51
|
Gaddis BD, Avramova LV, Chmielewski J. Inhibitors of anthrax lethal factor. Bioorg Med Chem Lett 2007; 17:4575-8. [PMID: 17574849 DOI: 10.1016/j.bmcl.2007.05.089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 05/24/2007] [Accepted: 05/30/2007] [Indexed: 10/23/2022]
Abstract
An inhibitor of anthrax lethal toxin mediated cell death (1) was identified by a medium throughput cell-based screen. This compound was determined to specifically inhibit anthrax lethal factor (LF), and subsequent SAR studies produced an even more potent inhibitor (4). Mechanistic studies identified these agents as uncompetitive inhibitors of LF with Ki values of 3.0 and 1.7 microM, respectively, with good cell potency and low cytotoxicity.
Collapse
Affiliation(s)
- Brandon D Gaddis
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
52
|
Li Y, Sherer K, Cui X, Eichacker PQ. New insights into the pathogenesis and treatment of anthrax toxin-induced shock. Expert Opin Biol Ther 2007; 7:843-54. [PMID: 17555370 DOI: 10.1517/14712598.7.6.843] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Inhalational Bacillus anthracis infection is a leading bioterrorist health threat in the US today. Lethal (LeTx) and edema toxin production are key to the virulent effects of this lethal bacteria. Recent insights into the structure and function of these toxins have increased the understanding of both the pathogenesis and treatment of anthrax. These are binary type toxins comprised of protective antigen necessary for their cellular uptake and either lethal or edema factors, the toxigenic moieties. Primary cellular receptors for protective antigen have been identified and the processing of the completed toxins clarified. Consistent with the ability of lethal factor to cleave mitogen activated protein kinase kinases, the evidence indicates that an excessive inflammatory response does not contribute to shock with LeTx. Rather, the immunosuppressive effects of LeTx could promote infection; however, direct endothelial dysfunction may have an important role in shock due to LeTx. Recent studies show that edema factor, a potent adenyl cyclase, may have a major role in shock during anthrax and that it may also be immunosuppresive. Therapies under development which target several steps in the cellular uptake and function of these two toxins have been effective in both in vitro and in vivo systems. Understanding how best to apply these agents in combination with conventional treatments should be a goal of future research.
Collapse
MESH Headings
- Adenylyl Cyclases/immunology
- Adenylyl Cyclases/metabolism
- Animals
- Anthrax/complications
- Anthrax/drug therapy
- Anthrax/metabolism
- Anthrax Vaccines/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Bacillus anthracis/immunology
- Bacillus anthracis/metabolism
- Bacillus anthracis/pathogenicity
- Bacterial Toxins/immunology
- Bacterial Toxins/metabolism
- Endothelium, Vascular/microbiology
- Endothelium, Vascular/physiopathology
- Humans
- Receptors, Peptide/metabolism
- Shock, Septic/drug therapy
- Shock, Septic/metabolism
- Shock, Septic/microbiology
- Shock, Septic/physiopathology
- Virulence
Collapse
Affiliation(s)
- Yan Li
- National Institutes of Health, Critical Care Medicine Department, Clinical Center, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
53
|
Pelat T, Hust M, Laffly E, Condemine F, Bottex C, Vidal D, Lefranc MP, Dübel S, Thullier P. High-affinity, human antibody-like antibody fragment (single-chain variable fragment) neutralizing the lethal factor (LF) of Bacillus anthracis by inhibiting protective antigen-LF complex formation. Antimicrob Agents Chemother 2007; 51:2758-64. [PMID: 17517846 PMCID: PMC1932538 DOI: 10.1128/aac.01528-06] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The anthrax lethal toxin (LT) consists of two subunits, the protective antigen (PA) and the lethal factor (LF), and is essential for anthrax pathogenesis. Several recombinant antibodies directed against PA and intended for medical use have been obtained, but none against LF, despite the recommendations of anthrax experts. Here we describe an anti-LF single-chain variable fragment (scFv) that originated from an immunized macaque (Macaca fascicularis) and was obtained by phage display. Panning of the library of 1.8 x 10(8) clones allowed the isolation of 2LF, a high-affinity (equilibrium dissociation constant, 1.02 nM) scFv, which is highly neutralizing in the standardized in vitro assay (50% inhibitory concentration, 1.20 +/- 0.06 nM) and in an in vivo assay. The scFv neutralizes anthrax LT by inhibiting the formation of the LF-PA complex. The genes encoding 2LF are very similar to those of human immunoglobulin germ line genes, sharing substantial (84.2%) identity with their most similar, germinally encoded counterparts; this feature favors medical applications. These results, and others formerly published, demonstrate that our approach can generate antibody fragments suitable for prophylaxis and therapeutics.
Collapse
Affiliation(s)
- Thibaut Pelat
- Groupe de Biotechnologie des Anticorps, Département de Biologie des Agents Transmissibles, La Tronche, France
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Kimura RH, Steenblock ER, Camarero JA. Development of a cell-based fluorescence resonance energy transfer reporter for Bacillus anthracis lethal factor protease. Anal Biochem 2007; 369:60-70. [PMID: 17586456 DOI: 10.1016/j.ab.2007.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 04/23/2007] [Accepted: 05/14/2007] [Indexed: 11/23/2022]
Abstract
We report the construction of a cell-based fluorescent reporter for anthrax lethal factor (LF) protease activity using the principle of fluorescence resonance energy transfer (FRET). This was accomplished by engineering an Escherichia coli cell line to express a genetically encoded FRET reporter and LF protease. Both proteins were encoded in two different expression plasmids under the control of different tightly controlled inducible promoters. The FRET-based reporter was designed to contain a LF recognition sequence flanked by the FRET pair formed by CyPet and YPet fluorescent proteins. The length of the linker between both fluorescent proteins was optimized using a flexible peptide linker containing several Gly-Gly-Ser repeats. Our results indicate that this FRET-based LF reporter was readily expressed in E. coli cells showing high levels of FRET in vivo in the absence of LF. The FRET signal, however, decreased five times after inducing LF expression in the same cell. These results suggest that this cell-based LF FRET reporter may be used to screen genetically encoded libraries in vivo against LF.
Collapse
Affiliation(s)
- Richard H Kimura
- Biosciences and Biotechnology Division, Livermore National Laboratory, University of California, Livermore, CA 94550, USA
| | | | | |
Collapse
|
55
|
Hanna ML, Tarasow TM, Perkins J. Mechanistic differences between in vitro assays for hydrazone-based small molecule inhibitors of anthrax lethal factor. Bioorg Chem 2007; 35:50-8. [PMID: 16949126 DOI: 10.1016/j.bioorg.2006.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 07/18/2006] [Accepted: 07/21/2006] [Indexed: 10/24/2022]
Abstract
A systematically generated series of hydrazones were analyzed as potential inhibitors of anthrax lethal factor. The hydrazones were screened using one UV-based and two fluorescence-based in vitro assays. The study identified several inhibitors with IC50 values in the micromolar range, and importantly, significant differences in the types of inhibition were observed with the different assays.
