1
|
Tessier E, Cheutin L, Garnier A, Vigne C, Tournier JN, Rougeaux C. Early Circulating Edema Factor in Inhalational Anthrax Infection: Does It Matter? Microorganisms 2024; 12:308. [PMID: 38399712 PMCID: PMC10891819 DOI: 10.3390/microorganisms12020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Anthrax toxins are critical virulence factors of Bacillus anthracis and Bacillus cereus strains that cause anthrax-like disease, composed of a common binding factor, the protective antigen (PA), and two enzymatic proteins, lethal factor (LF) and edema factor (EF). While PA is required for endocytosis and activity of EF and LF, several studies showed that these enzymatic factors disseminate within the body in the absence of PA after intranasal infection. In an effort to understand the impact of EF in the absence of PA, we used a fluorescent EF chimera to facilitate the study of endocytosis in different cell lines. Unexpectedly, EF was found inside cells in the absence of PA and showed a pole-dependent endocytosis. However, looking at enzymatic activity, PA was still required for EF to induce an increase in intracellular cAMP levels. Interestingly, the sequential delivery of EF and then PA rescued the rise in cAMP levels, indicating that PA and EF may functionally associate during intracellular trafficking, as well as it did at the cell surface. Our data shed new light on EF trafficking and the potential location of PA and EF association for optimal cytosolic delivery.
Collapse
Affiliation(s)
- Emilie Tessier
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Laurence Cheutin
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Annabelle Garnier
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Clarisse Vigne
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| | - Jean-Nicolas Tournier
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
- Institut Pasteur, 75015 Paris, France
| | - Clémence Rougeaux
- Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France (C.R.)
| |
Collapse
|
2
|
Karimi F, Dabbagh S. Gel green fluorescence ssDNA aptasensor based on carbon nanotubes for detection of anthrax protective antigen. Int J Biol Macromol 2019; 140:842-850. [PMID: 31470050 DOI: 10.1016/j.ijbiomac.2019.08.219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/16/2019] [Accepted: 08/26/2019] [Indexed: 01/09/2023]
Abstract
Bacillus anthracis, the causative agent of anthrax, is a harmful pathogen with potential ability as a biological weapon which persuades scientists to develop novel methods to detect anthrax from infected resources. In this study, a multi-walled carbon nanotube (MWCNTs)-based fluorescence aptasensor was fabricated to detect the recombinant protective antigen domain 4 (rPAD4) of Bacillus anthracis as the most important key factor in development of anthrax. First, PAD4 was recombinant expressed in E. coli and purified by Ni-NTA column. Second, the affinity of aptamer to rPAD4 was confirmed by ELAA assay. In aptasensor design, the aptamer was labeled with Gel Green and immobilized on MWCNTs. Upon the adsorption of labeled aptamer on MWCNTs, fluorescence emission was quenched. In contrast, by adding rPAD4 to hybridization reaction and incubation for 10 min, the fluorescence emission was significantly recovered to 85% compared to the control. Detection limit for the sensitivity and specificity of the aptasensor was determined 20 ng/ml and 62.5 ng/ml purified and unpurified rPAD4 protein, respectively. Also, applicability of aptasensor was showed in mouse serum sample. Finally, results indicated that nanosensor has the potential to be developed as a high-sensitive, cost-effective and fast-acting system for measuring of PA in anthrax diagnostic tests.
Collapse
Affiliation(s)
- Farrokh Karimi
- Department of Biotechnology, Faculty of Science, University of Maragheh, P.O. Box 55181-83111, Maragheh, Iran.
| | - Somayyeh Dabbagh
- Department of Biotechnology, Faculty of Science, University of Maragheh, P.O. Box 55181-83111, Maragheh, Iran
| |
Collapse
|
3
|
Mamillapalli S, Miyagi M, Bann JG. Stability of domain 4 of the anthrax toxin protective antigen and the effect of the VWA domain of CMG2 on stability. Protein Sci 2016; 26:355-364. [PMID: 27874231 DOI: 10.1002/pro.3087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/04/2023]
Abstract
The major immunogenic component of the current anthrax vaccine, anthrax vaccine adsorbed (AVA) is protective antigen (PA). We have shown recently that the thermodynamic stability of PA can be significantly improved by binding to the Von-Willebrand factor A (VWA) domain of capillary morphogenesis protein 2 (CMG2), and improvements in thermodynamic stability may improve storage and long-term stability of PA for use as a vaccine. In order to understand the origin of this increase in stability, we have isolated the receptor binding domain of PA, domain 4 (D4), and have studied the effect of the addition of CMG2 on thermodynamic stability. We are able to determine a binding affinity between D4 and CMG2 (∼300 nM), which is significantly weaker than that between full-length PA and CMG2 (170-300 pM). Unlike full-length PA, we observe very little change in stability of D4 on binding to CMG2, using either fluorescence or 19 F-NMR experiments. Because in previous experiments we could observe a stabilization of both domain 4 and domain 2, the mechanism of stabilization of PA by CMG2 is likely to involve a mutual stabilization of these two domains.
