenyl hapten (DNP) and then used to immunize mice, higher levels of anti-DNP antibodies were elicited by wild-type DNP-rPA than those elicited by the genetically deamidated DNP-rPA, indicating that wild-type rPA elicits more T-cell help than the genetically deamidated form of the protein. These results suggest that a decrease in the ability of deamidated rPA to elicit T-cell help for antibody production is a possible contributor to its lower immunogenicity.
Collapse
|
41
|
Rolando M, Gomez-Valero L, Buchrieser C. Bacterial remodelling of the host epigenome: functional role and evolution of effectors methylating host histones. Cell Microbiol 2016; 17:1098-107. [PMID: 26031999 DOI: 10.1111/cmi.12463] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/15/2015] [Accepted: 05/25/2015] [Indexed: 12/25/2022]
Abstract
The modulation of the chromatin organization of eukaryotic cells plays an important role in regulating key cellular processes including host defence mechanisms against pathogens. Thus, to successfully survive in a host cell, a sophisticated bacterial strategy is the subversion of nuclear processes of the eukaryotic cell. Indeed, the number of bacterial proteins that target host chromatin to remodel the host epigenetic machinery is expanding. Some of the identified bacterial effectors that target the chromatin machinery are 'eukaryotic-like' proteins as they mimic eukaryotic histone writers in carrying the same enzymatic activities. The best-studied examples are the SET domain proteins that methylate histones to change the chromatin landscape. In this review, we will discuss SET domain proteins identified in the Legionella, Chlamydia and Bacillus genomes that encode enzymatic activities targeting host histones. Moreover, we discuss their possible origin as having evolved from prokaryotic ancestors or having been acquired from their eukaryotic hosts during their co-evolution. The characterization of such bacterial effectors as modifiers of the host chromatin landscape is an exciting field of research as it elucidates new bacterial strategies to not only manipulate host functions through histone modifications but it may also identify new modifications of the mammalian host cells not known before.
Collapse
Affiliation(s)
- Monica Rolando
- Institut Pasteur, Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Laura Gomez-Valero
- Institut Pasteur, Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| | - Carmen Buchrieser
- Institut Pasteur, Biologie des Bactéries Intracellulaires, Paris, France.,CNRS UMR 3525, Paris, France
| |
Collapse
|
42
|
In vivo dynamics of active edema and lethal factors during anthrax. Sci Rep 2016; 6:23346. [PMID: 26996161 PMCID: PMC4800402 DOI: 10.1038/srep23346] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/26/2016] [Indexed: 01/21/2023] Open
Abstract
Lethal and edema toxins are critical virulence factors of Bacillus anthracis. However, little is known about their in vivo dynamics of production during anthrax. In this study, we unraveled for the first time the in vivo kinetics of production of the toxin components EF (edema factor) and LF (lethal factor) during cutaneous infection with a wild-type toxinogenic encapsulated strain in immuno-competent mice. We stratified the asynchronous infection process into defined stages through bioluminescence imaging (BLI), while exploiting sensitive quantitative methods by measuring the enzymatic activity of LF and EF. LF was produced in high amounts, while EF amounts steadily increased during the infectious process. This led to high LF/EF ratios throughout the infection, with variations between 50 to a few thousands. In the bloodstream, the early detection of active LF and EF despite the absence of bacteria suggests that they may exert long distance effects. Infection with a strain deficient in the protective antigen toxin component enabled to address its role in the diffusion of LF and EF within the host. Our data provide a picture of the in vivo complexity of the infectious process.