Collapse
Affiliation(s)
- M Leslie Hanna
- Chemistry and Materials Science Directorate, Lawrence Livermore National Laboratory, 7000 East Avenue, Livermore, CA 94551, USA
| | | | | |
Collapse
|
56
|
Sherer K, Li Y, Cui X, Eichacker PQ. Lethal and edema toxins in the pathogenesis of Bacillus anthracis septic shock: implications for therapy. Am J Respir Crit Care Med 2006; 175:211-21. [PMID: 17095744 PMCID: PMC2176088 DOI: 10.1164/rccm.200608-1239cp] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Recent research regarding the structure and function of Bacillus anthracis lethal (LeTx) and edema (ETx) toxins provides growing insights into the pathophysiology and treatment of shock with this lethal bacteria. These are both binary-type toxins composed of protective antigen necessary for their cellular uptake and either lethal or edema factors, the toxigenic moieties. The primary cellular receptors for protective antigen have been identified and constructed and key steps in the extracellular processing and internalization of the toxins clarified. Consistent with the lethal factor's primary action as an intracellular endopeptidase targeting mitogen-activated protein kinase kinases, growing evidence indicates that shock with this toxin does not result from an excessive inflammatory response. In fact, the potent immunosuppressive effects of LeTx may actually contribute to the establishment and persistence of infection. Instead, shock with LeTx may be related to the direct injurious effects of lethal factor on endothelial cell function. Despite the importance of LeTx, very recent studies show that edema factor, a potent adenyl cyclase, has the ability to make a substantial contribution to shock caused by B. anthracis and works additively with LeTx. Furthermore, ETx may contribute to the immunosuppressive effects of LeTx. Therapies under development that target several different steps in the cellular uptake and function of these two toxins have been effective in in vitro and in vivo systems. Understanding how best to apply these agents clinically and how they interact with conventional treatments should be goals for future research.
Collapse
Affiliation(s)
- Kevin Sherer
- Critical Care Medicine Department, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
57
|
Schepetkin IA, Khlebnikov AI, Kirpotina LN, Quinn MT. Novel small-molecule inhibitors of anthrax lethal factor identified by high-throughput screening. J Med Chem 2006; 49:5232-44. [PMID: 16913712 DOI: 10.1021/jm0605132] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Anthrax lethal factor (LF) is a key virulence factor of anthrax lethal toxin. We screened a chemolibrary of 10,000 drug-like molecules for their ability to inhibit LF and identified 18 novel small molecules with potent LF inhibitory activity. Three additional LF inhibitors were identified through further structure-activity relationship (SAR) analysis. All 21 compounds inhibited LF with an IC50 range of 0.8 to 11 muM, utilizing mixed-mode competitive inhibition. An evaluation of inhibitory activity against a range of unrelated proteases showed relatively high specificity for LF. Furthermore, pharmacophore modeling of these compounds showed a high degree of similarity to the model published by Panchal et al. (Nat. Struct. Mol. Biol. 2004, 11, 67-72), indicating that the conformational features of these inhibitors are structurally compatible with the steric constraints of the substrate-binding pocket. These novel LF inhibitors and the structural scaffolds identified as important for inhibitory activity represent promising leads to pursue for further LF inhibitor development.
Collapse
Affiliation(s)
- Igor A Schepetkin
- Department of Veterinary Molecular Biology, Montana State University, Bozeman, Montana 59717, USA
| | | | | | | |
Collapse
|
58
|
Jiao GS, Simo O, Nagata M, O'Malley S, Hemscheidt T, Cregar L, Millis SZ, Goldman ME, Tang C. Selectively guanidinylated derivatives of neamine. Syntheses and inhibition of anthrax lethal factor protease. Bioorg Med Chem Lett 2006; 16:5183-9. [PMID: 16870442 DOI: 10.1016/j.bmcl.2006.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 06/26/2006] [Accepted: 07/05/2006] [Indexed: 11/25/2022]
Abstract
A series of mono-, di-, and tri-guanidinylated derivatives of neamine were prepared via selective guanidinylation of neamine. These molecules represent a novel scaffold as inhibitors of anthrax lethal factor zinc metalloprotease. Methods for the synthesis of these compounds are described, and structure-activity relationships among the series are analyzed. In addition, initial findings regarding the mechanism of LF inhibition for these molecules are presented.
Collapse
Affiliation(s)
- Guan-Sheng Jiao
- Department of Chemistry, Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Suite 200, Aiea, 96701, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Lewis JA, Mongan J, McCammon JA, Cohen SM. Evaluation and Binding-Mode Prediction of Thiopyrone-Based Inhibitors of Anthrax Lethal Factor. ChemMedChem 2006; 1:694-7. [PMID: 16902919 DOI: 10.1002/cmdc.200600102] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jana A Lewis
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA
| | | | | | | |
Collapse
|
60
|
Fridman M, Belakhov V, Lee LV, Liang FS, Wong CH, Baasov T. Dual effect of synthetic aminoglycosides: antibacterial activity against Bacillus anthracis and inhibition of anthrax lethal factor. Angew Chem Int Ed Engl 2006; 44:447-52. [PMID: 15624157 DOI: 10.1002/anie.200462003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Micha Fridman
- Department of Chemistry and Institute of Catalysis Science and Technology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | | | | | | | | | | |
Collapse
|
61
|
Comer JE, Noffsinger DM, McHenry DJ, Weisbaum DM, Chatuev BM, Chopra AK, Peterson JW. Evaluation of the protective effects of quinacrine against Bacillus anthracis Ames. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2006; 69:1083-95. [PMID: 16840255 DOI: 10.1080/15287390500360281] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Bacillus anthracis has gained notoriety as a dangerous biological weapon because of its virulence and ability to produce highly resistant spores. In addition, the ability of this organism to produce plasmid-encoded edema toxin (EdTx) and lethal toxin (LeTx) plays a pivotal role in the pathogenesis of anthrax. In this study, the efficacy of quinacrine was evaluated against the effects of anthrax toxins in vitro and its ability to provide protection against challenge with B. anthracis Ames strain spores in an intranasal mouse and guinea pig model. Quinacrine protected murine macrophages in vitro against cytotoxicity and cAMP production induced by LeTx and EdTx, respectively, at concentrations of 40-80 microM, most likely by preventing acidification of the endosomes. However, animals dosed with human equivalent doses of quinacrine were not protected against respiratory spore challenge. The failure of quinacrine to provide protection against inhalation anthrax was attributed to our inability to attain inhibitory concentrations of the drug in the serum or tissues. After daily administration of 43.3 mg quinacrine to guinea pigs (300 g), serum levels after 96 h were only 9.9 microM, a concentration not sufficient to protect macrophages in vitro. Administration of high doses of quinacrine (86.6 mg/kg) was toxic to the animals. These results illustrate some of the difficulties in developing protective therapeutic strategies against inhalation anthrax even when antitoxic drugs appear effective in vitro.
Collapse
Affiliation(s)
- J E Comer
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas 77555-1070, USA
| | | | | | | | | | | | | |
Collapse
|
62
|
Guarise C, Pasquato L, De Filippis V, Scrimin P. Gold nanoparticles-based protease assay. Proc Natl Acad Sci U S A 2006; 103:3978-82. [PMID: 16537471 PMCID: PMC1449631 DOI: 10.1073/pnas.0509372103] [Citation(s) in RCA: 244] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We describe here a simple assay that allows the visual detection of a protease. The method takes advantage of the high molar absorptivity of the plasmon band of gold colloids and is based on the color change of their solution when treated with dithiols. We used C- and N-terminal cysteinyl derivatives of a peptide substrate exploiting its selective recognition and cleavage by a specific protease. Contrary to the native ones, cleaved peptides are unable to induce nanoparticles aggregation; hence, the color of the solution does not change. The detection of two proteases is reported: thrombin (involved in blood coagulation and thrombosis) and lethal factor (an enzyme component of the toxin produced by Bacillus anthracis). The sensitivity of this nanoparticle-based assay is in the low nanomolar range.