Collapse
Affiliation(s)
| | - Masaru Miyagi
- Case Center for Proteomics and Bioinformatics, Department of Pharmacology and Department of Opthamology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, 44106
| | - James G Bann
- Department of Chemistry, Wichita State University, Wichita, Kansas, 67260
| |
Collapse
|
4
|
Rabideau AE, Pentelute BL. Delivery of Non-Native Cargo into Mammalian Cells Using Anthrax Lethal Toxin. ACS Chem Biol 2016; 11:1490-501. [PMID: 27055654 DOI: 10.1021/acschembio.6b00169] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The intracellular delivery of peptide and protein therapeutics is a major challenge due to the plasma membrane, which acts as a barrier between the extracellular environment and the intracellular milieu. Over the past two decades, a nontoxic PA/LFN delivery platform derived from anthrax lethal toxin has been developed for the transport of non-native cargo into the cytosol of cells in order to understand the translocation process through a protective antigen (PA) pore and to probe intracellular biological functions. Enzyme-mediated ligation using sortase A and native chemical ligation are two facile methods used to synthesize these non-native conjugates, inaccessible by recombinant technology. Cargo molecules that translocate efficiently include enzymes from protein toxins, antibody mimic proteins, and peptides of varying lengths and non-natural amino acid compositions. The PA pore has been found to effectively convey over 30 known cargos other than native lethal factor (LF; i.e., non-native) with diverse sequences and functionalities on the LFN transporter protein. All together these studies demonstrated that non-native cargos must adopt an unfolded or extended conformation and contain a suitable charge composition in order to efficiently pass through the PA pore. This review provides insight into design parameters for the efficient delivery of new cargos using PA and LFN.
Collapse
Affiliation(s)
- Amy E. Rabideau
- Massachusetts Institute of Technology, Department of Chemistry, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Bradley Lether Pentelute
- Massachusetts Institute of Technology, Department of Chemistry, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
5
|
Zou J, Xu L, Ju Y, Zhang P, Wang Y, Zhang B. Cholesterol depletion induces ANTXR2-dependent activation of MMP-2 via ERK1/2 phosphorylation in neuroglioma U251 cell. Biochem Biophys Res Commun 2014; 452:186-90. [DOI: 10.1016/j.bbrc.2014.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/02/2014] [Indexed: 01/08/2023]
|
6
|
Markle JGM, Frank DN, Adeli K, von Bergen M, Danska JS. Microbiome manipulation modifies sex-specific risk for autoimmunity. Gut Microbes 2014; 5:485-93. [PMID: 25007153 DOI: 10.4161/gmic.29795] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Despite growing evidence for a causal role of environmental factors in autoimmune diseases including the rise in disease frequencies over the past several decades we lack an understanding of how particular environmental exposures modify disease risk. In addition, many autoimmune diseases display sex-biased incidence, with females being disproportionately affected but the mechanisms underlying this sex bias remain elusive. Emerging evidence suggests that both host metabolism and immune function is crucially regulated by the intestinal microbiome. Recently, we showed that in the non-obese diabetic (NOD) mouse model of Type 1 Diabetes (T1D), the gut commensal microbial community strongly impacts the pronounced sex bias in T1D risk by controlling serum testosterone and metabolic phenotypes (1). Here we present new data in the NOD model that explores the correlations between microbial phylogeny, testosterone levels, and metabolic phenotypes, and discuss the future of microbiome-centered analysis and microbe-based therapeutic approaches in autoimmune diseases.