Collapse
|
43
|
Animal Models for the Pathogenesis, Treatment, and Prevention of Infection by Bacillus anthracis. Microbiol Spectr 2016; 3:TBS-0001-2012. [PMID: 26104551 DOI: 10.1128/microbiolspec.tbs-0001-2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This article reviews the characteristics of the major animal models utilized for studies on Bacillus anthracis and highlights their contributions to understanding the pathogenesis and host responses to anthrax and its treatment and prevention. Advantages and drawbacks associated with each model, to include the major models (murine, guinea pig, rabbit, nonhuman primate, and rat), and other less frequently utilized models, are discussed. Although the three principal forms of anthrax are addressed, the main focus of this review is on models for inhalational anthrax. The selection of an animal model for study is often not straightforward and is dependent on the specific aims of the research or test. No single animal species provides complete equivalence to humans; however, each species, when used appropriately, can contribute to a more complete understanding of anthrax and its etiologic agent.
Collapse
|
44
|
Jouvion G, Corre JP, Khun H, Moya-Nilges M, Roux P, Latroche C, Tournier JN, Huerre M, Chrétien F, Goossens PL. Physical Sequestration of Bacillus anthracis in the Pulmonary Capillaries in Terminal Infection. J Infect Dis 2016; 214:281-7. [PMID: 26977051 DOI: 10.1093/infdis/jiw098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/29/2016] [Indexed: 11/14/2022] Open
Abstract
The lung is the terminal target of Bacillus anthracis before death, whatever the route of infection (cutaneous, inhalational, or digestive). During a cutaneous infection in absence of toxins, we observed encapsulated bacteria colonizing the alveolar capillary network, bacteria and hemorrhages in alveolar and bronchiolar spaces, and hypoxic foci in the lung (endothelial cells) and brain (neurons and neuropil). Circulating encapsulated bacteria were as chains of approximately 13 µm in length. Bacteria of such size were immediately trapped within the lung capillary network, but bacteria of shorter length were not. Controlling lung-targeted pathology would be beneficial for anthrax treatment.
Collapse
Affiliation(s)
- Gregory Jouvion
- Institut Pasteur, Histopathologie Humaine et Modèles Animaux PRES Sorbonne-Paris-Cité, Paris Descartes University
| | | | - Huot Khun
- Institut Pasteur, Histopathologie Humaine et Modèles Animaux
| | | | | | - Claire Latroche
- Institut Pasteur, Histopathologie Humaine et Modèles Animaux
| | - Jean-Nicolas Tournier
- Pathogénie des Toxi-Infections Bactériennes Ecole du Val-de-Grâce Unité Interactions Hôte-Agents Pathogènes, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Michel Huerre
- URE Histotechnologie et Pathologie, Institut Pasteur
| | - Fabrice Chrétien
- Institut Pasteur, Histopathologie Humaine et Modèles Animaux PRES Sorbonne-Paris-Cité, Paris Descartes University Neuropathology Unit, CH Sainte-Anne, Paris
| | - Pierre L Goossens
- Institut Pasteur, Histopathologie Humaine et Modèles Animaux Pathogénie des Toxi-Infections Bactériennes
| |
Collapse
|
45
|
Quantitative Determination of Lethal Toxin Proteins in Culture Supernatant of Human Live Anthrax Vaccine Bacillus anthracis A16R. Toxins (Basel) 2016; 8:toxins8030056. [PMID: 26927174 PMCID: PMC4810201 DOI: 10.3390/toxins8030056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/26/2022] Open
Abstract
Bacillus anthracis (B. anthracis) is the etiological agent of anthrax affecting both humans and animals. Anthrax toxin (AT) plays a major role in pathogenesis. It includes lethal toxin (LT) and edema toxin (ET), which are formed by the combination of protective antigen (PA) and lethal factor (LF) or edema factor (EF), respectively. The currently used human anthrax vaccine in China utilizes live-attenuated B. anthracis spores (A16R; pXO1+, pXO2−) that produce anthrax toxin but cannot produce the capsule. Anthrax toxins, especially LT, have key effects on both the immunogenicity and toxicity of human anthrax vaccines. Thus, determining quantities and biological activities of LT proteins expressed by the A16R strain is meaningful. Here, we explored LT expression patterns of the A16R strain in culture conditions using another vaccine strain Sterne as a control. We developed a sandwich ELISA and cytotoxicity-based method for quantitative detection of PA and LF. Expression and degradation of LT proteins were observed in culture supernatants over time. Additionally, LT proteins expressed by the A16R and Sterne strains were found to be monomeric and showed cytotoxic activity, which may be the main reason for side effects of live anthrax vaccines. Our work facilitates the characterization of anthrax vaccines components and establishment of a quality control standard for vaccine production which may ultimately help to ensure the efficacy and safety of the human anthrax vaccine A16R.
Collapse
|
46
|
Does Bacillus anthracis Lethal Toxin Directly Depress Myocardial Function? A Review of Clinical Cases and Preclinical Studies. Toxins (Basel) 2015; 7:5417-34. [PMID: 26703730 PMCID: PMC4690141 DOI: 10.3390/toxins7124891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 11/24/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022] Open
Abstract
The US outbreak of B.anthracis infection in 2001 and subsequent cases in the US and Europe demonstrate that anthrax is a continuing risk for the developed world. While several bacterial components contribute to the pathogenesis of B. anthracis, production of lethal toxin (LT) is strongly associated with the development of hypotension and lethality. However, the mechanisms underlying the cardiovascular instability LT produces are unclear. Some evidence suggests that LT causes shock by impairing the peripheral vasculature, effects consistent with the substantial extravasation of fluid in patients dying with B. anthracis. Other data suggests that LT directly depresses myocardial function. However a clinical correlate for this latter possibility is less evident since functional studies and post-mortem examination in patients demonstrate absent or minimal cardiac changes. The purposes of this review were to first present clinical studies of cardiac functional and histologic pathology with B. anthracis infection and to then examine in vivo, in vitro, and ex vivo preclinical studies of LT’s myocardial effects. Together, these data suggest that it is unclear whether that LT directly depresses cardiac function. This question is important for the clinical management and development of new therapies for anthrax and efforts should continue to be made to answer it.
Collapse
|
47
|
Bower WA, Hendricks K, Pillai S, Guarnizo J, Meaney-Delman D. Clinical Framework and Medical Countermeasure Use During an Anthrax Mass-Casualty Incident. MMWR Recomm Rep 2015; 64:1-22. [DOI: 10.15585/mmwr.rr6404a1] [Citation(s) in RCA: 337] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
| | - William A. Bower
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases
| | - Katherine Hendricks
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases
| | - Satish Pillai
- Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases
| | - Julie Guarnizo
- Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases
| | - Dana Meaney-Delman
- Office of the Director, National Center for Emerging and Zoonotic Infectious Diseases
| |
Collapse
|
48
|
Rolando M, Stefani C, Doye A, Acosta MI, Visvikis O, Yevick HG, Buchrieser C, Mettouchi A, Bassereau P, Lemichez E. Contractile actin cables induced by Bacillus anthracis lethal toxin depend on the histone acetylation machinery. Cytoskeleton (Hoboken) 2015; 72:542-56. [PMID: 26403219 DOI: 10.1002/cm.21256] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 09/18/2015] [Accepted: 09/22/2015] [Indexed: 12/25/2022]
Abstract
It remains a challenge to decode the molecular basis of the long-term actin cytoskeleton rearrangements that are governed by the reprogramming of gene expression. Bacillus anthracis lethal toxin (LT) inhibits mitogen-activated protein kinase (MAPK) signaling, thereby modulating gene expression, with major consequences for actin cytoskeleton organization and the loss of endothelial barrier function. Using a laser ablation approach, we characterized the contractile and tensile mechanical properties of LT-induced stress fibers. These actin cables resist pulling forces that are transmitted at cell-matrix interfaces and at cell-cell discontinuous adherens junctions. We report that treating the cells with trichostatin A (TSA), a broad range inhibitor of histone deacetylases (HDACs), or with MS-275, which targets HDAC1, 2 and 3, induces stress fibers. LT decreased the cellular levels of HDAC1, 2 and 3 and reduced the global HDAC activity in the nucleus. Both the LT and TSA treatments induced Rnd3 expression, which is required for the LT-mediated induction of actin stress fibers. Furthermore, we reveal that treating the LT-intoxicated cells with garcinol, an inhibitor of histone acetyl-transferases (HATs), disrupts the stress fibers and limits the monolayer barrier dysfunctions. These data demonstrate the importance of modulating the flux of protein acetylation in order to control actin cytoskeleton organization and the endothelial cell monolayer barrier.
Collapse
Affiliation(s)
- Monica Rolando
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France.,Biologie Des Bactéries Intracellulaires, Institut Pasteur, Paris, France.,UMR 3525, CNRS, Paris, France
| | - Caroline Stefani
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France.,Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Anne Doye
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France
| | - Maria I Acosta
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France
| | - Orane Visvikis
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France
| | - Hannah G Yevick
- Institut Curie-Centre de Recherche, Membrane and Cell Functions Group; CNRS UMR 168, Physico-Chimie Curie, Université Pierre et Marie Curie, 26 Rue d'ulm, Paris Cedex 05, 75248, France
| | - Carmen Buchrieser
- Biologie Des Bactéries Intracellulaires, Institut Pasteur, Paris, France.,UMR 3525, CNRS, Paris, France
| | - Amel Mettouchi
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France
| | - Patricia Bassereau
- Institut Curie-Centre de Recherche, Membrane and Cell Functions Group; CNRS UMR 168, Physico-Chimie Curie, Université Pierre et Marie Curie, 26 Rue d'ulm, Paris Cedex 05, 75248, France
| | - Emmanuel Lemichez
- Microbial Toxins in Host-Pathogen Interactions, Equipe Labellisée La Ligue Contre Le Cancer, INSERM, U1065, Centre Méditerranéen De Médecine Moléculaire (C3M), 151 Route St Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex, France.,UFR Médecine, IFR50, Faculté De Médecine, Université De Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
49
|
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: 0.7] [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
|
50
|
Goossens PL, Tournier JN. Crossing of the epithelial barriers by Bacillus anthracis: the Known and the Unknown. Front Microbiol 2015; 6:1122. [PMID: 26500645 PMCID: PMC4598578 DOI: 10.3389/fmicb.2015.01122] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/28/2015] [Indexed: 12/15/2022] Open
Abstract
Anthrax, caused by Bacillus anthracis, a Gram-positive spore-forming bacterium, is initiated by the entry of spores into the host body. There are three types of human infection: cutaneous, inhalational, and gastrointestinal. For each form, B. anthracis spores need to cross the cutaneous, respiratory or digestive epithelial barriers, respectively, as a first obligate step to establish infection. Anthrax is a toxi-infection: an association of toxemia and rapidly spreading infection progressing to septicemia. The pathogenicity of Bacillus anthracis mainly depends on two toxins and a capsule. The capsule protects bacilli from the immune system, thus promoting systemic dissemination. The toxins alter host cell signaling, thereby paralyzing the immune response of the host and perturbing the endocrine and endothelial systems. In this review, we will mainly focus on the events and mechanisms leading to crossing of the respiratory epithelial barrier, as the majority of studies have addressed inhalational infection. We will discuss the critical gaps of knowledge that need to be addressed to gain a comprehensive view of the initial steps of inhalational anthrax. We will then discuss the few data available on B. anthracis crossing the cutaneous and digestive epithelia.
Collapse
Affiliation(s)
- Pierre L Goossens
- Pathogénie des Toxi-Infections Bactériennes, Institut Pasteur , Paris, France
| | - Jean-Nicolas Tournier
- Pathogénie des Toxi-Infections Bactériennes, Institut Pasteur , Paris, France ; Unité Interactions Hôte-Agents Pathogènes, Institut de Recherche Biomédicale des Armées , Brétigny-sur-Orge, France ; Ecole du Val-de-Grâce , Paris, France
| |
Collapse
|