Collapse
Affiliation(s)
- Cristian Guarise
- *Department of Chemical Sciences and Institute for Membrane Technology–National Research Council, Padova Section, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Lucia Pasquato
- Department of Chemical Sciences, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy; and
| | - Vincenzo De Filippis
- Department of Pharmaceutical Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Paolo Scrimin
- *Department of Chemical Sciences and Institute for Membrane Technology–National Research Council, Padova Section, University of Padova, Via Marzolo 1, 35131 Padova, Italy
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
63
|
Jiao GS, Cregar L, Goldman ME, Millis SZ, Tang C. Guanidinylated 2,5-dideoxystreptamine derivatives as anthrax lethal factor inhibitors. Bioorg Med Chem Lett 2006; 16:1527-31. [PMID: 16386899 DOI: 10.1016/j.bmcl.2005.12.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2005] [Revised: 12/09/2005] [Accepted: 12/09/2005] [Indexed: 11/23/2022]
Abstract
Anthrax lethal factor is a Zn(2+)-dependent metalloprotease and the key virulence factor of tripartite anthrax toxin secreted by Bacillus anthracis, the causative agent of anthrax. A series of guanidinylated 2,5-dideoxystreptamine derivatives were designed and synthesized as inhibitors of lethal factor, some of which show strong inhibitory activity against lethal factor in an in vitro FRET assay. Preparation and structure-activity relationships of these compounds are presented.
Collapse
Affiliation(s)
- Guan-Sheng Jiao
- Department of Chemistry, Hawaii Biotech Inc., 99-193 Aiea Heights Drive, Suite 200, Aiea, HI 96701, USA.
| | | | | | | | | |
Collapse
|
64
|
Saunders MJ, Kim H, Woods TA, Nolan JP, Sklar LA, Edwards BS, Graves SW. Microsphere-based protease assays and screening application for lethal factor and factor Xa. Cytometry A 2006; 69:342-52. [PMID: 16604538 DOI: 10.1002/cyto.a.20268] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Proteases regulate many biological pathways in humans and are components of several bacterial toxins. Protease studies and development of protease inhibitors do not follow a single established methodology and are mostly protease specific. METHODS We have created recombinant fusion proteins consisting of a biotinylated attachment sequence linked to a GFP via a protease cleavage site to develop a multiplexable microsphere-based protease assay system. Using the proteases lethal factor and factor Xa, we performed kinetic experiments to determine optimal conditions for inhibitor screens and detect known inhibitors using the HyperCyt flow cytometry system. RESULTS We have demonstrated specific cleavage of lethal factor and factor Xa substrates, optimized screening conditions for these substrates, shown specific inhibition of the proteases, and demonstrated high throughput detection of these inhibitors. CONCLUSIONS The assay developed here is adaptable to any site-specific protease, compatible with high throughput flow cytometry systems, and multiplexable. Coupled with flow cytometry, which provides continuous time resolution and intrinsic resolution of free vs. bound fluorophores, this assay will be useful for high throughput screening of protease inhibitors in general and could simplify assays designed to determine protease mechanism.
Collapse
Affiliation(s)
- Matthew J Saunders
- National Flow Cytometry Resource, Biosciences Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, USA
| | | | | | | | | | | | | |
Collapse
|
65
|
Opal SM, Artenstein AW, Cristofaro PA, Jhung JW, Palardy JE, Parejo NA, Lim YP. Inter-alpha-inhibitor proteins are endogenous furin inhibitors and provide protection against experimental anthrax intoxication. Infect Immun 2005; 73:5101-5. [PMID: 16041026 PMCID: PMC1201260 DOI: 10.1128/iai.73.8.5101-5105.2005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inter-alpha-inhibitor protein (IalphaIp) functions as an endogenous serine protease inhibitor in human plasma, and IalphaIp levels diminish rapidly during acute inflammatory states. One potential target for IalphaIp is furin, a cell-associated serine endopeptidase essential for the activation of protective antigen and the formation of anthrax lethal toxin (LT). IalphaIp blocks furin activity in vitro and provides significant protection against cytotoxicity for murine peritoneal macrophages exposed to up to 500 ng/ml LT. A monoclonal antibody (MAb), 69.31, that specifically blocks the enzymatic activity of IalphaIp eliminates its protective effect against LT-induced cytotoxicity. IalphaIp (30 mg/kg of body weight) administered to BALB/c mice 1 hour prior to an intravenous LT challenge resulted in 71% survival after 7 days compared with no survivors among the control animals (P < 0.001). We conclude that human IalphaIp may be an effective preventative or therapeutic agent against anthrax intoxication.
Collapse
Affiliation(s)
- Steven M Opal
- Center for Biodefense and Emerging Pathogens, Memorial Hospital of RI, 111 Brewster Street, Pawtucket, RI 02860, USA.
| | | | | | | | | | | | | |
Collapse
|
66
|
Kim J, Choi MK, Koo BS, Yoon MY. Development of high-throughput assay of lethal factor using native substrate. Anal Biochem 2005; 341:33-9. [PMID: 15866525 DOI: 10.1016/j.ab.2005.03.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Indexed: 11/22/2022]
Abstract
The design of inhibitors for anthrax lethal factor (LF) is currently of interest as an approach for the treatment of anthrax because LF plays a major role in the cytotoxicity of target cells. LF is a zinc-dependent metalloprotease that specifically cleaves the mitogen-activated protein kinase kinase (MKK) family. Current assay systems for the screening of LF inhibitor use the optimized synthetic peptide coupled with various kinds of fluorophores, enabling fast, sensitive, and robust assays suited to high-throughput screening. However, evidence suggests that the regions beside the cleavage site are also involved in specificity and proteolytic activity of LF. In the current study, we tried to develop a high-throughput assay for LF activity based on native substrate, mitogen-activated ERK kinase 1 (MEK1). The assay system relies on the enhanced chemiluminescence signal resulting from a specific antibody against the C-terminal region of native substrate. A glutathione-coated multiwell plate was used as a solid support to immobilize the native substrate by its N-terminal glutathione-S-transferase moiety. Immobilized substrate increases the specificity and sensitivity of LF-catalyzed substrate hydrolysis compared with the solution phase assay. This assay system might be used to discover a wide spectrum of anthrax inhibitors.
Collapse
Affiliation(s)
- Joungmok Kim
- Department of Chemistry, College of Natural Science, Hanyang University, Seoul 133-791, Republic of Korea
| | | | | | | |
Collapse
|
67
|
Burnett JC, Henchal EA, Schmaljohn AL, Bavari S. The evolving field of biodefence: therapeutic developments and diagnostics. Nat Rev Drug Discov 2005; 4:281-97. [PMID: 15803193 PMCID: PMC7096857 DOI: 10.1038/nrd1694] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bioweapons are a clear threat to both military and civilian populations. Here, the latest advances in the pursuit of inhibitors against biothreat threat toxins, current therapeutic strategies for treating biodefence related pathogens, and strategies for improving detection and exposure survivability are covered. There are numerous lead therapeutics that have emerged from drug discovery efforts. However, many of these are toxic and/or fail to possess conventional drug-like properties. One clear advantage of small (non-peptidic) molecules is that they possess scaffolds that are inherently more likely to evolve into real therapeutics. One of the major obstacles impeding the translation of these lead therapeutics into viable drugs is the lack of involvement of the pharmaceutical industry, which has been discovering leads and translating them into drugs for decades. The expertise of the pharmaceutical industry therefore needs to be more effectively engaged in developing drugs against biothreat agents. New methods for rapidly detecting and diagnosing biothreat agents are also in development. The detection and diagnosis of biothreats is inherently linked with treatment. The means for detecting the release of bioweapons are being deployed, and new technologies are shortening the timeframe between initial sample collection and conclusive agent determination. However, the organization of this process is imperfect. At present, a unifying entity that orchestrates the biodefence response is clearly needed to reduce the time-to-drug process and redundancies in drug development efforts. Such a central entity could formulate and implement plans to coordinate all participants, including academic institutions, government agencies and the private sector. This could accelerate the development of countermeasures against high probability biothreat agents.