Collapse
Affiliation(s)
- Janet G M Markle
- Department of Immunology; University of Toronto; Toronto, ON Canada; Program in Genetics and Genomic Biology; The Hospital for Sick Children; Toronto, ON Canada
| | - Daniel N Frank
- Division of Infectious Diseases; University of Colorado; Aurora, CO USA
| | - Khosrow Adeli
- Department of Laboratory Medicine; The Hospital for Sick Children; Toronto, ON Canada; Department of Biochemistry; University of Toronto; Toronto, ON Canada
| | - Martin von Bergen
- Department of Metabolomics and Department of Proteomics; Helmholtz Center for Environmental Research; Leipzig, Germany; Department of Biotechnology, Chemistry, and Environmental Engineering; Aalborg University; Aalborg, Denmark
| | - Jayne S Danska
- Department of Immunology; University of Toronto; Toronto, ON Canada; Program in Genetics and Genomic Biology; The Hospital for Sick Children; Toronto, ON Canada; Department of Medical Biophysics; University of Toronto; Toronto, ON Canada
| |
Collapse
|
7
|
Tournier JN, Ulrich RG, Quesnel-Hellmann A, Mohamadzadeh M, Stiles BG. Anthrax, toxins and vaccines: a 125-year journey targetingBacillus anthracis. Expert Rev Anti Infect Ther 2014; 7:219-36. [DOI: 10.1586/14787210.7.2.219] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
8
|
Markle JGM, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ, Danska JS. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 2013; 339:1084-8. [PMID: 23328391 DOI: 10.1126/science.1233521] [Citation(s) in RCA: 1448] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Microbial exposures and sex hormones exert potent effects on autoimmune diseases, many of which are more prevalent in women. We demonstrate that early-life microbial exposures determine sex hormone levels and modify progression to autoimmunity in the nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D). Colonization by commensal microbes elevated serum testosterone and protected NOD males from T1D. Transfer of gut microbiota from adult males to immature females altered the recipient's microbiota, resulting in elevated testosterone and metabolomic changes, reduced islet inflammation and autoantibody production, and robust T1D protection. These effects were dependent on androgen receptor activity. Thus, the commensal microbial community alters sex hormone levels and regulates autoimmune disease fate in individuals with high genetic risk.
Collapse
Affiliation(s)
- Janet G M Markle
- Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Anthrax lethal toxin and the induction of CD4 T cell immunity. Toxins (Basel) 2012; 4:878-99. [PMID: 23162703 PMCID: PMC3496994 DOI: 10.3390/toxins4100878] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 10/08/2012] [Accepted: 10/10/2012] [Indexed: 12/27/2022] Open
Abstract
Bacillus anthracis secretes exotoxins which act through several mechanisms including those that can subvert adaptive immunity with respect both to antigen presenting cell and T cell function. The combination of Protective Antigen (PA) and Lethal Factor (LF) forming Lethal Toxin (LT), acts within host cells to down-regulate the mitogen activated protein kinase (MAPK) signaling cascade. Until recently the MAPK kinases were the only known substrate for LT; over the past few years it has become evident that LT also cleaves Nlrp1, leading to inflammasome activation and macrophage death. The predicted downstream consequences of subverting these important cellular pathways are impaired antigen presentation and adaptive immunity. In contrast to this, recent work has indicated that robust memory T cell responses to B. anthracis antigens can be identified following natural anthrax infection. We discuss how LT affects the adaptive immune response and specifically the identification of B. anthracis epitopes that are both immunogenic and protective with the potential for inclusion in protein sub-unit based vaccines.
Collapse
|
10
|
Rajapaksha M, Lovell S, Janowiak BE, Andra KK, Battaile KP, Bann JG. pH effects on binding between the anthrax protective antigen and the host cellular receptor CMG2. Protein Sci 2012; 21:1467-80. [PMID: 22855243 DOI: 10.1002/pro.2136] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/25/2012] [Accepted: 07/26/2012] [Indexed: 11/11/2022]
Abstract
The anthrax protective antigen (PA) binds to the host cellular receptor capillary morphogenesis protein 2 (CMG2) with high affinity. To gain a better understanding of how pH may affect binding to the receptor, we have investigated the kinetics of binding as a function of pH to the full-length monomeric PA and to two variants: a 2-fluorohistidine-labeled PA (2-FHisPA), which is ∼1 pH unit more stable to variations in pH than WT, and an ∼1 pH unit less stable variant in which Trp346 in the domain 2β(3) -2β(4) loop is substituted with a Phe (W346F). We show using stopped-flow fluorescence that the binding rate increases as the pH is lowered for all proteins, with little influence on the rate of dissociation. In addition, we have crystallized PA and the two variants and examine the influence of pH on structure. In contrast to previous X-ray studies, the domain 2β(3) -2β(4) loop undergoes little change in structure from pH ∼8 to 5.5 for the WT protein, but for the 2-FHis labeled and W346F mutant there are changes in structure consistent with previous X-ray studies. In accord with pH stability studies, we find that the average B-factor values increase by ∼20-30% for all three proteins at low pH. Our results suggest that for the full-length PA, low pH increases the binding affinity, likely through a change in structure that favors a more "bound-like" conformation.
Collapse
|
11
|
Bann JG. Anthrax toxin protective antigen--insights into molecular switching from prepore to pore. Protein Sci 2012; 21:1-12. [PMID: 22095644 DOI: 10.1002/pro.752] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The protective antigen is a key component of the anthrax toxin, as it allows entry of the enzymatic components edema factor and lethal factor into the host cell, through the formation of a membrane spanning pore. This event is absolutely critical for the pathogenesis of anthrax, and although we have yet to understand the mechanism of pore formation, recent developments have provided key insights into how this process may occur. Based on the available data, a model is proposed for the kinetic steps for protective antigen conversion from prepore to pore. In this model, the driving force for pore formation is the formation of the phi (ϕ)-clamp, a region that forms a leak-free seal around the translocating polypeptide. Formation of the ϕ-clamp elicits movements within the prepore that provide steric freedom for the subsequent conformational changes required to form the membrane spanning pore.
Collapse
Affiliation(s)
- James G Bann
- Department of Chemistry, Wichita State University, Wichita, Kansas 67260-0051, USA.
| |
Collapse
|
12
|
Reeves CV, Wang X, Charles-Horvath PC, Vink JY, Borisenko VY, Young JAT, Kitajewski JK. Anthrax toxin receptor 2 functions in ECM homeostasis of the murine reproductive tract and promotes MMP activity. PLoS One 2012; 7:e34862. [PMID: 22529944 PMCID: PMC3328497 DOI: 10.1371/journal.pone.0034862] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 03/08/2012] [Indexed: 11/19/2022] Open
Abstract
Anthrax Toxin Receptor proteins function as receptors for anthrax toxin, however physiological activity remains unclear. To evaluate the biological role of Antxr2, we generated Antxr2-/- mice. Antxr2-/- mice were viable, however Antxr2 is required for parturition in young females and for preserving fertility in older female mice. Histological analysis of the uterus and cervix revealed aberrant deposition of extracellular matrix proteins such as type I collagen, type VI collagen and fibronectin. A marked disruption of both the circular and longitudinal myometrial cell layers was evident in Antxr2-/- mice. These changes progressed as the mice aged, resulting in a thickened, collagen dense, acellular stroma and the disappearance of normal uterine architecture. To investigate the molecular mechanism underlying the uterine fibrosis we performed immunoblotting for MMP2 using uterine lysates and zymography using conditioned medium from Antxr2-/- mouse embryonic fibroblasts and found reduced levels of activated MMP2 in both. This prompted us to investigate MT1-MMP status, as MMP2 processing is regulated by MT1-MMP. We found MT1-MMP activity, as measured by MMP2 processing and activation, was enhanced by expression of either ANTXR1 or ANTXR2. We identified an ANTXR2/MT1-MMP complex and demonstrated that MT1-MMP activity is dependent on ANTXR2 expression levels in cells. Thus, we have discovered that ANTXR1 and ANTXR2 function as positive regulators of MT1-MMP activity.
Collapse
Affiliation(s)
- Claire V. Reeves
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York, United States of America
| | - Xing Wang
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York, United States of America
| | - Pelisa C. Charles-Horvath
- Department of Pharmacology, Columbia University Medical Center, New York, New York, United States of America
| | - Joy Y. Vink
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York, United States of America
| | - Valeriya Y. Borisenko
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York, United States of America
| | - John A. T. Young
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Jan K. Kitajewski
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York, United States of America
- Department of Pathology, Columbia University Medical Center, New York, New York, United States of America
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
13
|
Thomas D, Naughton J, Cote C, Welkos S, Manchester M, Young JAT. Delayed toxicity associated with soluble anthrax toxin receptor decoy-Ig fusion protein treatment. PLoS One 2012; 7:e34611. [PMID: 22511955 PMCID: PMC3325282 DOI: 10.1371/journal.pone.0034611] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 03/05/2012] [Indexed: 11/21/2022] Open
Abstract
Soluble receptor decoy inhibitors, including receptor-immunogloubulin (Ig) fusion proteins, have shown promise as candidate anthrax toxin therapeutics. These agents act by binding to the receptor-interaction site on the protective antigen (PA) toxin subunit, thereby blocking toxin binding to cell surface receptors. Here we have made the surprising observation that co-administration of receptor decoy-Ig fusion proteins significantly delayed, but did not protect, rats challenged with anthrax lethal toxin. The delayed toxicity was associated with the in vivo assembly of a long-lived complex comprised of anthrax lethal toxin and the receptor decoy-Ig inhibitor. Intoxication in this system presumably results from the slow dissociation of the toxin complex from the inhibitor following their prolonged circulation. We conclude that while receptor decoy-Ig proteins represent promising candidates for the early treatment of B. anthracis infection, they may not be suitable for therapeutic use at later stages when fatal levels of toxin have already accumulated in the bloodstream.
Collapse
Affiliation(s)
- Diane Thomas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - John Naughton
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Christopher Cote
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Susan Welkos
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Marianne Manchester
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
- * E-mail: (MM); (JATY)
| | - John A. T. Young
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail: (MM); (JATY)
| |
Collapse
|
14
|
Feld GK, Kintzer AF, Tang II, Thoren KL, Krantz BA. Domain flexibility modulates the heterogeneous assembly mechanism of anthrax toxin protective antigen. J Mol Biol 2012; 415:159-74. [PMID: 22063095 PMCID: PMC3249527 DOI: 10.1016/j.jmb.2011.10.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 10/01/2011] [Accepted: 10/18/2011] [Indexed: 01/07/2023]
Abstract
The three protein components of anthrax toxin are nontoxic individually, but they form active holotoxin complexes upon assembly. The role of the protective antigen (PA) component of the toxin is to deliver two other enzyme components, lethal factor and edema factor, across the plasma membrane and into the cytoplasm of target cells. PA is produced as a proprotein, which must be proteolytically activated; generally, cell surface activation is mediated by a furin family protease. Activated PA can then assemble into one of two noninterconverting oligomers, a homoheptamer and a homooctamer, which have unique properties. Herein we describe molecular determinants that influence the stoichiometry of PA in toxin complexes. By tethering PA domain 4 (D4) to domain 2 with two different-length cross-links, we can control the relative proportions of PA heptamers and octamers. The longer cross-link favors octamer formation, whereas the shorter one favors formation of the heptamer. X-ray crystal structures of PA (up to 1.45 Å resolution), including these cross-linked PA constructs, reveal that a hinge-like movement of D4 correlates with the relative preference for each oligomeric architecture. Furthermore, we report the conformation of the flexible loop containing the furin cleavage site and show that, for efficient processing, the furin site cannot be moved ~5 or 6 residues within the loop. We propose that there are different orientations of D4 relative to the main body of PA that favor the formation of either the heptamer or the octamer.
Collapse
Affiliation(s)
- Geoffrey K. Feld
- Department of Chemistry, University of California, Berkeley, CA, 94720, U.S.A.
| | | | - Iok I Tang
- California Institute for Quantitative Biomedical Research, University of California, Berkeley, CA, 94720, U.S.A.
| | - Katie L. Thoren
- Department of Chemistry, University of California, Berkeley, CA, 94720, U.S.A.
| | - Bryan A. Krantz
- Department of Chemistry, University of California, Berkeley, CA, 94720, U.S.A.
,California Institute for Quantitative Biomedical Research, University of California, Berkeley, CA, 94720, U.S.A.
,Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, U.S.A.
,Address correspondence to: Bryan Krantz, Ph.D., University of California, Berkeley 492 Stanley Hall, #3220 Berkeley, CA 94720-3220. Phone: 510-666-2788, (B.A.K.)
| |
Collapse
|
15
|
Pilpa RM, Bayrhuber M, Marlett JM, Riek R, Young JAT. A receptor-based switch that regulates anthrax toxin pore formation. PLoS Pathog 2011; 7:e1002354. [PMID: 22174672 PMCID: PMC3234216 DOI: 10.1371/journal.ppat.1002354] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 09/19/2011] [Indexed: 11/19/2022] Open
Abstract
Cellular receptors can act as molecular switches, regulating the sensitivity of microbial proteins to conformational changes that promote cellular entry. The activities of these receptor-based switches are only partially understood. In this paper, we sought to understand the mechanism that underlies the activity of the ANTXR2 anthrax toxin receptor-based switch that binds to domains 2 and 4 of the protective antigen (PA) toxin subunit. Receptor-binding restricts structural changes within the heptameric PA prepore that are required for pore conversion to an acidic endosomal compartment. The transfer cross-saturation (TCS) NMR approach was used to monitor changes in the heptameric PA-receptor contacts at different steps during prepore-to-pore conversion. These studies demonstrated that receptor contact with PA domain 2 is weakened prior to pore conversion, defining a novel intermediate in this pathway. Importantly, ANTXR2 remained bound to PA domain 4 following pore conversion, suggesting that the bound receptor might influence the structure and/or function of the newly formed pore. These studies provide new insights into the function of a receptor-based molecular switch that controls anthrax toxin entry into cells. The bacterium that causes anthrax produces a toxin called anthrax toxin that is largely responsible for causing disease symptoms. The first step in anthrax intoxication involves binding of the toxin to a specific protein, called a receptor, on the cell surface. Receptor-binding acts like a switch to prevent the toxin from forming a pore in a cell membrane until the toxin-receptor complex is taken up into cells and delivered to a specific location (called an endosome) where it is exposed to an “acid bath”. This acidic environment promotes structural changes in the toxin leading to pore formation in the endosomal membrane. In this report, we have studied how the receptor regulates pore formation by following the associated changes in toxin-receptor contacts. These studies have defined a new toxin-receptor intermediate in the pathway leading to pore conversion and demonstrate that the receptor remains bound after pore conversion. Our results provide important new insights into how the receptor regulates anthrax toxin pore formation, information that could be useful for designing new therapeutic strategies to treat this disease.
Collapse
Affiliation(s)
- Rosemarie M. Pilpa
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Monika Bayrhuber
- Structural Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | - John M. Marlett
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Roland Riek
- Structural Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
- * E-mail: (JATY); (RR)
| | - John A. T. Young
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail: (JATY); (RR)
| |
Collapse
|
16
|
Cai C, Che J, Xu L, Guo Q, Kong Y, Fu L, Xu J, Cheng Y, Chen W. Tumor endothelium marker-8 based decoys exhibit superiority over capillary morphogenesis protein-2 based decoys as anthrax toxin inhibitors. PLoS One 2011; 6:e20646. [PMID: 21674060 PMCID: PMC3107238 DOI: 10.1371/journal.pone.0020646] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 05/06/2011] [Indexed: 01/06/2023] Open
Abstract
Anthrax toxin is the major virulence factor produced by Bacillus anthracis. The toxin consists of three protein subunits: protective antigen (PA), lethal factor, and edema factor. Inhibition of PA binding to its receptors, tumor endothelium marker-8 (TEM8) and capillary morphogenesis protein-2 (CMG2) can effectively block anthrax intoxication, which is particularly valuable when the toxin has already been overproduced at the late stage of anthrax infection, thus rendering antibiotics ineffectual. Receptor-like agonists, such as the mammalian cell-expressed von Willebrand factor type A (vWA) domain of CMG2 (sCMG2), have demonstrated potency against the anthrax toxin. However, the soluble vWA domain of TEM8 (sTEM8) was ruled out as an anthrax toxin inhibitor candidate due to its inferior affinity to PA. In the present study, we report that L56A, a PA-binding-affinity-elevated mutant of sTEM8, could inhibit anthrax intoxication as effectively as sCMG2 in Fisher 344 rats. Additionally, pharmacokinetics showed that L56A and sTEM8 exhibit advantages over sCMG2 with better lung-targeting and longer plasma retention time, which may contribute to their enhanced protective ability in vivo. Our results suggest that receptor decoys based on TEM8 are promising anthrax toxin inhibitors and, together with the pharmacokinetic studies in this report, may contribute to the development of novel anthrax drugs.
Collapse
Affiliation(s)
- Chenguang Cai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jinjing Che
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Long Xu
- Laboratory of protein engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Qiang Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yirong Kong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Ling Fu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Junjie Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- * E-mail: (JX) (JX); (YC) (YC); (WC) (WC)
| | - Yuanguo Cheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- * E-mail: (JX) (JX); (YC) (YC); (WC) (WC)
| | - Wei Chen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- * E-mail: (JX) (JX); (YC) (YC); (WC) (WC)
| |
Collapse
|
17
|
Venter PA, Dirksen A, Thomas D, Manchester M, Dawson PE, Schneemann A. Multivalent display of proteins on viral nanoparticles using molecular recognition and chemical ligation strategies. Biomacromolecules 2011; 12:2293-301. [PMID: 21545187 DOI: 10.1021/bm200369e] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multivalent display of heterologous proteins on viral nanoparticles forms a basis for numerous applications in nanotechnology, including vaccine development, targeted therapeutic delivery, and tissue-specific bioimaging. In many instances, precise placement of proteins is required for optimal functioning of the supramolecular assemblies, but orientation- and site-specific coupling of proteins to viral scaffolds remains a significant technical challenge. We have developed two strategies that allow for controlled attachment of a variety of proteins on viral particles using covalent and noncovalent principles. In one strategy, an interaction between domain 4 of anthrax protective antigen and its receptor was used to display multiple copies of a target protein on virus-like particles. In the other, expressed protein ligation and aniline-catalyzed oximation was used to display covalently a model protein. The latter strategy, in particular, yielded nanoparticles that induced potent immune responses to the coupled protein, suggesting potential applications in vaccine development.
Collapse
Affiliation(s)
- P Arno Venter
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
18
|
Constitutive MEK1 activation rescues anthrax lethal toxin-induced vascular effects in vivo. Infect Immun 2010; 78:5043-53. [PMID: 20855511 DOI: 10.1128/iai.00604-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anthrax lethal toxin (LT) increases vascular leakage in a number of mammalian models and in human anthrax disease. Using a zebrafish model, we determined that vascular delivery of LT increased permeability, which was phenocopied by treatment with a selective chemical inhibitor of MEK1 and MEK2 (also known as mitogen-activated protein kinase [MAPK] kinase, MEK, or MKK). Here we investigate further the role of MEK1/phospho-ERK (pERK) in the action of LT. Overexpression of wild-type zebrafish MEK1 at high levels did not induce detrimental effects. However, a constitutively activated version, MEK1(S219D,S223D) (MEK1DD), induced early defects in embryonic development that correlated with increased ERK/MAPK phosphorylation. To bypass these early developmental defects and to provide a genetic tool for examining the action of lethal factor (LF), we generated inducible transgenic zebrafish lines expressing either wild-type or activated MEK1 under the control of a heat shock promoter. Remarkably, induction of MEK1DD transgene expression prior to LT delivery prevented vascular damage, while the wild-type MEK1 line did not. In the presence of both LT and MEK1DD transgene expression, cardiovascular development and function proceeded normally in most embryos. The resistance to microsphere leakage in transgenic animals demonstrated a protective role against LT-induced vascular permeability. A consistent increase in ERK phosphorylation among LT-resistant MEK1DD transgenic animals provided additional confirmation of transgene activation. These findings provide a novel genetic approach to examine mechanism of action of LT in vivo through one of its known targets. This approach may be generally applied to investigate additional pathogen-host interactions and to provide mechanistic insights into host signaling pathways affected by pathogen entry.
Collapse
|
19
|
The structure of tumor endothelial marker 8 (TEM8) extracellular domain and implications for its receptor function for recognizing anthrax toxin. PLoS One 2010; 5:e11203. [PMID: 20585457 PMCID: PMC2887854 DOI: 10.1371/journal.pone.0011203] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 05/20/2010] [Indexed: 12/02/2022] Open
Abstract
Anthrax toxin, which is released from the Gram-positive bacterium Bacillus anthracis, is composed of three proteins: protective antigen (PA), lethal factor (LF), and edema factor (EF). PA binds a receptor on the surface of the target cell and further assembles into a homo-heptameric pore through which EF and LF translocate into the cytosol. Two distinct cellular receptors for anthrax toxin, TEM8/ANTXR1 and CMG2/ANTXR2, have been identified, and it is known that their extracellular domains bind PA with low and high affinities, respectively. