151
|
Schwan C, Aktories K. Formation of Nanotube-Like Protrusions, Regulation of Septin Organization and Re-guidance of Vesicle Traffic by Depolymerization of the Actin Cytoskeleton Induced by Binary Bacterial Protein Toxins. Curr Top Microbiol Immunol 2016; 399:35-51. [PMID: 27726005 DOI: 10.1007/82_2016_25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A large group of bacterial protein toxins, including binary ADP-ribosylating toxins, modify actin at arginine-177, thereby actin polymerization is blocked and the actin cytoskeleton is redistributed. Modulation of actin functions largely affects other components of the cytoskeleton, especially microtubules and septins. Here, recent findings about the functional interconnections of the actin cytoskeleton with microtubules and septins, affected by bacterial toxins, are reviewed.
Collapse
Affiliation(s)
- Carsten Schwan
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany
| | - Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| |
Collapse
|
152
|
Abstract
The stoichiometry of a protein complex can be calculated from an accurate measurement of the complex's molecular weight. Multiangle laser light scattering in combination with size exclusion chromatography and interferometric refractometry provides a powerful means for determining the molecular weights of proteins and protein complexes. In contrast to conventional size exclusion chromatography and analytical centrifugation, measurements do not rely on the use of molecular weight standards and are not affected by the shape of the proteins. The technique is based on the direct relationship between the amount of light scattered by a protein in solution, and the product of its concentration and molecular weight. A typical experimental configuration includes a size exclusion column to fractionate the sample, a light scattering detector to measure scattered light, and an interferometric refractometer to measure protein concentration. The determination of the molecular weight of an anthrax toxin complex will be used to illustrate how multiangle laser light scattering can be used to determine the stoichiometry of protein complexes.
Collapse
Affiliation(s)
- Jeremy Mogridge
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, Room 6308, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8,
| |
Collapse
|
153
|
Nair MS, Dean DH. Composition of the Putative Prepore Complex of Bacillus thuringiensis Cry1Ab Toxin. ACTA ACUST UNITED AC 2015; 5:179-188. [PMID: 26702367 PMCID: PMC4686277 DOI: 10.4236/abc.2015.54014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prepore formation is hypothesized to be an obligate step in the insertion of Cry1Ab toxin into insect brush border membrane vesicles. We examined the architecture of the putative prepore when isolated using the published protocols [1] [2]. Our results demonstrate that the putative prepore form of Cry1Ab is a combination of receptor proteins attached to the toxin, when purified. The results also suggest that this prepore form as prepared by the methods published is different from other membrane-extracted oligomeric forms of Cry toxins and prepore of other toxins in general. While most other known prepores are composed of multimers of a single protein, the Cry1Ab prepore, as generated, is a protein-receptor complex oligomer and monomers of Cry toxins.
Collapse
Affiliation(s)
- Manoj S Nair
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA ; Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY, USA
| | - Donald H Dean
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
154
|
Zhang Z, Zhang Y, Shi M, Ye B, Shen W, Li P, Xing L, Zhang X, Hou L, Xu J, Zhao Z, Chen W. Anthrax Susceptibility: Human Genetic Polymorphisms Modulating ANTXR2 Expression. Toxins (Basel) 2015; 8:toxins8010001. [PMID: 26703731 PMCID: PMC4728523 DOI: 10.3390/toxins8010001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/29/2015] [Accepted: 12/09/2015] [Indexed: 01/03/2023] Open
Abstract
Anthrax toxin causes anthrax pathogenesis and expression levels of ANTXR2 (anthrax toxin receptor 2) are strongly correlated with anthrax toxin susceptibility. Previous studies found that ANTXR2 transcript abundance varies considerably in individuals of different ethnic/geographical groups, but no eQTLs (expression quantitative trait loci) have been identified. By using 3C (chromatin conformation capture), CRISPR-mediated genomic deletion and dual-luciferase reporter assay, gene loci containing cis-regulatory elements of ANTXR2 were localized. Two SNPs (single nucleotide polymorphism) at the conserved CREB-binding motif, rs13140055 and rs80314910 in the promoter region of the gene, modulating ANTXR2 promoter activity were identified. Combining these two regulatory SNPs with a previously reported SNP, rs12647691, for the first time, a statistically significant correlation between human genetic variations and anthrax toxin sensitivity was observed. These findings further our understanding of human variability in ANTXR2 expression and anthrax toxin susceptibility.
Collapse
Affiliation(s)
- Zhang Zhang
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Yan Zhang
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Minglei Shi
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Bingyu Ye
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Wenlong Shen
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Ping Li
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Lingyue Xing
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Xiaopeng Zhang
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Lihua Hou
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Junjie Xu
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Zhihu Zhao
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| | - Wei Chen
- Beijing Institute of Biotechnology, No. 20, Dongdajie str., Fengtai District, Beijing 100071, China.
| |
Collapse
|
155
|
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
|
156
|
Molecular assembly of lethal factor enzyme and pre-pore heptameric protective antigen in early stage of translocation. J Mol Model 2015; 22:7. [PMID: 26659402 DOI: 10.1007/s00894-015-2878-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022]
Abstract
During intoxication, the anthrax toxin lethal (LF) and edema (EF) factors initially assemble with the protective antigen (PA) on the plasma membrane of cells expressing the membrane-bound surface-exposed anthrax toxin receptor (ATR). This takes place at the physiological pH prior to entering the acidic environment of the endosome. We elucidated the molecular dynamics (MD) behaviors of the three-dimensional structure of the (PA63)7LF3 complex in various conformations and analyzed the dynamical properties of the fully loaded pre-pore complex on the plasma membrane at the physiological pH. The analysis points to the interaction networks of amino acids conserved between PA63 octamer and heptamer, which are not affected during the initial stage of the LFs binding. The simulations show an asymmetrical movement of the complex domains that directly affect LFs conformations. The conformational and structural alterations of the 2β2-2β3 loops of PA subunits are associated with pore formation. The early conformational changes of the loops appear as they peel off from the domain 2 toward domain 4 of each PA subunit. The LFs unfold in 1α1 segments of their N-terminal initiating the early stage of the pre-pore formation. The results indicate instable regions within the complex and provide important clues concerning the detail of fluctuating residues of the LF-PA interface regions at the early steps of toxins translocation.
Collapse
|
157
|
Das R, Goel AK, Sharma MK, Upadhyay S. Electrochemical DNA sensor for anthrax toxin activator gene atxA-detection of PCR amplicons. Biosens Bioelectron 2015; 74:939-46. [DOI: 10.1016/j.bios.2015.07.066] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/15/2015] [Accepted: 07/28/2015] [Indexed: 02/08/2023]
|
158
|
Rabideau AE, Pentelute BL. A d-Amino Acid at the N-Terminus of a Protein Abrogates Its Degradation by the N-End Rule Pathway. ACS CENTRAL SCIENCE 2015; 1:423-430. [PMID: 26807441 PMCID: PMC4711398 DOI: 10.1021/acscentsci.5b00308] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Indexed: 06/05/2023]
Abstract
Eukaryotes have evolved the ubiquitin (Ub)/proteasome system to degrade polypeptides. The Ub/proteasome system is one way that cells regulate cytosolic protein and amino acids levels through the recognition and ubiquitination of a protein's N-terminus via E1, E2, and E3 enzymes. The process by which the N-terminus stimulates intracellular protein degradation is referred to as the N-end rule. Characterization of the N-end rule has been limited to only the natural l-amino acids. Using a cytosolic delivery platform derived from anthrax lethal toxin, we probed the stability of mixed chirality proteins, containing one d-amino acid on the N-terminus of otherwise all l-proteins. In all cases, we observed that one N-terminal d-amino acid stabilized the cargo protein to proteasomal degradation with respect to the N-end rule. We found that since the mixed chirality proteins were not polyubiquitinated, they evaded N-end-mediated proteasomal degradation. Evidently, a subtle change on the N-terminus of a natural protein can enhance its intracellular lifetime.
Collapse
|
159
|
Di Paola L, Platania CBM, Oliva G, Setola R, Pascucci F, Giuliani A. Characterization of Protein-Protein Interfaces through a Protein Contact Network Approach. Front Bioeng Biotechnol 2015; 3:170. [PMID: 26579512 PMCID: PMC4626657 DOI: 10.3389/fbioe.2015.00170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/12/2015] [Indexed: 11/13/2022] Open
Abstract
Anthrax toxin comprises three different proteins, jointly acting to exert toxic activity: a non-toxic protective agent (PA), toxic edema factor (EF), and lethal factor (LF). Binding of PA to anthrax receptors promotes oligomerization of PA, binding of EF and LF, and then endocytosis of the complex. Homomeric forms of PA, complexes of PA bound to LF and to the endogenous receptor capillary morphogenesis gene 2 (CMG2) were analyzed. In this work, we characterized protein–protein interfaces (PPIs) and identified key residues at PPIs of complexes, by means of a protein contact network (PCN) approach. Flexibility and global and local topological properties of each PCN were computed. The vulnerability of each PCN was calculated using different node removal strategies, with reference to specific PCN topological descriptors, such as participation coefficient, contact order, and degree. The participation coefficient P, the topological descriptor of the node’s ability to intervene in protein inter-module communication, was the key descriptor of PCN vulnerability of all structures. High P residues were localized both at PPIs and other regions of complexes, so that we argued an allosteric mechanism in protein–protein interactions. The identification of residues, with key role in the stability of PPIs, has a huge potential in the development of new drugs, which would be designed to target not only PPIs but also residues localized in allosteric regions of supramolecular complexes.
Collapse
Affiliation(s)
- Luisa Di Paola
- Facoltà Dipartimentale di Ingegneria, Università Campus Bio-Medico di Roma , Rome , Italy
| | | | - Gabriele Oliva
- Facoltà Dipartimentale di Ingegneria, Università Campus Bio-Medico di Roma , Rome , Italy
| | - Roberto Setola
- Facoltà Dipartimentale di Ingegneria, Università Campus Bio-Medico di Roma , Rome , Italy
| | - Federica Pascucci
- Dipartimento di Informatica e Automazione, Università degli studi Roma Tre , Rome , Italy
| | - Alessandro Giuliani
- Dipartimento di Ambiente e Connessa Prevenzione Primaria, Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
160
|
Zuverink M, Barbieri JT. From GFP to β-lactamase: advancing intact cell imaging for toxins and effectors. Pathog Dis 2015; 73:ftv097. [PMID: 26500183 DOI: 10.1093/femspd/ftv097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Canonical reporters such as green fluorescent protein (GFP) and luciferase have assisted researchers in probing cellular pathways and processes. Prior research in pathogenesis depended on sensitivity of biochemical and biophysical techniques to identify effectors and elucidate entry mechanisms. Recently, the β-lactamase (βlac) reporter system has advanced toxin and effector reporting by permitting measurement of βlac delivery into the cytosol or host βlac expression in intact cells. βlac measurement in cells was facilitated by the development of the fluorogenic substrate, CCF2-AM, to identify novel effectors, target cells, and domains involved in bacterial pathogenesis. The assay is also adaptable for high-throughput screening of small molecule inhibitors against toxins, providing information on mechanism and potential therapeutic agents. The versatility and limitations of the βlac reporter system as applied to toxins and effectors are discussed in this review.
Collapse
Affiliation(s)
- Madison Zuverink
- Medical College of Wisconsin, Microbiology and Molecular Genetics, Milwaukee, WI 53226, USA
| | - Joseph T Barbieri
- Medical College of Wisconsin, Microbiology and Molecular Genetics, Milwaukee, WI 53226, USA
| |
Collapse
|
161
|
Peng X, Sun J. Mechanism of ESAT-6 membrane interaction and its roles in pathogenesis of Mycobacterium tuberculosis. Toxicon 2015; 116:29-34. [PMID: 26456678 DOI: 10.1016/j.toxicon.2015.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/21/2015] [Accepted: 10/05/2015] [Indexed: 10/22/2022]
Abstract
The 6-kDa early secreted antigenic target (ESAT-6; EsxA) of Mycobacterium tuberculosis was first identified as a potent T-cell antigen, and it is now recognized as a pore-forming toxin that is essential for virulence of M. tuberculosis. ESAT-6 is secreted through the ESX-1 secretion system (Type VII) of M. tuberculosis and has been implicated to mediate mycobacterial cytosolic translocation within the host macrophages by rupturing the phagosomal membranes. Recent studies have made significant progresses in understanding of the mechanism of ESAT-6 membrane interaction and its role in M. tuberculosis pathogenesis, but important questions still remain to be answered. Here, we summarize the current progress in study of ESAT-6 membrane interaction and its roles in pathogenesis and discuss some of the key remaining questions for future investigation.
Collapse
Affiliation(s)
- Xiuli Peng
- Key Laboratory Agriculture Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agriculture University, Wuhan, 430070, China
| | - Jianjun Sun
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA.
| |
Collapse
|
162
|
Enhanced Immune Response to DNA Vaccine Encoding Bacillus anthracis PA-D4 Protects Mice against Anthrax Spore Challenge. PLoS One 2015; 10:e0139671. [PMID: 26430894 PMCID: PMC4591996 DOI: 10.1371/journal.pone.0139671] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 09/16/2015] [Indexed: 11/19/2022] Open
Abstract
Anthrax has long been considered the most probable bioweapon-induced disease. The protective antigen (PA) of Bacillus anthracis plays a crucial role in the pathogenesis of anthrax. In the current study, we evaluated the efficiency of a genetic vaccination with the fourth domain (D4) of PA, which is responsible for initial binding of the anthrax toxin to the cellular receptor. The eukaryotic expression vector was designed with the immunoglobulin M (IgM) signal sequence encoding for PA-D4, which contains codon-optimized genes. The expression and secretion of recombinant protein was confirmed in vitro in 293T cells transfected with plasmid and detected by western blotting, confocal microscopy, and enzyme-linked immunosorbent assay (ELISA). The results revealed that PA-D4 protein can be efficiently expressed and secreted at high levels into the culture medium. When plasmid DNA was given intramuscularly to mice, a significant PA-D4-specific antibody response was induced. Importantly, high titers of antibodies were maintained for nearly 1 year. Furthermore, incorporation of the SV40 enhancer in the plasmid DNA resulted in approximately a 15-fold increase in serum antibody levels in comparison with the plasmid without enhancer. The antibodies produced were predominantly the immunoglobulin G2 (IgG2) type, indicating the predominance of the Th1 response. In addition, splenocytes collected from immunized mice produced PA-D4-specific interferon gamma (IFN-γ). The biodistribution study showed that plasmid DNA was detected in most organs and it rapidly cleared from the injection site. Finally, DNA vaccination with electroporation induced a significant increase in immunogenicity and successfully protected the mice against anthrax spore challenge. Our approach to enhancing the immune response contributes to the development of DNA vaccines against anthrax and other biothreats.
Collapse
|
163
|
Streatfield SJ, Kushnir N, Yusibov V. Plant-produced candidate countermeasures against emerging and reemerging infections and bioterror agents. PLANT BIOTECHNOLOGY JOURNAL 2015; 13:1136-59. [PMID: 26387510 PMCID: PMC7167919 DOI: 10.1111/pbi.12475] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 05/20/2023]
Abstract
Despite progress in the prevention and treatment of infectious diseases, they continue to present a major threat to public health. The frequency of emerging and reemerging infections and the risk of bioterrorism warrant significant efforts towards the development of prophylactic and therapeutic countermeasures. Vaccines are the mainstay of infectious disease prophylaxis. Traditional vaccines, however, are failing to satisfy the global demand because of limited scalability of production systems, long production timelines and product safety concerns. Subunit vaccines are a highly promising alternative to traditional vaccines. Subunit vaccines, as well as monoclonal antibodies and other therapeutic proteins, can be produced in heterologous expression systems based on bacteria, yeast, insect cells or mammalian cells, in shorter times and at higher quantities, and are efficacious and safe. However, current recombinant systems have certain limitations associated with production capacity and cost. Plants are emerging as a promising platform for recombinant protein production due to time and cost efficiency, scalability, lack of harboured mammalian pathogens and possession of the machinery for eukaryotic post-translational protein modification. So far, a variety of subunit vaccines, monoclonal antibodies and therapeutic proteins (antivirals) have been produced in plants as candidate countermeasures against emerging, reemerging and bioterrorism-related infections. Many of these have been extensively evaluated in animal models and some have shown safety and immunogenicity in clinical trials. Here, we overview ongoing efforts to producing such plant-based countermeasures.
Collapse
Affiliation(s)
| | - Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| |
Collapse
|
164
|
Kyrychenko A. Using fluorescence for studies of biological membranes: a review. Methods Appl Fluoresc 2015; 3:042003. [DOI: 10.1088/2050-6120/3/4/042003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
165
|
Passive Immunotherapy Protects against Enteric Invasion and Lethal Sepsis in a Murine Model of Gastrointestinal Anthrax. Toxins (Basel) 2015; 7:3960-76. [PMID: 26426050 PMCID: PMC4626714 DOI: 10.3390/toxins7103960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/19/2015] [Indexed: 01/09/2023] Open
Abstract
The principal portal for anthrax infection in natural animal outbreaks is the digestive tract. Enteric exposure to anthrax, which is difficult to detect or prevent in a timely manner, could be exploited as an act of terror through contamination of human or animal food. Our group has developed a novel animal model of gastrointestinal (GI) anthrax for evaluation of disease pathogenesis and experimental therapeutics, utilizing vegetative Bacillus anthracis (Sterne strain) administered to A/J mice (a complement-deficient strain) by oral gavage. We hypothesized that a humanized recombinant monoclonal antibody (mAb) * that neutralizes the protective antigen (PA) component of B. anthracis lethal toxin (LT) and edema toxin (ET) could be an effective treatment. Although the efficacy of this anti-anthrax PA mAb has been shown in animal models of inhalational anthrax, its activity in GI infection had not yet been ascertained. We hereby demonstrate that passive immunotherapy with anti-anthrax PA mAb, administered at the same time as gastrointestinal exposure to B. anthracis, prevents lethal sepsis in nearly all cases (>90%), while a delay of up to forty-eight hours in treatment still greatly reduces mortality following exposure (65%). Moreover, passive immunotherapy protects against enteric invasion, associated mucosal injury and subsequent dissemination by gastrointestinal B. anthracis, indicating that it acts to prevent the initial stages of infection. * Expired raxibacumab being cycled off the Strategic National Stockpile; biological activity confirmed by in vitro assay.
Collapse
|
166
|
Cosentino K, Ros U, García-Sáez AJ. Assembling the puzzle: Oligomerization of α-pore forming proteins in membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:457-466. [PMID: 26375417 DOI: 10.1016/j.bbamem.2015.09.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/26/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
Pore forming proteins (PFPs) share the ability of creating pores that allow the passage of ions, proteins or other constituents through a wide variety of target membranes, ranging from bacteria to humans. They often cause cell death, as pore formation disrupts the membrane permeability barrier required for maintaining cell homeostasis. The organization into supramolecular complexes or oligomers that pierce the membrane is a common feature of PFPs. However, the molecular pathway of self-assembly and pore opening remains unclear. Here, we review the most recent discoveries in the mechanism of membrane oligomerization and pore formation of a subset of PFPs, the α-PFPs, whose pore-forming domains are formed by helical segments. Only now we are starting to grasp the molecular details of their function, mainly thanks to the introduction of single molecule microscopy and nanoscopy techniques. This article is part of a Special Issue entitled: Pore-forming toxins edited by Mauro Dalla Serra and Franco Gambale.
Collapse
Affiliation(s)
- Katia Cosentino
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Tübingen, Germany.,Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Uris Ros
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Tübingen, Germany.,Max-Planck Institute for Intelligent Systems, Stuttgart, Germany.,Center for Protein Studies, Havana University, Havana, Cuba
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Tübingen, Germany.,Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| |
Collapse
|
167
|
Colby JM, Krantz BA. Peptide Probes Reveal a Hydrophobic Steric Ratchet in the Anthrax Toxin Protective Antigen Translocase. J Mol Biol 2015; 427:3598-3606. [PMID: 26363343 DOI: 10.1016/j.jmb.2015.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/03/2015] [Accepted: 09/03/2015] [Indexed: 11/27/2022]
Abstract
Anthrax toxin is a tripartite virulence factor produced by Bacillus anthracis during infection. Under acidic endosomal pH conditions, the toxin's protective antigen (PA) component forms a transmembrane channel in host cells. The PA channel then translocates its two enzyme components, lethal factor and edema factor, into the host cytosol under the proton motive force. Protein translocation under a proton motive force is catalyzed by a series of nonspecific polypeptide binding sites, called clamps. A 10-residue guest/host peptide model system, KKKKKXXSXX, was used to functionally probe polypeptide-clamp interactions within wild-type PA channels. The guest residues were Thr, Ala, Leu, Phe, Tyr, and Trp. In steady-state translocation experiments, the channel blocked most tightly with peptides that had increasing amounts of nonpolar surface area. Cooperative peptide binding was observed in the Trp-containing peptide sequence but not the other tested sequences. Trp substitutions into a flexible, uncharged linker between the lethal factor amino-terminal domain and diphtheria toxin A chain expedited translocation. Therefore, peptide-clamp sites in translocase channels can sense large steric features (like tryptophan) in peptides, and while these steric interactions may make a peptide translocate poorly, in the context of folded domains, they can make the protein translocate more rapidly presumably via a hydrophobic steric ratchet mechanism.
Collapse
Affiliation(s)
- Jennifer M Colby
- Molecular Toxicology Graduate Program, University of California, Berkeley, CA 94720, USA
| | - Bryan A Krantz
- Department of Chemistry, University of California, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, 650 West Baltimore St., Baltimore, MD 21201, USA.
| |
Collapse
|
168
|
Brown MJ, Thoren KL, Krantz BA. Role of the α Clamp in the Protein Translocation Mechanism of Anthrax Toxin. J Mol Biol 2015; 427:3340-3349. [PMID: 26344833 DOI: 10.1016/j.jmb.2015.08.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 08/31/2015] [Accepted: 08/31/2015] [Indexed: 10/23/2022]
Abstract
Membrane-embedded molecular machines are utilized to move water-soluble proteins across these barriers. Anthrax toxin forms one such machine through the self-assembly of its three component proteins--protective antigen (PA), lethal factor, and edema factor. Upon endocytosis into host cells, acidification of the endosome induces PA to form a membrane-inserted channel, which unfolds lethal factor and edema factor and translocates them into the host cytosol. Translocation is driven by the proton motive force, composed of the chemical potential, the proton gradient (ΔpH), and the membrane potential (Δψ). A crystal structure of the lethal toxin core complex revealed an "α clamp" structure that binds to substrate helices nonspecifically. Here, we test the hypothesis that, through the recognition of unfolding helical structure, the α clamp can accelerate the rate of translocation. We produced a synthetic PA mutant in which an α helix was crosslinked into the α clamp to block its function. This synthetic construct impairs translocation by raising a yet uncharacterized translocation barrier shown to be much less force dependent than the known unfolding barrier. We also report that the α clamp more stably binds substrates that can form helices than those, such as polyproline, that cannot. Hence, the α clamp recognizes substrates by a general shape-complementarity mechanism. Substrates that are incapable of forming compact secondary structure (due to the introduction of a polyproline track) are severely deficient for translocation. Therefore, the α clamp and its recognition of helical structure in the translocating substrate play key roles in the molecular mechanism of protein translocation.
Collapse
Affiliation(s)
- Michael J Brown
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Katie L Thoren
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Bryan A Krantz
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; Department of Chemistry, University of California, Berkeley, CA 94720, USA; Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, 650 West Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|
169
|
Pore-forming activity of clostridial binary toxins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:512-25. [PMID: 26278641 DOI: 10.1016/j.bbamem.2015.08.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/13/2015] [Accepted: 08/11/2015] [Indexed: 11/24/2022]
Abstract
Clostridial binary toxins (Clostridium perfringens Iota toxin, Clostridium difficile transferase, Clostridium spiroforme toxin, Clostridium botulinum C2 toxin) as Bacillus binary toxins, including Bacillus anthracis toxins consist of two independent proteins, one being the binding component which mediates the internalization into cell of the intracellularly active component. Clostridial binary toxins induce actin cytoskeleton disorganization through mono-ADP-ribosylation of globular actin and are responsible for enteric diseases. Clostridial and Bacillus binary toxins share structurally and functionally related binding components which recognize specific cell receptors, oligomerize, form pores in endocytic vesicle membrane, and mediate the transport of the enzymatic component into the cytosol. Binding components retain the global structure of pore-forming toxins (PFTs) from the cholesterol-dependent cytotoxin family such as perfringolysin. However, their pore-forming activity notably that of clostridial binding components is more related to that of heptameric PFT family including aerolysin and C. perfringens epsilon toxin. This review focuses upon pore-forming activity of clostridial binary toxins compared to other related PFTs. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.
Collapse
|
170
|
Polymer antidotes for toxin sequestration. Adv Drug Deliv Rev 2015; 90:81-100. [PMID: 26026975 DOI: 10.1016/j.addr.2015.05.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/09/2015] [Accepted: 05/21/2015] [Indexed: 12/24/2022]
Abstract
Toxins delivered by envenomation, secreted by microorganisms, or unintentionally ingested can pose an immediate threat to life. Rapid intervention coupled with the appropriate antidote is required to mitigate the threat. Many antidotes are biological products and their cost, methods of production, potential for eliciting immunogenic responses, the time needed to generate them, and stability issues contribute to their limited availability and effectiveness. These factors exacerbate a world-wide challenge for providing treatment. In this review we evaluate a number of polymer constructs that may serve as alternative antidotes. The range of toxins investigated includes those from sources such as plants, animals and bacteria. The development of polymeric heavy metal sequestrants for use as antidotes to heavy metal poisoning faces similar challenges, thus recent findings in this area have also been included. Two general strategies have emerged for the development of polymeric antidotes. In one, the polymer acts as a scaffold for the presentation of ligands with a known affinity for the toxin. A second strategy is to generate polymers with an intrinsic affinity, and in some cases selectivity, to a range of toxins. Importantly, in vivo efficacy has been demonstrated for each of these strategies, which suggests that these approaches hold promise as an alternative to biological or small molecule based treatments.
Collapse
|
171
|
Fang RH, Luk BT, Hu CMJ, Zhang L. Engineered nanoparticles mimicking cell membranes for toxin neutralization. Adv Drug Deliv Rev 2015; 90:69-80. [PMID: 25868452 DOI: 10.1016/j.addr.2015.04.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/08/2015] [Accepted: 04/01/2015] [Indexed: 11/27/2022]
Abstract
Protein toxins secreted from pathogenic bacteria and venomous animals rely on multiple mechanisms to overcome the cell membrane barrier to inflict their virulence effect. A promising therapeutic concept toward developing a broadly applicable anti-toxin platform is to administer cell membrane mimics as decoys to sequester these virulence factors. As such, lipid membrane-based nanoparticulates are an ideal candidate given their structural similarity to cellular membranes. This article reviews the virulence mechanisms employed by toxins at the cell membrane interface and highlights the application of cell-membrane mimicking nanoparticles as toxin decoys for systemic detoxification. In addition, the implication of particle/toxin nanocomplexes in the development of toxoid vaccines is discussed.
Collapse
|
172
|
Ryou JH, Sohn YK, Hwang DE, Kim HS. Shiga-like toxin-based high-efficiency and receptor-specific intracellular delivery system for a protein. Biochem Biophys Res Commun 2015. [PMID: 26220340 DOI: 10.1016/j.bbrc.2015.07.122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cell-specific cytosolic delivery of functional macromolecules with high efficiency is of great significance in molecular medicine and biotechnology. Herein, we present a Shiga-like toxin II-based high-efficiency and receptor-specific intracellular delivery system. We designed and constructed the Shiga-like toxin-based carrier (STC) to comprise the targeting and translocation domains, and used it for delivering a protein cargo. The STC was shown to deliver a protein cargo into the cytosol with high efficiency in a receptor-specific manner, exhibiting much higher efficiency than the most widely used cell-penetrating peptide. The general utility of the STC was demonstrated by modulating the targeting domain. The present delivery platform can be widely used for the intracellular delivery of diverse biomolecules in a receptor-specific and genetically encodable manner.
Collapse
Affiliation(s)
- Jeong-Hyun Ryou
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea
| | - Yoo-Kyoung Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea
| | - Da-Eun Hwang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea.
| |
Collapse
|
173
|
Immunization with a Recombinant, Pseudomonas fluorescens-Expressed, Mutant Form of Bacillus anthracis-Derived Protective Antigen Protects Rabbits from Anthrax Infection. PLoS One 2015. [PMID: 26207820 PMCID: PMC4514824 DOI: 10.1371/journal.pone.0130952] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Protective antigen (PA), one of the components of the anthrax toxin, is the major component of human anthrax vaccine (Biothrax). Human anthrax vaccines approved in the United States and Europe consist of an alum-adsorbed or precipitated (respectively) supernatant material derived from cultures of toxigenic, non-encapsulated strains of Bacillus anthracis. Approved vaccination schedules in humans with either of these vaccines requires several booster shots and occasionally causes adverse injection site reactions. Mutant derivatives of the protective antigen that will not form the anthrax toxins have been described. We have cloned and expressed both mutant (PA SNKE167-ΔFF-315-E308D) and native PA molecules recombinantly and purified them. In this study, both the mutant and native PA molecules, formulated with alum (Alhydrogel), elicited high titers of anthrax toxin neutralizing anti-PA antibodies in New Zealand White rabbits. Both mutant and native PA vaccine preparations protected rabbits from lethal, aerosolized, B. anthracis spore challenge subsequent to two immunizations at doses of less than 1 μg.
Collapse
|
174
|
Translocation of Non-Canonical Polypeptides into Cells Using Protective Antigen. Sci Rep 2015; 5:11944. [PMID: 26178180 PMCID: PMC4503955 DOI: 10.1038/srep11944] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 04/09/2015] [Indexed: 11/09/2022] Open
Abstract
A variety of pathogenic bacteria infect host eukaryotic cells using protein toxins, which enter the cytosol and exert their cytotoxic effects. Anthrax lethal toxin, for example, utilizes the membrane-spanning translocase, protective antigen (PA) pore, to deliver the protein toxin lethal factor (LF) from the endosome into the cytosol of cells. Previous work has investigated the delivery of natural peptides and enzymatic domains appended to the C-terminus of the PA-binding domain of lethal factor (LFN) into the cytosol via PA pore. Here, we move beyond natural amino acids and systematically investigate the translocation of polypeptide cargo containing non-canonical amino acids and functionalities through PA pore. Our results indicate translocation is not perturbed with alterations to the peptide backbone or side-chain. Moreover, despite their structural complexity, we found that the small molecule drugs, doxorubicin and monomethyl auristatin F (MMAF) translocated efficiently through PA pore. However, we found cyclic peptides and the small molecule drug docetaxel abrogated translocation due to their large size and structural rigidity. For cargos that reached the cytosol, we demonstrated that each remained intact after translocation. These studies show PA is capable of translocating non-canonical cargo provided it is in a conformational state conducive for passage through the narrow pore.
Collapse
|
175
|
Schiffmiller A, Anderson D, Finkelstein A. Ion selectivity of the anthrax toxin channel and its effect on protein translocation. ACTA ACUST UNITED AC 2015; 146:183-92. [PMID: 26170174 PMCID: PMC4516782 DOI: 10.1085/jgp.201511388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/09/2015] [Indexed: 11/20/2022]
Abstract
Anthrax toxin consists of three ∼ 85-kD proteins: lethal factor (LF), edema factor (EF), and protective antigen (PA). PA63 (the 63-kD, C-terminal portion of PA) forms heptameric channels ((PA63)7) in planar phospholipid bilayer membranes that enable the translocation of LF and EF across the membrane. These mushroom-shaped channels consist of a globular cap domain and a 14-stranded β-barrel stem domain, with six anionic residues lining the interior of the stem to form rings of negative charges. (PA63)7 channels are highly cation selective, and, here, we investigate the effects on both cation selectivity and protein translocation of mutating each of these anionic residues to a serine. We find that although some of these mutations reduce cation selectivity, selectivity alone does not directly predict the rate of protein translocation; local changes in electrostatic forces must be considered as well.
Collapse
Affiliation(s)
- Aviva Schiffmiller
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Alan Finkelstein
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
176
|
Jacquez P, Avila G, Boone K, Altiyev A, Puschhof J, Sauter R, Arigi E, Ruiz B, Peng X, Almeida I, Sherman M, Xiao C, Sun J. The Disulfide Bond Cys255-Cys279 in the Immunoglobulin-Like Domain of Anthrax Toxin Receptor 2 Is Required for Membrane Insertion of Anthrax Protective Antigen Pore. PLoS One 2015; 10:e0130832. [PMID: 26107617 PMCID: PMC4479931 DOI: 10.1371/journal.pone.0130832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/25/2015] [Indexed: 11/19/2022] Open
Abstract
Anthrax toxin receptors act as molecular clamps or switches that control anthrax toxin entry, pH-dependent pore formation, and translocation of enzymatic moieties across the endosomal membranes. We previously reported that reduction of the disulfide bonds in the immunoglobulin-like (Ig) domain of the anthrax toxin receptor 2 (ANTXR2) inhibited the function of the protective antigen (PA) pore. In the present study, the disulfide linkage in the Ig domain was identified as Cys255-Cys279 and Cys230-Cys315. Specific disulfide bond deletion mutants were achieved by replacing Cys residues with Ala residues. Deletion of the disulfide bond C255-C279, but not C230-C315, inhibited the PA pore-induced release of the fluorescence dyes from the liposomes, suggesting that C255-C279 is essential for PA pore function. Furthermore, we found that deletion of C255-C279 did not affect PA prepore-to-pore conversion, but inhibited PA pore membrane insertion by trapping the PA membrane-inserting loops in proteinaceous hydrophobic pockets. Fluorescence spectra of Trp59, a residue adjacent to the PA-binding motif in von Willebrand factor A (VWA) domain of ANTXR2, showed that deletion of C255-C279 resulted in a significant conformational change on the receptor ectodomain. The disulfide deletion-induced conformational change on the VWA domain was further confirmed by single-particle 3D reconstruction of the negatively stained PA-receptor heptameric complexes. Together, the biochemical and structural data obtained in this study provides a mechanistic insight into the role of the receptor disulfide bond C255-C279 in anthrax toxin action. Manipulation of the redox states of the receptor, specifically targeting to C255-C279, may become a novel strategy to treat anthrax.
Collapse
Affiliation(s)
- Pedro Jacquez
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Gustavo Avila
- Department of Chemistry, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Kyle Boone
- Bioinformatics Program of University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Agamyrat Altiyev
- Bioinformatics Program of University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Jens Puschhof
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Roland Sauter
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Emma Arigi
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Blanca Ruiz
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Xiuli Peng
- China National Key Laboratory of Agricultural Microbiology, Huazhong Agriculture University, Wuhan, 430070, P. R. China
| | - Igor Almeida
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Michael Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas, 77555, United States of America
| | - Chuan Xiao
- Department of Chemistry, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
- * E-mail: (CX); (JS)
| | - Jianjun Sun
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
- * E-mail: (CX); (JS)
| |
Collapse
|
177
|
Ros U, García-Sáez AJ. More Than a Pore: The Interplay of Pore-Forming Proteins and Lipid Membranes. J Membr Biol 2015; 248:545-61. [PMID: 26087906 DOI: 10.1007/s00232-015-9820-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/12/2015] [Indexed: 01/09/2023]
Abstract
Pore-forming proteins (PFPs) punch holes in their target cell membrane to alter their permeability. Permeabilization of lipid membranes by PFPs has received special attention to study the basic molecular mechanisms of protein insertion into membranes and the development of biotechnological tools. PFPs act through a general multi-step mechanism that involves (i) membrane partitioning, (ii) insertion into the hydrophobic core of the bilayer, (iii) oligomerization, and (iv) pore formation. Interestingly, PFPs and membranes show a dynamic interplay. As PFPs are usually produced as soluble proteins, they require a large conformational change for membrane insertion. Moreover, membrane structure is modified upon PFPs insertion. In this context, the toroidal pore model has been proposed to describe a pore architecture in which not only protein molecules but also lipids are directly involved in the structure. Here, we discuss how PFPs and lipids cooperate and remodel each other to achieve pore formation, and explore new evidences of protein-lipid pore structures.
Collapse
Affiliation(s)
- Uris Ros
- Center for Protein Studies, Faculty of Biology, Calle 25 # 455, Plaza de la Revolución, Havana, Cuba
| | | |
Collapse
|
178
|
Effects of metalloprotease anthrax lethal factor on its peptide-based inhibitor R9LF-1. Mol Cell Biochem 2015; 406:293-9. [PMID: 25981534 DOI: 10.1007/s11010-015-2447-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/07/2015] [Indexed: 10/23/2022]
Abstract
The metalloprotease lethal factor (LF) from Bacillus anthracis plays a vital role in anthrax toxin action, and thus becomes a target for anti-anthrax therapy. Following the guidelines based on existing metalloprotease inhibitors, we designed a 'first-generation' LF inhibitor R9LF-1. This inhibitor was shown to be very stable by itself in a wide range of pH and temperature and able to inhibit LF activity in vitro. However, as we reported previously in the presence of LF, this inhibitor was degraded to a small molecular weight species, resulting in a significantly decreased ability to protect MAPKK from cleavage by LF as well as to protect murine macrophages from lethal toxin. In order to elucidate this unusual phenomenon to build solid basis for high-efficiency LF inhibitor development, we performed extensive research to study the effect of LF on its peptide-based inhibitor. Effects of temperature and incubation period of time on generation of the smaller peptide (short version R9LF-1) by LF as well as its catalytic domain were analyzed. We found that LF degraded R9LF-1 with maximum efficiency in the pH range of 7.0-8.5, which correlates well with the range of LF enzymatic activity with its native substrate. The degradation showed a deviation from normal hyperbolic kinetics but a similarity to the kinetics profile of an enzyme-catalyzed reaction with positive cooperativity. The short version R9LF-1 had decreased inhibitory activity toward LF; surprisingly, BIAcore results suggested a better affinity for its binding to LF. In addition, R9LF-1 was not hydrolyzed by other common proteases, such as chymotrypsin and pepsin, suggesting hydrolysis of the bond between amino acid and hydroxamate groups is unique to LF. This study calls for caution when designing peptide-based LF inhibitors and when interpreting effects of these types of inhibitors.
Collapse
|
179
|
Advances in Anthrax Detection: Overview of Bioprobes and Biosensors. Appl Biochem Biotechnol 2015; 176:957-77. [PMID: 25987133 DOI: 10.1007/s12010-015-1625-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 04/08/2015] [Indexed: 12/22/2022]
Abstract
Anthrax is an infectious disease caused by Bacillus anthracis. Although anthrax commonly affects domestic and wild animals, it causes a rare but lethal infection in humans. A variety of techniques have been introduced and evaluated to detect anthrax using cultures, polymerase chain reaction, and immunoassays to address the potential threat of anthrax being used as a bioweapon. The high-potential harm of anthrax in bioterrorism requires sensitive and specific detection systems that are rapid, field-ready, and real-time monitoring. Here, we provide a systematic overview of anthrax detection probes with their potential applications in various ultra-sensitive diagnostic systems.
Collapse
|
180
|
Hardes K, Becker GL, Lu Y, Dahms SO, Köhler S, Beyer W, Sandvig K, Yamamoto H, Lindberg I, Walz L, von Messling V, Than ME, Garten W, Steinmetzer T. Novel Furin Inhibitors with Potent Anti-infectious Activity. ChemMedChem 2015; 10:1218-31. [PMID: 25974265 DOI: 10.1002/cmdc.201500103] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Indexed: 11/10/2022]
Abstract
New peptidomimetic furin inhibitors with unnatural amino acid residues in the P3 position were synthesized. The most potent compound 4-guanidinomethyl-phenylacteyl-Arg-Tle-Arg-4-amidinobenzylamide (MI-1148) inhibits furin with a Ki value of 5.5 pM. The derivatives also strongly inhibit PC1/3, whereas PC2 is less affected. Selected inhibitors were tested in cell culture for antibacterial and antiviral activity against infectious agents known to be dependent on furin activity. A significant protective effect against anthrax and diphtheria toxin was observed in the presence of the furin inhibitors. Furthermore, the spread of the highly pathogenic H5N1 and H7N1 avian influenza viruses and propagation of canine distemper virus was strongly inhibited. Inhibitor MI-1148 was crystallized in complex with human furin. Its N-terminal guanidinomethyl group in the para position of the P5 phenyl ring occupies the same position as that found previously for a structurally related inhibitor containing this substitution in the meta position, thereby maintaining all of the important P5 interactions. Our results confirm that the inhibition of furin is a promising strategy for a short-term treatment of acute infectious diseases.
Collapse
Affiliation(s)
- Kornelia Hardes
- Institute of Pharmaceutical Chemistry, Philipps University, Marbacher Weg 6, 35032 Marburg (Germany)
| | - Gero L Becker
- Institute of Pharmaceutical Chemistry, Philipps University, Marbacher Weg 6, 35032 Marburg (Germany)
| | - Yinghui Lu
- Institute of Virology, Philipps University, Hans-Meerwein-Strasse 2, Marburg (Germany)
| | - Sven O Dahms
- Protein Crystallography Group, Leibniz Institute for Age Research-Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena (Germany)
| | - Susanne Köhler
- Institute of Environmental and Animal Hygiene, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart (Germany)
| | - Wolfgang Beyer
- Institute of Environmental and Animal Hygiene, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart (Germany)
| | - Kirsten Sandvig
- Department of Biochemistry and Centre for Cancer Biomedicine, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0310 Oslo (Norway)
| | - Hiroyuki Yamamoto
- Department of Anatomy and Neurobiology, University of Maryland, Baltimore, Maryland 21201 (USA)
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland, Baltimore, Maryland 21201 (USA)
| | - Lisa Walz
- Veterinary Medicine Division, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Strasse 51-59, 63225 Langen (Germany)
| | - Veronika von Messling
- Veterinary Medicine Division, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Strasse 51-59, 63225 Langen (Germany)
| | - Manuel E Than
- Protein Crystallography Group, Leibniz Institute for Age Research-Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena (Germany)
| | - Wolfgang Garten
- Institute of Virology, Philipps University, Hans-Meerwein-Strasse 2, Marburg (Germany)
| | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Philipps University, Marbacher Weg 6, 35032 Marburg (Germany).
| |
Collapse
|
181
|
Ohanjanian L, Remy KE, Li Y, Cui X, Eichacker PQ. An overview of investigational toxin-directed therapies for the adjunctive management of Bacillus anthracis infection and sepsis. Expert Opin Investig Drugs 2015; 24:851-65. [PMID: 25920540 DOI: 10.1517/13543784.2015.1041587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sepsis with Bacillus anthracis infection has a very high mortality rate despite appropriate antibiotic and supportive therapies. Over the past 15 years, recent outbreaks in the US and in Europe, coupled with anthrax's bioterrorism weapon potential, have stimulated efforts to develop adjunctive therapies to improve clinical outcomes. Since lethal toxin and edema toxin (LT and ET) make central contributions to the pathogenesis of B. anthracis, these have been major targets in this effort. AREAS COVERED Here, the authors review different investigative biopharmaceuticals that have been recently identified for their therapeutic potential as inhibitors of LT or ET. Among these inhibitors are two antibody preparations that have been included in the Strategic National Stockpile (SNS) and several more that have reached Phase I testing. Presently, however, many of these candidate agents have only been studied in vitro and very few tested in bacteria-challenged models. EXPERT OPINION Although a large number of drugs have been identified as potential therapeutic inhibitors of LT and ET, in most cases their testing has been limited. The use of the two SNS antibody therapies during a large-scale exposure to B. anthracis will be difficult. Further testing and development of agents with oral bioavailability and relatively long shelf lives should be a focus for future research.
Collapse
Affiliation(s)
- Lernik Ohanjanian
- National Institutes of Health, Clinical Center, Critical Care Medicine Department , Building 10, Room 2C145, Bethesda, MD 20892 , USA +1 301 402 2914 ; +1 301 402 1213 ;
| | | | | | | | | |
Collapse
|
182
|
Noskov AN. [Molecular model of anthrax toxin translocation into target-cells]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 40:399-404. [PMID: 25898749 DOI: 10.1134/s1068162014040098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anthrax toxin is formed from three components: protective antigen (PA), lethal (LF) and edema (EF) factors. PA83 is cleaved by cell surface protease furin to produce a 63-kDa fragment (PA63). PA63 and LF/EF molecules are assembled to anthrax toxin complexes: oligomer PA63 x 7 + LF/EF x 3. Assembly is occurred during of binding with cellular receptor or near surface of target-cell. This toxin complex forms pore and induces receptor-mediated endocytosis. Formed endosome consists extracellular liquid with LF/EF and membrane-associated ferments (H+ and K+/Na+-ATPases) and proteins (receptors and others). H+ concentration is increased into endosome as result of K/Na-ATPase-dependent- activity of H+-ATPase. Difference of potentials (between endosome and intracellular liquid) is increased and LF/EF molecules are moved to pore and bound with PA63-oligomer to PA63 x 7 + LF/EF x 7 and full block pore (ion-selective channel). Endosome is increased in volume and induces increasing of PA63-oligomer pore to.size of effector complex: LF/EF x 7 + PAl7 x 7 = 750 kDa. Effector complex is translocated from endosome to cytosol by means high difference of potentials (H+) and dissociates from PA47 x 7 complex after cleavage of FFD315-sait by intracellular chymotrypsin-like proteases in all 7 molecules PA63. PA47 x 7 complex (strongly fixed in membrane with debris of hydrophobic loops) return into endosome and pore is destroyed. Endosome pH is decreased rapidly and PA47 x 7 complex is destroyed by endosomal/lysosomal proteases. Receptor-mediated endocytosis is ended by endosome recycling in cell-membrane.
Collapse
|
183
|
Hemmasi S, Czulkies BA, Schorch B, Veit A, Aktories K, Papatheodorou P. Interaction of the Clostridium difficile Binary Toxin CDT and Its Host Cell Receptor, Lipolysis-stimulated Lipoprotein Receptor (LSR). J Biol Chem 2015; 290:14031-44. [PMID: 25882847 DOI: 10.1074/jbc.m115.650523] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Indexed: 12/17/2022] Open
Abstract
CDT (Clostridium difficile transferase) is a binary, actin ADP-ribosylating toxin frequently associated with hypervirulent strains of the human enteric pathogen C. difficile, the most serious cause of antibiotic-associated diarrhea and pseudomembranous colitis. CDT leads to the collapse of the actin cytoskeleton and, eventually, to cell death. Low doses of CDT result in the formation of microtubule-based protrusions on the cell surface that increase the adherence and colonization of C. difficile. The lipolysis-stimulated lipoprotein receptor (LSR) is the host cell receptor for CDT, and our aim was to gain a deeper insight into the interplay between both proteins. We show that CDT interacts with the extracellular, Ig-like domain of LSR with an affinity in the nanomolar range. We identified LSR splice variants in the colon carcinoma cell line HCT116 and disrupted the LSR gene in these cells by applying the CRISPR-Cas9 technology. LSR truncations ectopically expressed in LSR knock-out cells indicated that intracellular parts of LSR are not essential for plasma membrane targeting of the receptor and cellular uptake of CDT. By generating a series of N- and C-terminal truncations of the binding component of CDT (CDTb), we found that amino acids 757-866 of CDTb are sufficient for binding to LSR. With a transposon-based, random mutagenesis approach, we identified potential LSR-interacting epitopes in CDTb. This study increases our understanding about the interaction between CDT and its receptor LSR, which is key to the development of anti-toxin strategies for preventing cell entry of the toxin.
Collapse
Affiliation(s)
- Sarah Hemmasi
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Fakultät für Biologie
| | - Bernd A Czulkies
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Hermann Staudinger Graduate School
| | - Björn Schorch
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Fakultät für Biologie, the Spemann Graduate School of Biology and Medicine, and
| | - Antonia Veit
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie
| | - Klaus Aktories
- From the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, the Centre for Biological Signalling Studies (BIOSS), Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany
| | | |
Collapse
|
184
|
Lo SY, Säbel CE, Mapletoft JP, Siemann S. Influence of chemical denaturants on the activity, fold and zinc status of anthrax lethal factor. Biochem Biophys Rep 2015; 1:68-77. [PMID: 29124135 PMCID: PMC5668564 DOI: 10.1016/j.bbrep.2015.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/17/2022] Open
Abstract
Anthrax lethal factor (LF) is a zinc-dependent endopeptidase which, through a process facilitated by protective antigen, translocates to the host cell cytosol in a partially unfolded state. In the current report, the influence of urea and guanidine hydrochloride (GdnHCl) on LF׳s catalytic function, fold and metal binding was assessed at neutral pH. Both urea and GdnHCl were found to inhibit LF prior to the onset of unfolding, with the inhibition by the latter denaturant being a consequence of its ionic strength. With the exception of demetallated LF (apoLF) in urea, unfolding, as monitored by tryptophan fluorescence spectroscopy, was found to follow a two-state (native to unfolded) mechanism. Analysis of the metal status of LF with 4-(2-pyridylazoresorcinol) (PAR) following urea or GdnHCl exposure suggests the enzyme to be capable of maintaining its metal ion passed the observed unfolding transition in a chelator-inaccessible form. Although an increase in the concentration of the denaturants eventually allowed the chelator access to the protein׳s zinc ion, such process is not correlated with the release of the metal ion. Indeed, significant dissociation of the zinc ion from LF was not observed even at 6 M urea, and only high concentrations of GdnHCl (>3 M) were capable of inducing the release of the metal ion from the protein. Hence, the current study demonstrates not only the propensity of LF to tightly bind its zinc ion beyond the spectroscopically determined unfolding transition, but also the utility of PAR as a structural probe. Lethal factor (LF) is strongly inhibited by guanidine hydrochloride. Except of apoLF in urea, unfolding follows a two-state mechanism. LF shields and retains its zinc ion in an unfolded state. Pyridylazoresorcinol is a useful probe to assess metal accessibility and release.
Collapse
Key Words
- 4-(2-pyridylazo)resorcinol
- CD, circular dichroism
- Chemical denaturants
- DPA, dipicolinic acid
- EDTA, ethylenediaminetetraacetic acid
- EF, edema factor
- LF, anthrax lethal factor
- Lethal factor
- MWCO, molecular weight cut-off
- PA, protective antigen
- PAR, 4-(2-pyridylazo)resorcinol
- Protein folding
- S-pNA, lethal factor substrate
- SASA, solvent-accessible surface area
- SOD, superoxide dismutase
- Tryptophan fluorescence
- Zinc
- ZnLF, zinc-containing lethal factor
- cps, counts per second
Collapse
Affiliation(s)
- Suet Y. Lo
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Crystal E. Säbel
- Bharti School of Engineering, Laurentian University, Sudbury, Ontario, Canada
| | | | - Stefan Siemann
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
- Correspondence to: Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario, Canada P3E 2C6. Tel.: +1 705 675 1151; fax: +1 705 675 4844.
| |
Collapse
|
185
|
Generation and Characterization of Human Monoclonal Antibodies Targeting Anthrax Protective Antigen following Vaccination with a Recombinant Protective Antigen Vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:553-60. [PMID: 25787135 DOI: 10.1128/cvi.00792-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/03/2015] [Indexed: 12/20/2022]
Abstract
The anthrax protective antigen (PA) is the central component of the three-part anthrax toxin, and it is the primary immunogenic component in the approved AVA anthrax vaccine and the "next-generation" recombinant PA (rPA) anthrax vaccines. Animal models have indicated that PA-specific antibodies (AB) are sufficient to protect against infection with Bacillus anthracis. In this study, we investigated the PA domain specificity, affinity, mechanisms of neutralization, and synergistic effects of PA-specific antibodies from a single donor following vaccination with the rPA vaccine. Antibody-secreting cells were isolated 7 days after the donor received a boost vaccination, and 34 fully human monoclonal antibodies (hMAb) were identified. Clones 8H6, 4A3, and 22F1 were able to neutralize lethal toxin (LeTx) both in vitro and in vivo. Clone 8H6 neutralized LeTx by preventing furin cleavage of PA in a dose-dependent manner. Clone 4A3 enhanced degradation of nicked PA, thereby interfering with PA oligomerization. The mechanism of 22F1 is still unclear. A fourth clone, 2A6, that was protective only in vitro was found to be neutralizing in vivo in combination with a toxin-enhancing antibody, 8A7, which binds to domain 3 of PA and PA oligomers. These results provide novel insights into the antibody response elicited by the rPA vaccine and may be useful for PA-based vaccine and immunotherapeutic cocktail design.
Collapse
|
186
|
Atomic structure of anthrax protective antigen pore elucidates toxin translocation. Nature 2015; 521:545-9. [PMID: 25778700 PMCID: PMC4519040 DOI: 10.1038/nature14247] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/19/2015] [Indexed: 12/11/2022]
Abstract
Anthrax toxin, comprising protective antigen (PA), lethal factor (LF) and edema factor (EF), is the major virulence factor of Bacillus anthracis, an agent that causes high mortality in human and animals. PA forms oligomeric prepores that undergo conversion to membrane-spanning pores by endosomal acidification, and these pores translocate the enzymes LF and EF into the cytosol of target cells1. PA is not only a vaccine component and therapeutic target for anthrax infections but also an excellent model system for understanding the mechanism of protein translocation. Based on biochemical and electrophysiological results, researchers have proposed that a Φ-clamp composed of Phe427 residues of PA catalyzes protein translocation via a charge-state dependent Brownian ratchet2–9. Although atomic structures of PA prepores are available10–14, how PA senses low pH, converts to active pore and translocates LF and EF are not well defined without an atomic model of the PA pore. Here, by cryo electron microscopy (cryoEM) with direct electron counting, we have determined the PA pore structure at 2.9-Å resolution. The structure reveals the long-sought-after catalytic Φ-clamp and the membrane-spanning translocation channel, and supports the Brownian ratchet model for protein translocation. Comparisons of four structures reveal conformational changes in prepore to pore conversion that support a multi-step mechanism by which low-pH is sensed and the membrane-spanning channel is formed.
Collapse
|
187
|
Shen J, Cai C, Yu Z, Pang Y, Zhou Y, Qian L, Wei W, Huang Y. A microfluidic live cell assay to study anthrax toxin induced cell lethality assisted by conditioned medium. Sci Rep 2015; 5:8651. [PMID: 25731605 PMCID: PMC4346806 DOI: 10.1038/srep08651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/29/2015] [Indexed: 01/13/2023] Open
Abstract
It is technically challenging to investigate the function of secreted protein in real time by supply of conditioned medium that contains secreted protein of interest. The internalization of anthrax toxin is facilitated by a secreted protein Dickkopf-1 (DKK1) and its receptor, and eventually leads to cell lethality. To monitor the dynamic interplay between these components in live cells, we use an integrated microfluidic device to perform the cell viability assays with real-time controlled culture microenvironment in parallel. Conditioned medium, which contains the secreted proteins from specific cell lines, can be continuously pumped towards the cells that exposed to toxin. The exogenous DKK1 secreted from distant cells is able to rescue the sensitivity to toxin for those DKK1-knocked-down cells. This high-throughput assay allows us to precisely quantify the dynamic interaction between key components that cause cell death, and provide independent evidence of the function of DKK1 in the complex process of anthrax toxin internalization.
Collapse
Affiliation(s)
- Jie Shen
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing, 100871, China [2] College of Engineering, Peking University, Beijing, 100871, China [3] School of Life Sciences, Peking University, Beijing, 100871, China
| | - Changzu Cai
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhilong Yu
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing, 100871, China [2] College of Engineering, Peking University, Beijing, 100871, China
| | - Yuhong Pang
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing, 100871, China [2] School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ying Zhou
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing, 100871, China [2] College of Engineering, Peking University, Beijing, 100871, China
| | - Lili Qian
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Wensheng Wei
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yanyi Huang
- 1] Biodynamic Optical Imaging Center (BIOPIC), Peking University, Beijing, 100871, China [2] College of Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
188
|
Verdurmen WPR, Luginbühl M, Honegger A, Plückthun A. Efficient cell-specific uptake of binding proteins into the cytoplasm through engineered modular transport systems. J Control Release 2015; 200:13-22. [PMID: 25526701 DOI: 10.1016/j.jconrel.2014.12.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 12/13/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Abstract
Through advances in protein scaffold engineering and selection technologies, highly specific binding proteins, which fold under reducing conditions, can be generated against virtually all targets. Despite tremendous therapeutic opportunities, intracellular applications are hindered by difficulties associated with achieving cytosolic delivery, compounded by even correctly measuring it. Here, we addressed cytosolic delivery systematically through the development of a biotin ligase-based assay that objectively quantifies cytosolic delivery in a generic fashion. We developed modular transport systems that consist of a designed ankyrin repeat protein (DARPin) for receptor targeting and a different DARPin for intracellular recognition and a bacterial toxin-derived component for cytosolic translocation. We show that both anthrax pores and the translocation domain of Pseudomonas exotoxin A (ETA) efficiently deliver DARPins into the cytosol. We found that the cargo must not exceed a threshold thermodynamic stability for anthrax pores, which can be addressed by engineering, while the ETA pathway does not appear to have this restriction.
Collapse
Affiliation(s)
- Wouter P R Verdurmen
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| | - Manuel Luginbühl
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| | - Annemarie Honegger
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| | - Andreas Plückthun
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| |
Collapse
|
189
|
Remy KE, Cui X, Li Y, Sun J, Solomon SB, Fitz Y, Barochia AV, Al-Hamad M, Moayeri M, Leppla SH, Eichacker PQ. Raxibacumab augments hemodynamic support and improves outcomes during shock with B. anthracis edema toxin alone or together with lethal toxin in canines. Intensive Care Med Exp 2015; 3:9. [PMID: 26097803 PMCID: PMC4473792 DOI: 10.1186/s40635-015-0043-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Lethal and edema toxin contribute to shock and lethality with Bacillus anthracis. We showed previously in a 96-h sedated canine model that raxibacumab, a monoclonal antibody against protective antigen, augmented hemodynamic support (HS) and improved survival with lethal toxin challenge. Here we study raxibacumab further. Using this model, we have now studied raxibacumab with 24 h edema toxin challenges (Study 1), and lethal and edema toxin challenges together (Study 2). METHODS Using our canine model, we have now studied raxibacumab with 24h edema toxin challenges (Study-1), and lethal and edema toxin challenges together (Study-2). RESULTS In Study 1, compared to no treatment, HS (titrated fluid and norepinephrine) increased mean arterial blood pressure (MAP, p ≤ 0.05) but not survival [0 of 10 (0/10) animals survived in each group] or median survival time [43.8 h (range 16.8 to 80.3) vs. 45.2 h (21.0 to 57.1)]. Compared to HS, HS with raxibacumab treatment at or 6 h after the beginning of edema toxin increased MAP and survival rate (6/7 and 7/8, respectively) and time [96.0 h (39.5 to 96.0) and 96.0 h (89.5 to 96.0), respectively]; (p ≤ 0.05). HS with raxibacumab at 12 h increased MAP (p ≤ 0.05) but not survival [1/5; 55.3 h (12.6 to 96.0)]. In Study-2, survival rate and time increased with HS and raxibacumab at 0 h (4/4) or 6 h after (3/3) beginning lethal and edema toxin compared to HS [0/5; 71.5 h (65 to 93)] (p = 0.01 averaged over raxibacumab groups). CONCLUSIONS Raxibacumab augments HS and improves survival during shock with lethal and edema toxin.
Collapse
Affiliation(s)
- Kenneth E Remy
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Steven B Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Yvonne Fitz
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Amisha V Barochia
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Mariam Al-Hamad
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| | - Mahtab Moayeri
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| | - Stephen H Leppla
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| | - Peter Q Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bldg 10, Rm 2C145, Bethesda, MD, 20892, USA.
| |
Collapse
|
190
|
High-sensitivity MALDI-TOF MS quantification of anthrax lethal toxin for diagnostics and evaluation of medical countermeasures. Anal Bioanal Chem 2015; 407:2847-58. [PMID: 25673244 PMCID: PMC4369318 DOI: 10.1007/s00216-015-8509-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/13/2015] [Accepted: 01/21/2015] [Indexed: 11/15/2022]
Abstract
Inhalation anthrax has a rapid progression and high fatality rate. Pathology and death from inhalation of Bacillus anthracis spores are attributed to the actions of secreted protein toxins. Protective antigen (PA) binds and imports the catalytic component lethal factor (LF), a zinc endoprotease, and edema factor (EF), an adenylyl cyclase, into susceptible cells. PA-LF is termed lethal toxin (LTx) and PA-EF, edema toxin. As the universal transporter for both toxins, PA is an important target for vaccination and immunotherapeutic intervention. However, its quantification has been limited to methods of relatively low analytic sensitivity. Quantification of LTx may be more clinically relevant than LF or PA alone because LTx is the toxic form that acts on cells. A method was developed for LTx-specific quantification in plasma using anti-PA IgG magnetic immunoprecipitation of PA and quantification of LF activity that co-purified with PA. The method was fast (<4 h total time to detection), sensitive at 0.033 ng/mL LTx in plasma for the fast analysis (0.0075 ng/mL LTx in plasma for an 18 h reaction), precise (6.3–9.9 % coefficient of variation), and accurate (0.1–12.7 %error; n ≥ 25). Diagnostic sensitivity was 100 % (n = 27 animal/clinical cases). Diagnostic specificity was 100 % (n = 141). LTx was detected post-antibiotic treatment in 6/6 treated rhesus macaques and 3/3 clinical cases of inhalation anthrax and as long as 8 days post-treatment. Over the course of infection in two rhesus macaques, LTx was first detected at 0.101 and 0.237 ng/mL at 36 h post-exposure and increased to 1147 and 12,107 ng/mL in late-stage anthrax. This demonstrated the importance of LTx as a diagnostic and therapeutic target. This method provides a sensitive, accurate tool for anthrax toxin detection and evaluation of PA-directed therapeutics. Method schematic for analysis of anthrax lethal toxin activity by ID-MALDI-TOF MS ![]()
Collapse
|
191
|
Schmidt G, Papatheodorou P, Aktories K. Novel receptors for bacterial protein toxins. Curr Opin Microbiol 2015; 23:55-61. [DOI: 10.1016/j.mib.2014.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 01/05/2023]
|
192
|
Selection of nanobodies that block the enzymatic and cytotoxic activities of the binary Clostridium difficile toxin CDT. Sci Rep 2015; 5:7850. [PMID: 25597743 PMCID: PMC4297958 DOI: 10.1038/srep07850] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/16/2014] [Indexed: 01/01/2023] Open
Abstract
The spore-forming gut bacterium Clostridium difficile is the leading cause of antibiotic-associated diarrhea in hospitalized patients. The major virulence factors are two large glucosylating cytotoxins. Hypervirulent strains (e.g. ribotype 027) with higher morbidity and mortality additionally produce the binary CDT toxin (Clostridium difficile transferase) that ADP-ribosylates actin and induces microtubule-based cell protrusions. Nanobodies are robust single domain antibodies derived from camelid heavy chain antibodies. Here we report the generation of functional nanobodies against the enzymatic CDTa and the heptameric receptor binding subunit CDTb. The nanobodies were obtained from a variable-domain repertoire library isolated from llamas immunized with recombinant CDTa or CDTb. Five CDTa-specific nanobodies blocked CDTa-mediated ADP-ribosylation of actin. Three CDTa-specific and two CDTb-specific nanobodies neutralized the cytotoxicity of CDTa+b. These nanobodies hold promise as new tools for research, diagnosis and therapy of C. difficile associated disease.
Collapse
|
193
|
Goel AK. Anthrax: A disease of biowarfare and public health importance. World J Clin Cases 2015; 3:20-33. [PMID: 25610847 PMCID: PMC4295216 DOI: 10.12998/wjcc.v3.i1.20] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/23/2014] [Accepted: 10/31/2014] [Indexed: 02/05/2023] Open
Abstract
Bioterrorism has received a lot of attention in the first decade of this century. Biological agents are considered attractive weapons for bioterrorism as these are easy to obtain, comparatively inexpensive to produce and exhibit widespread fear and panic than the actual potential of physical damage. Bacillus anthracis (B. anthracis), the etiologic agent of anthrax is a Gram positive, spore forming, non-motile bacterium. This is supposed to be one of the most potent BW agents because its spores are extremely resistant to natural conditions and can survive for several decades in the environment. B. anthracis spores enter the body through skin lesion (cutaneous anthrax), lungs (pulmonary anthrax), or gastrointestinal route (gastrointestinal anthrax) and germinate, giving rise to the vegetative form. Anthrax is a concern of public health also in many countries where agriculture is the main source of income including India. Anthrax has been associated with human history for a very long time and regained its popularity after Sept 2001 incidence in United States. The present review article describes the history, biology, life cycle, pathogenicity, virulence, epidemiology and potential of B. anthracis as biological weapon.
Collapse
|
194
|
Regulatory mechanisms of anthrax toxin receptor 1-dependent vascular and connective tissue homeostasis. Matrix Biol 2015; 42:56-73. [PMID: 25572963 PMCID: PMC4409530 DOI: 10.1016/j.matbio.2014.12.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 01/06/2023]
Abstract
It is well known that angiogenesis is linked to fibrotic processes in fibroproliferative diseases, but insights into pathophysiological processes are limited, due to lack of understanding of molecular mechanisms controlling endothelial and fibroblastic homeostasis. We demonstrate here that the matrix receptor anthrax toxin receptor 1 (ANTXR1), also known as tumor endothelial marker 8 (TEM8), is an essential component of these mechanisms. Loss of TEM8 function in mice causes reduced synthesis of endothelial basement membrane components and hyperproliferative and leaky blood vessels in skin. In addition, endothelial cell alterations in mutants are almost identical to those of endothelial cells in infantile hemangioma lesions, including activated VEGF receptor signaling in endothelial cells, increased expression of the downstream targets VEGF and CXCL12, and increased numbers of macrophages and mast cells. In contrast, loss of TEM8 in fibroblasts leads to increased rates of synthesis of fiber-forming collagens, resulting in progressive fibrosis in skin and other organs. Compromised interactions between TEM8-deficient endothelial and fibroblastic cells cause dramatic reduction in the activity of the matrix-degrading enzyme MMP2. In addition to insights into mechanisms of connective tissue homeostasis, our data provide molecular explanations for vascular and connective tissue abnormalities in GAPO syndrome, caused by loss-of-function mutations in ANTXR1. Furthermore, the loss of MMP2 activity suggests that fibrotic skin abnormalities in GAPO syndrome are, in part, the consequence of pathophysiological mechanisms underlying syndromes (NAO, Torg and Winchester) with multicentric skin nodulosis and osteolysis caused by homozygous loss-of-function mutations in MMP2.
Collapse
|
195
|
Vibrio cholerae Cytolysin: Structure–Function Mechanism of an Atypical β-Barrel Pore-Forming Toxin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 842:109-25. [DOI: 10.1007/978-3-319-11280-0_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
196
|
Owen JL, Yang T, Mohamadzadeh M. New insights into gastrointestinal anthrax infection. Trends Mol Med 2014; 21:154-63. [PMID: 25577136 DOI: 10.1016/j.molmed.2014.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/16/2014] [Accepted: 12/11/2014] [Indexed: 02/08/2023]
Abstract
Bacterial infections are the primary cause of gastrointestinal (GI) disorders in both developing and developed countries, and are particularly dangerous for infants and children. Bacillus anthracis is the 'archetype zoonotic' pathogen; no other infectious disease affects such a broad range of species, including humans. Importantly, there are more case reports of GI anthrax infection in children than inhalational disease. Early diagnosis is difficult and widespread systemic disease develops rapidly. This review highlights new findings concerning the roles of the gut epithelia, commensal microbiota, and innate lymphoid cells (ILCs) in initiation of disease and systemic dissemination in animal models of GI anthrax, the understanding of which is crucial to designing alternative therapies that target the establishment of infection.
Collapse
Affiliation(s)
- Jennifer L Owen
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tao Yang
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
197
|
Abstract
Plasmid-encoded virulence factors are important in the pathogenesis of diseases caused by spore-forming bacteria. Unlike many other bacteria, the most common virulence factors encoded by plasmids in Clostridium and Bacillus species are protein toxins. Clostridium perfringens causes several histotoxic and enterotoxin diseases in both humans and animals and produces a broad range of toxins, including many pore-forming toxins such as C. perfringens enterotoxin, epsilon-toxin, beta-toxin, and NetB. Genetic studies have led to the determination of the role of these toxins in disease pathogenesis. The genes for these toxins are generally carried on large conjugative plasmids that have common core replication, maintenance, and conjugation regions. There is considerable functional information available about the unique tcp conjugation locus carried by these plasmids, but less is known about plasmid maintenance. The latter is intriguing because many C. perfringens isolates stably maintain up to four different, but closely related, toxin plasmids. Toxin genes may also be plasmid-encoded in the neurotoxic clostridia. The tetanus toxin gene is located on a plasmid in Clostridium tetani, but the botulinum toxin genes may be chromosomal, plasmid-determined, or located on bacteriophages in Clostridium botulinum. In Bacillus anthracis it is well established that virulence is plasmid determined, with anthrax toxin genes located on pXO1 and capsule genes on a separate plasmid, pXO2. Orthologs of these plasmids are also found in other members of the Bacillus cereus group such as B. cereus and Bacillus thuringiensis. In B. thuringiensis these plasmids may carry genes encoding one or more insecticidal toxins.
Collapse
|
198
|
Knapp O, Maier E, Waltenberger E, Mazuet C, Benz R, Popoff MR. Residues involved in the pore-forming activity of theClostridium perfringensiota toxin. Cell Microbiol 2014; 17:288-302. [DOI: 10.1111/cmi.12366] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 07/09/2014] [Accepted: 09/22/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Oliver Knapp
- Institut Pasteur, Bactéries anaérobies et Toxines; 28 rue du Dr Roux, F-75724 Paris Cedex 15 France
| | - Elke Maier
- Rudolf-Virchow-Center, DFG-Research Center for Experimental Biomedicine; University of Würzburg; Versbacher Str. 9 D-97078 Würzburg Germany
| | - Eva Waltenberger
- School of Engineering and Science; Jacobs University Bremen; Campusring 1 D-28759 Bremen Germany
| | - Christelle Mazuet
- Institut Pasteur, Bactéries anaérobies et Toxines; 28 rue du Dr Roux, F-75724 Paris Cedex 15 France
| | - Roland Benz
- Rudolf-Virchow-Center, DFG-Research Center for Experimental Biomedicine; University of Würzburg; Versbacher Str. 9 D-97078 Würzburg Germany
- School of Engineering and Science; Jacobs University Bremen; Campusring 1 D-28759 Bremen Germany
| | - Michel R. Popoff
- Institut Pasteur, Bactéries anaérobies et Toxines; 28 rue du Dr Roux, F-75724 Paris Cedex 15 France
| |
Collapse
|
199
|
Lo SY, Säbel CE, Webb MI, Walsby CJ, Siemann S. High metal substitution tolerance of anthrax lethal factor and characterization of its active copper-substituted analogue. J Inorg Biochem 2014; 140:12-22. [DOI: 10.1016/j.jinorgbio.2014.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/27/2014] [Accepted: 06/16/2014] [Indexed: 01/19/2023]
|
200
|
Sharma O, Collier RJ. Polylysine-mediated translocation of the diphtheria toxin catalytic domain through the anthrax protective antigen pore. Biochemistry 2014; 53:6934-40. [PMID: 25317832 PMCID: PMC4230326 DOI: 10.1021/bi500985v] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The protective antigen (PA) moiety of anthrax toxin forms oligomeric pores in the endosomal membrane, which translocate the effector proteins of the toxin to the cytosol. Effector proteins bind to oligomeric PA via their respective N-terminal domains and undergo N- to C-terminal translocation through the pore. Earlier we reported that a tract of basic amino acids fused to the N-terminus of an unrelated effector protein (the catalytic domain diphtheria toxin, DTA) potentiated that protein to undergo weak PA-dependent translocation. In this study, we varied the location of the tract (N-terminal or C-terminal) and the length of a poly-Lys tract fused to DTA and examined the effects of these variations on PA-dependent translocation into cells and across planar bilayers in vitro. Entry into cells was most efficient with ∼12 Lys residues (K12) fused to the N-terminus but also occurred, albeit 10-100-fold less efficiently, with a C-terminal tract of the same length. Similarly, K12 tracts at either terminus occluded PA pores in planar bilayers, and occlusion was more efficient with the N-terminal tag. We used biotin-labeled K12 constructs in conjunction with streptavidin to show that a biotinyl-K12 tag at either terminus is transiently exposed to the trans compartment of planar bilayers at 20 mV; this partial translocation in vitro was more efficient with an N-terminal tag than a C-terminal tag. Significantly, our studies with polycationic tracts fused to the N- and C-termini of DTA suggest that PA-mediated translocation can occur not only in the N to C direction but also in the C to N direction.
Collapse
Affiliation(s)
- Onkar Sharma
- Department of Microbiology and Immunobiology, Harvard Medical School , 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | | |
Collapse
|