The threat of bioterrorism and the potential use of biological weapons against both military and civilian populations has become a major concern for governments around the world. For example, in 2001 anthrax-tainted letters resulted in several deaths, caused widespread public panic and exerted a heavy economic toll. If such a small-scale act of bioterrorism could have such a huge impact, then the effects of a large-scale attack would be catastrophic. This review covers recent progress in developing therapeutic countermeasures against, and diagnostics for, such agents.
Collapse
Affiliation(s)
- James C. Burnett
- Developmental Therapeutics Program, Target Structure-Based Drug Discovery Group, National Cancer Institute-SAIC, Frederick, 21702 Maryland USA
| | - Erik A. Henchal
- United States Army Medical Research Institute of Infectious Diseases, Frederick, 21702 Maryland USA
| | - Alan L. Schmaljohn
- United States Army Medical Research Institute of Infectious Diseases, Frederick, 21702 Maryland USA
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Frederick, 21702 Maryland USA
| |
Collapse
|
68
|
Kim C, Gajendran N, Mittrücker HW, Weiwad M, Song YH, Hurwitz R, Wilmanns M, Fischer G, Kaufmann SHE. Human alpha-defensins neutralize anthrax lethal toxin and protect against its fatal consequences. Proc Natl Acad Sci U S A 2005; 102:4830-5. [PMID: 15772169 PMCID: PMC555714 DOI: 10.1073/pnas.0500508102] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Anthrax caused by Bacillus anthracis represents a major bioterroristic threat. B. anthracis produces lethal toxin (LeTx), a combination of lethal factor (LF) and protective antigen that plays a major role in anthrax pathogenesis. We demonstrate that human neutrophil alpha-defensins are potent inhibitors of LF. The inhibition of LF by human neutrophil protein (HNP-1) was noncompetitive. HNP-1 inhibited cleavage of a mitogen-activated protein kinase kinase and restored impaired mitogen-activated protein kinase signaling in LeTx-treated macrophages. HNP-1 rescued murine macrophages from B. anthracis-induced cytotoxicity, and in vivo treatment with HNP-1-3 protected mice against the fatal consequences of LeTx.
Collapse
Affiliation(s)
- Chun Kim
- Department of Immunology, Max Planck Institute for Infection Biology, Schumannstrasse 21-22, D-10117 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Fridman M, Belakhov V, Lee LV, Liang FS, Wong CH, Baasov T. Dual Effect of Synthetic Aminoglycosides: Antibacterial Activity againstBacillus anthracis and Inhibition of Anthrax Lethal Factor. Angew Chem Int Ed Engl 2005. [DOI: 10.1002/ange.200462003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
70
|
Liang X, Young JJ, Boone SA, Waugh DS, Duesbery NS. Involvement of Domain II in Toxicity of Anthrax Lethal Factor. J Biol Chem 2004; 279:52473-8. [PMID: 15465830 DOI: 10.1074/jbc.m409105200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anthrax lethal factor (LF) is a Zn2+ -metalloprotease that cleaves and inactivates mitogen-activated protein kinase kinases (MEKs). We have used site-directed mutagenesis to identify a cluster of residues in domain II of LF that lie outside the active site and are required for cellular proteolytic activity toward MEKs. Alanine substituted for Leu293, Lys294, Leu514, Asn516, or Arg491 caused a 10-50-fold reduction in LF toxicity. Further, whereas pairwise substitution of alanine for Leu514 and either Leu293, Lys294, or Arg491 completely abrogated LF toxicity, pairwise mutation of Leu514 and Asn516 resulted in toxicity comparable with N516A alone. The introduction of these mutations reduced LF-mediated cleavage of MEK2 in cell-based assays but altered neither the ability of LF to bind protective antigen nor its ability to translocate across a membrane. Interestingly, direct in vitro measurement of LF activity indicated that decreased toxicity was not always accompanied by reduced proteolytic activity. However, mutations in this region significantly reduced the ability of LF to competitively inhibit B-Raf phosphorylation of MEK. These results provide evidence that elements of domain II are involved in the association of LF into productive complex with MEKs.
Collapse
Affiliation(s)
- Xudong Liang
- Laboratory of Cancer and Developmental Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | | | | | | | | |
Collapse
|
71
|
Lee VJ. Biowarfare Pathogens. Is the Research Flavor Different Than That of Clinically Relevant Pathogens? ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2004; 39:211-221. [PMID: 32287465 PMCID: PMC7127116 DOI: 10.1016/s0065-7743(04)39017-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
This chapter introduces four chemical warfare agents: bacillus anthracis (anthrax), yersinia pestis (plague), variola major (smallpox), and francesella tularensis (tularemia). Anthrax is a dimorphic bacterium that normally exists as spores. The clinical presentation can be as cutaneous, inhalational or gastrointestinal forms that are fortuitously not transmissible from person to person. The insidious nature of anthrax has both vegetative and spore morphology. The vegetative state, being the growth phase, is typically responsive to most classes of antibiotics, while the spore phase is not. Plague is caused by a bacterium carried by a rodent flea. While current antibiotics are effective against plague, the worry is the possibility of a bioengineered chimeric construct that would be resistant to all classes of antibiotics. Tularemia is a zoonosis that occurs naturally in the United States, with animal transmission to man. Sometimes an insect vector may also be the primary route of infection. It is highly pathogenic and the inhalation of 10 organisms would be adequate for infection. Smallpox is the most feared of all biowarfare pathogens, primarily due to its high transmissibility versus other pathogens whose etiologic affects are episodic.
Collapse
Affiliation(s)
- Ving J Lee
- Anacor Pharmaceuticals, Inc., Palo Alto, CA 94303, USA
| |
Collapse
|
72
|
Barth H, Aktories K, Popoff MR, Stiles BG. Binary bacterial toxins: biochemistry, biology, and applications of common Clostridium and Bacillus proteins. Microbiol Mol Biol Rev 2004; 68:373-402, table of contents. [PMID: 15353562 PMCID: PMC515256 DOI: 10.1128/mmbr.68.3.373-402.2004] [Citation(s) in RCA: 285] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Certain pathogenic species of Bacillus and Clostridium have developed unique methods for intoxicating cells that employ the classic enzymatic "A-B" paradigm for protein toxins. The binary toxins produced by B. anthracis, B. cereus, C. botulinum, C. difficile, C. perfringens, and C. spiroforme consist of components not physically associated in solution that are linked to various diseases in humans, animals, or insects. The "B" components are synthesized as precursors that are subsequently activated by serine-type proteases on the targeted cell surface and/or in solution. Following release of a 20-kDa N-terminal peptide, the activated "B" components form homoheptameric rings that subsequently dock with an "A" component(s) on the cell surface. By following an acidified endosomal route and translocation into the cytosol, "A" molecules disable a cell (and host organism) via disruption of the actin cytoskeleton, increasing intracellular levels of cyclic AMP, or inactivation of signaling pathways linked to mitogen-activated protein kinase kinases. Recently, B. anthracis has gleaned much notoriety as a biowarfare/bioterrorism agent, and of primary interest has been the edema and lethal toxins, their role in anthrax, as well as the development of efficacious vaccines and therapeutics targeting these virulence factors and ultimately B. anthracis. This review comprehensively surveys the literature and discusses the similarities, as well as distinct differences, between each Clostridium and Bacillus binary toxin in terms of their biochemistry, biology, genetics, structure, and applications in science and medicine. The information may foster future studies that aid novel vaccine and drug development, as well as a better understanding of a conserved intoxication process utilized by various gram-positive, spore-forming bacteria.
Collapse
Affiliation(s)
- Holger Barth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie der Albert-Ludwigs-Universität Freiburg, Otto-Krayer-Haus, Albertstrasse 25, D-79104 Freiburg, Germany.
| | | | | | | |
Collapse
|
73
|
Peinado JR, Kacprzak MM, Leppla SH, Lindberg I. Cross-inhibition between furin and lethal factor inhibitors. Biochem Biophys Res Commun 2004; 321:601-5. [PMID: 15358148 DOI: 10.1016/j.bbrc.2004.07.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Indexed: 11/15/2022]
Abstract
Bacillus anthracis synthesizes two toxins composed of the three proteins: protective antigen (PA), lethal factor (LF), and edema factor (EF). The cleavage of PA on the cell surface by the convertase furin leads to the translocation of LF and EF into the cytosol. We have investigated the cross-inhibitory activities of the furin inhibitors hexa-d-arginine amide (D6R) and nona-d-arginine amide (D9R), which block the proteolytic activation of PA; and of the LF inhibitor In-2-LF, a peptide hydroxamate. D6R and D9R inhibit LF with IC(50s) of 300 and 10microM, respectively; conversely, In-2-LF also inhibits furin (IC(50) 2microM). In-2-LF was efficiently cleaved by furin with the concomitant loss of inhibitory activity on both LF and furin. Incubation of In-2-LF with LF however generated a product that retained partial inhibitory activity against LF. Combined treatment of cells with D6R and In-2-LF enhanced protection against anthrax lethal toxin, indicating that combined administration of inhibitors could represent an effective therapeutic approach.
Collapse
Affiliation(s)
- Juan R Peinado
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
74
|
Abstract
Never before has there been such a strong possibility that biological agents might be used indiscriminately on civilian populations. This review focuses on the use of antitoxins - antibodies, receptor decoys, dominant-negative inhibitors of translocation, small-molecule inhibitors and substrate analogues - to counteract those biological weapons for which toxins are an important mechanism of disease pathogenesis.
Collapse
Affiliation(s)
- G Jonah A Rainey
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037-1099, USA
| | | |
Collapse
|
75
|
Abstract
Bacillus anthracis, the etiological agent of anthrax, secretes three polypeptides that assemble into toxic complexes on the cell surfaces of the host it infects. One of these polypeptides, protective antigen (PA), binds to the integrin-like domains of ubiquitously expressed membrane proteins of mammalian cells. PA is then cleaved by membrane endoproteases of the furin family. Cleaved PA molecules assemble into heptamers, which can then associate with the two other secreted polypeptides: edema factor (EF) and/or lethal factor (LF). The heptamers of PA are relocalized to lipid rafts where they are quickly endocytosed and routed to an acidic compartment. The low pH triggers a conformational change in the heptamers, resulting in the formation of cation-specific channels and the translocation of EF/LF. EF is a calcium- and calmodulin-dependent adenylate cyclase that dramatically raises the intracellular concentration of cyclic adenosine monophosphate (cAMP). LF is a zinc-dependent endoprotease that cleaves the amino terminus of mitogen-activated protein kinase kinases (Meks). Cleaved Meks cannot bind to their substrates and have reduced kinase activity, resulting in alterations of the signaling pathways they govern. The structures of PA, PA heptamer, EF, and LF have been solved and much is now known about the molecular details of the intoxication mechanism. The in vivo action of the toxins, on the other hand, is still poorly understood and hotly debated. A better understanding of the toxins will help in the design of much-needed anti-toxin drugs and the development of new toxin-based medical applications.
Collapse
Affiliation(s)
- M Mourez
- Faculté de Médecine Vétérinaire, Département de Pathologie et Microbiologie, Université de Montréal, J2S 7C6, Saint Hyacinthe, QC, Canada.
| |
Collapse
|
76
|
Min DH, Tang WJ, Mrksich M. Chemical screening by mass spectrometry to identify inhibitors of anthrax lethal factor. Nat Biotechnol 2004; 22:717-23. [PMID: 15146199 DOI: 10.1038/nbt973] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Accepted: 03/17/2004] [Indexed: 11/09/2022]
Abstract
Mass spectrometry (MS) analysis is applicable to a broad range of biological analytes and has the important advantage that it does not require analytes to be labeled. A drawback of MS methods, however, is the need for chromatographic steps to prepare the analyte, precluding MS from being used in chemical screening and rapid analysis. Here, we report that surfaces that are chemically tailored for characterization by matrix-assisted laser-desorption ionization time-of-flight MS eliminate the need for sample processing and make this technique adaptable to parallel screening experiments. The tailored substrates are based on self-assembled monolayers that present ligands that interact with target proteins and enzymes. We apply this method to screen a chemical library against protease activity of anthrax lethal factor, and report a compound that inhibits lethal factor activity with a K(i) of 1.1 microM and blocks the cleavage of MEK1 in 293 cells.
Collapse
Affiliation(s)
- Dal-Hee Min
- Department of Chemistry, Institute for Biophysical Dynamics, The University of Chicago, Illinois 60637, USA
| | | | | |
Collapse
|
77
|
Lee LV, Bower KE, Liang FS, Shi J, Wu D, Sucheck SJ, Vogt PK, Wong CH. Inhibition of the Proteolytic Activity of Anthrax Lethal Factor by Aminoglycosides. J Am Chem Soc 2004; 126:4774-5. [PMID: 15080670 DOI: 10.1021/ja0495359] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The anthrax lethal factor (LF), a Zn-dependent endopeptidase, is considered the dominant virulence factor of anthrax. Because pharmacological inhibition of the catalytic activity of LF is considered a plausible mechanism for preventing the lethality of anthrax, a high-throughput screening experiment based on LF-catalyzed cleavage of a fluorescent substrate was performed to identify novel inhibitors of LF. The RNA-targeting antibiotics, neomycin B and some synthetic dimeric aminoglycosides, were found to be nanomolar active-site inhibitors of LF.
Collapse
Affiliation(s)
- Lac V Lee
- Department of Chemistry and the Skaggs Institute for Chemical Biology, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Dell'Aica I, Donà M, Tonello F, Piris A, Mock M, Montecucco C, Garbisa S. Potent inhibitors of anthrax lethal factor from green tea. EMBO Rep 2004; 5:418-22. [PMID: 15031715 PMCID: PMC1299029 DOI: 10.1038/sj.embor.7400118] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2003] [Revised: 01/27/2004] [Accepted: 01/28/2004] [Indexed: 11/09/2022] Open
Abstract
The anthrax lethal factor (LF) has a major role in the development of anthrax. LF is delivered by the protective antigen (PA) inside the cell, where it exerts its metalloprotease activity on the N-terminus of MAPK-kinases. PA+LF are cytotoxic to macrophages in culture and kill the Fischer 344 rat when injected intravenously. We describe here the properties of some polyphenols contained in green tea as powerful inhibitors of LF metalloproteolytic activity, and how the main catechin of green tea, (-)epigallocatechin-3-gallate, prevents the LF-induced death of macrophages and Fischer 344 rats.
Collapse
Affiliation(s)
| | - Massimo Donà
- Dipartimento di Scienze Biomediche, Università di Padova, Padova, Italy
| | | | - Alejandro Piris
- Toxines et Pathogénie Bactérienne (CNRS URA 2172), Institut Pasteur, Paris, France
| | - Michèle Mock
- Toxines et Pathogénie Bactérienne (CNRS URA 2172), Institut Pasteur, Paris, France
| | - Cesare Montecucco
- Dipartimento di Scienze Biomediche, Università di Padova, Padova, Italy
- Tel: +39 049 8276058; Fax: +39 049 8276049; E-mail:
| | | |
Collapse
|
79
|
Shen Y, Zhukovskaya NL, Zimmer MI, Soelaiman S, Bergson P, Wang CR, Gibbs CS, Tang WJ. Selective inhibition of anthrax edema factor by adefovir, a drug for chronic hepatitis B virus infection. Proc Natl Acad Sci U S A 2004; 101:3242-7. [PMID: 14978283 PMCID: PMC365774 DOI: 10.1073/pnas.0306552101] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Edema factor (EF), a key virulence factor in anthrax pathogenesis, has calmodulin (CaM)-activated adenylyl cyclase activity. We have found that adefovir dipivoxil, a drug approved to treat chronic infection of hepatitis B virus, effectively inhibits EF-induced cAMP accumulation and changes in cytokine production in mouse primary macrophages. Adefovir diphosphate (PMEApp), the active cellular metabolite of adefovir dipivoxil, inhibits the adenylyl cyclase activity of EF in vitro with high affinity (K(i) = 27 nM). A crystal structure of EF-CaM-PMEApp reveals that the catalytic site of EF forms better van der Waals contacts and more hydrogen bonds with PMEApp than with its endogenous substrate, ATP, providing an explanation for the approximately 10,000-fold higher affinity EF-CaM has for PMEApp versus ATP. Adefovir dipivoxil is a clinically approved drug that can block the action of an anthrax toxin. It can be used to address the role of EF in anthrax pathogenesis.
Collapse
Affiliation(s)
- Yuequan Shen
- Ben-May Institute for Cancer Research, University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
Anthrax toxin consists of three nontoxic proteins that associate in binary or ternary combinations to form toxic complexes at the surface of mammalian cells. One of these proteins, protective antigen (PA), transports the other two, edema factor (EF) and lethal factor (LF), to the cytosol. LF is a Zn2+-protease that cleaves certain MAP kinase kinases, leading to death of the host via a poorly defined sequence of events. EF, a calmodulin- and Ca2+-dependent adenylate cyclase, is responsible for the edema seen in the disease. Both enzymes are believed to benefit the bacteria by inhibiting cells of the host's innate immune system. Assembly of toxic complexes begins after PA binds to cellular receptors and is cleaved into two fragments by furin proteases. The smaller fragment dissociates, allowing the receptor-bound fragment, PA63 (63 kDa), to self-associate and form a ring-shaped, heptameric pore precursor (prepore). The prepore binds up to three molecules of EF and/or LF, and the resulting complexes are endocytosed and trafficked to an acidic compartment. There, the prepore converts to a transmembrane pore, mediating translocation of EF and LF to the cytosol. Recent studies have revealed (a) the identity of receptors; (b) crystallographic structures of the three toxin proteins and the heptameric PA63 prepore; and (c) information about toxin assembly, entry, and action within the cytosol. Knowledge of the structure and mode of action of the toxin has unveiled potential applications in medicine, including approaches to treating anthrax infections.
Collapse
Affiliation(s)
- R John Collier
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
81
|
Sarac MS, Peinado JR, Leppla SH, Lindberg I. Protection against anthrax toxemia by hexa-D-arginine in vitro and in vivo. Infect Immun 2004; 72:602-5. [PMID: 14688144 PMCID: PMC343991 DOI: 10.1128/iai.72.1.602-605.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The anthrax toxin protective antigen precursor is activated by proteolytic cleavage by furin or a furin-like protease. We present here data demonstrating that the small stable furin inhibitor hexa-D-arginine amide delays anthrax toxin-induced toxemia both in cells and in live animals, suggesting that furin inhibition may represent a reasonable avenue for therapeutic intervention in anthrax.
Collapse
Affiliation(s)
- Miroslav S Sarac
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | |
Collapse
|
82
|
Tonello F, Naletto L, Romanello V, Dal Molin F, Montecucco C. Tyrosine-728 and glutamic acid-735 are essential for the metalloproteolytic activity of the lethal factor of Bacillus anthracis. Biochem Biophys Res Commun 2004; 313:496-502. [PMID: 14697216 DOI: 10.1016/j.bbrc.2003.11.134] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The lethal factor (LF) of Bacillus anthracis is a Zn2+-endopeptidase specific for the MAPK-kinase family of proteins. The catalytic zinc atom is coordinated by a first shell of residues including the two histidines and the glutamate of the zinc-binding motif HExxH and by Glu-735. A characteristic feature of LF is the presence, within the second shell of residues, of a tyrosine (Tyr-728) in close proximity (3.3 A) to the zinc atom. To investigate the role of Tyr-728 and Glu-735, LF mutants with one or both of these two residues replaced by Ala were cloned, expressed, and purified from Escherichia coli. A fourth mutant was obtained by replacing Tyr-728 with Phe. Spectroscopic analysis of these mutants indicates that they fold in the same way as the parental molecule and that zinc stabilizes the structure of LF. These mutants have neither proteolytic activity nor in vivo toxicity. The possible role of Tyr-728 in catalysis is discussed.
Collapse
Affiliation(s)
- Fiorella Tonello
- Istituto di Neuroscienze del CNR, Università di Padova, Via G Colombo 3, 35121 Padua, Italy.
| | | | | | | | | |
Collapse
|
83
|
Turk BE, Wong TY, Schwarzenbacher R, Jarrell ET, Leppla SH, Collier RJ, Liddington RC, Cantley LC. The structural basis for substrate and inhibitor selectivity of the anthrax lethal factor. Nat Struct Mol Biol 2003; 11:60-6. [PMID: 14718924 DOI: 10.1038/nsmb708] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2003] [Accepted: 10/23/2003] [Indexed: 11/09/2022]
Abstract
Recent events have created an urgent need for new therapeutic strategies to treat anthrax. We have applied a mixture-based peptide library approach to rapidly determine the optimal peptide substrate for the anthrax lethal factor (LF), a metalloproteinase with an important role in the pathogenesis of the disease. Using this approach we have identified peptide analogs that inhibit the enzyme in vitro and that protect cultured macrophages from LF-mediated cytolysis. The crystal structures of LF bound to an optimized peptide substrate and to peptide-based inhibitors provide a rationale for the observed selectivity and may be exploited in the design of future generations of LF inhibitors.
Collapse
Affiliation(s)
- Benjamin E Turk
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Panchal RG, Hermone AR, Nguyen TL, Wong TY, Schwarzenbacher R, Schmidt J, Lane D, McGrath C, Turk BE, Burnett J, Aman MJ, Little S, Sausville EA, Zaharevitz DW, Cantley LC, Liddington RC, Gussio R, Bavari S. Identification of small molecule inhibitors of anthrax lethal factor. Nat Struct Mol Biol 2003; 11:67-72. [PMID: 14718925 DOI: 10.1038/nsmb711] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2003] [Accepted: 10/30/2003] [Indexed: 12/14/2022]
Abstract
The virulent spore-forming bacterium Bacillus anthracis secretes anthrax toxin composed of protective antigen (PA), lethal factor (LF) and edema factor (EF). LF is a Zn-dependent metalloprotease that inactivates key signaling molecules, such as mitogen-activated protein kinase kinases (MAPKK), to ultimately cause cell death. We report here the identification of small molecule (nonpeptidic) inhibitors of LF. Using a two-stage screening assay, we determined the LF inhibitory properties of 19 compounds. Here, we describe six inhibitors on the basis of a pharmacophoric relationship determined using X-ray crystallographic data, molecular docking studies and three-dimensional (3D) database mining from the US National Cancer Institute (NCI) chemical repository. Three of these compounds have K(i) values in the 0.5-5 microM range and show competitive inhibition. These molecular scaffolds may be used to develop therapeutically viable inhibitors of LF.
Collapse
Affiliation(s)
- Rekha G Panchal
- Developmental Therapeutics Program, NCI Frederick, Frederick, Maryland 21702-1201, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Tonello F, Ascenzi P, Montecucco C. The metalloproteolytic activity of the anthrax lethal factor is substrate-inhibited. J Biol Chem 2003; 278:40075-8. [PMID: 12888555 DOI: 10.1074/jbc.m306466200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The anthrax lethal factor (LF) is a Zn2+ endopeptidase specific for mitogen-activated protein kinase kinases (MAPKKs), which are cleaved within their N termini. Here, the proteolytic activity of LF has been investigated using novel chromogenic MAPKK-derived peptide substrates, which allowed us to determine the kinetic parameters of the reaction. LF displayed maximal proteolytic activity at the pH and temperature values of the cell cytosol, which is its site of action. LF undergoes substrate inhibition, in keeping with the non-productive binding geometry of the MAPPK-2 N terminus to LF.
Collapse
Affiliation(s)
- Fiorella Tonello
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche, Via G. Colombo 3, 35121 Padova, Italy.
| | | | | |
Collapse
|
86
|
Rivera VR, Merrill GA, White JA, Poli MA. An enzymatic electrochemiluminescence assay for the lethal factor of anthrax. Anal Biochem 2003; 321:125-30. [PMID: 12963063 DOI: 10.1016/s0003-2697(03)00424-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The lethal factor (LF) of anthrax toxin is the toxic component of the exotoxin (lethal toxin) secreted by toxic strains of Bacillus anthracis. The lethal factor is a zinc-dependent metalloprotease that specifically cleaves the mitogen-activated protein kinase kinase (MAPKK) family of enzymes. We took advantage of this substrate specificity to develop an electrochemiluminescence (ECL) peptide cleavage assay. The ECL assay uses the stable ruthenium (Ru) metal chelate that, in the presence of tripropylamine, generates a light reaction triggered by the application of an electric potential. The Ru label is specifically incorporated into the C-terminal CYS residue of a synthetic peptide (23mer) containing the MAPKK2 cleavage sequence of LF. Streptavidin-coated paramagnetic beads were the solid phase and facilitated separation and characterization of the enzymatic reaction products based upon N-terminal biotinylation of the peptide substrate. Intact peptide bound via the biotin moiety generated high signal due to the Ru label, whereas binding of the cleaved peptide fragment devoid of Ru label reduced the ECL signal. The proposed assay provides a novel opportunity for the screening of potential therapeutics against anthrax.
Collapse
Affiliation(s)
- Victor R Rivera
- Toxinology and Aerobiology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | | | | | | |
Collapse
|
87
|
Haug G, Leemhuis J, Tiemann D, Meyer DK, Aktories K, Barth H. The host cell chaperone Hsp90 is essential for translocation of the binary Clostridium botulinum C2 toxin into the cytosol. J Biol Chem 2003; 278:32266-74. [PMID: 12805360 DOI: 10.1074/jbc.m303980200] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Clostridium botulinum C2 toxin is the prototype of the binary actin-ADP-ribosylating toxins and consists of the binding component C2II and the enzyme component C2I. The activated binding component C2IIa forms heptamers, which bind to carbohydrates on the cell surface and interact with the enzyme component C2I. This toxin complex is taken up by receptor-mediated endocytosis. In acidic endosomes, heptameric C2IIa forms pores and mediates the translocation of C2I into the cytosol. We report that the heat shock protein (Hsp) 90-specific inhibitors, geldanamycin or radicicol, block intoxication of Vero cells, rat astrocytes, and HeLa cells by C2 toxin. ADP-ribosylation of actin in the cytosol of toxin-treated cells revealed that less active C2I was translocated into the cytosol after treatment with Hsp90 inhibitors. Under control conditions, C2I was localized in the cytosol of toxin-treated rat astrocytes, whereas geldanamycin blocked the cytosolic distribution of C2I. At low extracellular pH (pH 4.5), which allows the direct translocation of C2I via C2IIa heptamers across the cell membrane into the cytosol, Hsp90 inhibitors retarded intoxication by C2I. Geldanamycin did not affect toxin binding, endocytosis, and pore formation by C2IIa. The ADP-ribosyltransferase activity of C2I was not affected by Hsp90 inhibitors in vitro. The cytotoxic actions of the actin-ADP-ribosylating Clostridium perfringens iota toxin and the Rho-ADP-ribosylating C2-C3 fusion toxin was similarly blocked by Hsp90 inhibitors. In contrast, radicicol and geldanamycin had no effect on anthrax lethal toxin-induced cytotoxicity of J774-A1 macrophage-like cells or on cytotoxic effects of the glucosylating Clostridium difficile toxin B in Vero cells. The data indicate that Hsp90 is essential for the membrane translocation of ADP-ribosylating toxins delivered by C2II.
Collapse
Affiliation(s)
- Gerd Haug
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie der Albert-Ludwigs-Universität Freiburg, Albertstrasse 25 (Otto-Krayer-Haus), D-79104 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
88
|
Kim J, Kim YM, Koo BS, Chae YK, Yoon MY. Production and proteolytic assay of lethal factor from Bacillus anthracis. Protein Expr Purif 2003; 30:293-300. [PMID: 12880779 DOI: 10.1016/s1046-5928(03)00132-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bacillus anthracis is the causative agent of anthrax. The major virulence factors are a poly-D-glutamic acid capsule and three-protein component exotoxin, protective antigen (PA, 83 kDa), lethal factor (LF, 90 kDa), and edema factor (EF, 89 kDa), respectively. These three proteins individually have no known toxic activities, but in combination with PA form two toxins (lethal toxin or edema toxin), causing different pathogenic responses in animals and cultured cells. In this study, we constructed and produced rLF as a form of GST fusion protein in Escherichia coli. rLF was rapidly purified through a single affinity purification step to near homogeneity. Furthermore, we developed an in vitro immobilized proteolytic assay of LF under the condition containing full-length native substrate, MEK1, rather than short synthetic peptide. The availability of full-length substrate and of an immobilized LF assay could facilitate not only the in-depth investigation of structure-function relationship of the enzyme toward its substrate but also wide spectrum screening of inhibitor collections based on the 96-well plate system.
Collapse
Affiliation(s)
- Joungmok Kim
- Department of Chemistry, Hanyang University, 133-791 Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
89
|
Soelaiman S, Wei BQ, Bergson P, Lee YS, Shen Y, Mrksich M, Shoichet BK, Tang WJ. Structure-based inhibitor discovery against adenylyl cyclase toxins from pathogenic bacteria that cause anthrax and whooping cough. J Biol Chem 2003; 278:25990-7. [PMID: 12676933 DOI: 10.1074/jbc.m301232200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Edema factor (EF) and CyaA are adenylyl cyclase toxins secreted by pathogenic bacteria that cause anthrax and whooping cough, respectively. Using the structure of the catalytic site of EF, we screened a data base of commercially available, small molecular weight chemicals for those that could specifically inhibit adenylyl cyclase activity of EF. From 24 compounds tested, we have identified one quinazoline compound, ethyl 5-aminopyrazolo[1,5-a]quinazoline-3-carboxylate, that specifically inhibits adenylyl cyclase activity of EF and CyaA with approximately 20 microm Ki. This compound neither affects the activity of host resident adenylyl cyclases type I, II, and V nor exhibits promiscuous inhibition. The compound is a competitive inhibitor, consistent with the prediction that it binds to the adenine portion of the ATP binding site on EF. EF is activated by the host calcium sensor, calmodulin. Surface plasmon resonance spectroscopic analysis shows that this compound does not affect the binding of calmodulin to EF. This compound is dissimilar from a previously described, non-nucleoside inhibitor of host adenylyl cyclase. It may serve as a lead to design antitoxins to address the role of adenylyl cyclase toxins in bacterial pathogenesis and to fight against anthrax and whooping cough.
Collapse
Affiliation(s)
- Sandriyana Soelaiman
- Ben-May Institute for Cancer Research, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Chiavolini D, Memmi G, Maggi T, Iannelli F, Pozzi G, Oggioni MR. The three extra-cellular zinc metalloproteinases of Streptococcus pneumoniae have a different impact on virulence in mice. BMC Microbiol 2003; 3:14. [PMID: 12841855 PMCID: PMC166150 DOI: 10.1186/1471-2180-3-14] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Accepted: 07/03/2003] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Streptococcus pneumoniae possesses large zinc metalloproteinases on its surface. To analyse the importance in virulence of three of these metalloproteinases, intranasal challenge of MF1 outbred mice was carried out using a range of infecting doses of wild type and knock-out pneumococcal mutant strains, in order to compare mice survival. RESULTS Observation of survival percentages over time and detection of LD50s of knock out mutants in the proteinase genes in comparison to the type 4 TIGR4 wild type strain revealed two major aspects: i) Iga and ZmpB, present in all strains of S. pneumoniae, strongly contribute to virulence in mice; (ii) ZmpC, only present in about 25% of pneumococcal strains, has a lower influence on virulence in mice. CONCLUSIONS These data suggest Iga, ZmpB and ZmpC as candidate surface proteins responsible for pneumococcal infection and potentially involved in distinct stages of pneumococcal disease.
Collapse
Affiliation(s)
- Damiana Chiavolini
- Dipartimento di Biologia Molecolare, Laboratorio di Microbiologia Molecolare e Biotecnologia, Università di Siena, Siena, Italy
| | - Guido Memmi
- Dipartimento di Biologia Molecolare, Laboratorio di Microbiologia Molecolare e Biotecnologia, Università di Siena, Siena, Italy
| | - Tiziana Maggi
- Dipartimento di Biologia Molecolare, Laboratorio di Microbiologia Molecolare e Biotecnologia, Università di Siena, Siena, Italy
| | - Francesco Iannelli
- Dipartimento di Biologia Molecolare, Laboratorio di Microbiologia Molecolare e Biotecnologia, Università di Siena, Siena, Italy
| | - Gianni Pozzi
- Dipartimento di Biologia Molecolare, Laboratorio di Microbiologia Molecolare e Biotecnologia, Università di Siena, Siena, Italy
| | - Marco R Oggioni
- Dipartimento di Biologia Molecolare, Laboratorio di Microbiologia Molecolare e Biotecnologia, Università di Siena, Siena, Italy
| |
Collapse
|
91
|
Abstract
Although the dramatic events of the year 2001 have revitalized the interest in anthrax, research on Bacillus anthracis and its major virulence factors is one of the oldest theme in microbiology and started with the early works of Robert Koch and Louis Pasteur. The anthrax toxins are central to anthrax pathogenesis. They were discovered in the mid-1950s and since then there has been an enormous amount of work to elucidate both the molecular and physiopathological details of their mode of action. In this review, after a brief introduction of B. anthracis, we will focus on the latest findings that concern two aspects of anthrax toxin research: the environmental signals and the molecular mechanisms that regulate toxin synthesis, and the mechanisms of intoxication. We hope to convince the reader that the anthrax toxins are highly specialized determinants of B. anthracis pathogenicity: their synthesis is integrated within a global virulence programme and they target key eukaryotic cell proteins. We conclude with a consideration of the therapeutic perspectives arising from our current knowledge of how the toxins work.
Collapse
Affiliation(s)
- Michèle Mock
- Toxines et Pathogénie Bactériennes (URA 2172, CNRS), Institut Pasteur, 28 rue du Dr Roux, 75724, Paris cedex 15, France.
| | | |
Collapse
|
92
|
Abstract
Bacillus anthracis, the agent responsible for inhalation anthrax, exerts its lethal effects via the production of anthrax toxin (protective antigen, lethal factor and oedema factor); anthrax kills because the toxin overwhelms the patient before innate host defence systems have a chance to eradicate the invaders. Structural studies on these three components provide a starting point for the design of novel drugs to neutralize toxin action, yielding a valuable adjunct to conventional antibiotic treatment.
Collapse
Affiliation(s)
- Milton T Stubbs
- Institut für Biotechnologie, Martin-Luther-Universität Halle-Wittenberg, Kurt-Mothes-Strasse 3, D-06120 Halle an der Saale, Germany.
| |
Collapse
|
93
|
Ascenzi P, Visca P, Ippolito G, Spallarossa A, Bolognesi M, Montecucco C. Anthrax toxin: a tripartite lethal combination. FEBS Lett 2002; 531:384-8. [PMID: 12435580 DOI: 10.1016/s0014-5793(02)03609-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Anthrax is a severe bacterial infection that occurs when Bacillus anthracis spores gain access into the body and germinate in macrophages, causing septicemia and toxemia. Anthrax toxin is a binary A-B toxin composed of protective antigen (PA), lethal factor (LF), and edema factor (EF). PA mediates the entry of either LF or EF into the cytosol of host cells. LF is a zinc metalloprotease that inactivates mitogen-activated protein kinase kinase inducing cell death, and EF is an adenylyl cyclase impairing host defences. Inhibitors targeting different steps of toxin activity have recently been developed. Anthrax toxin has also been exploited as a therapeutic agent against cancer.
Collapse
Affiliation(s)
- Paolo Ascenzi
- Department of Biology and Interdepartmental Laboratory for Electron Microscopy, University Roma Tre, Viale G. Marconi 446, 00146, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
94
|
In Brief. Nat Rev Drug Discov 2002. [DOI: 10.1038/nrd905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|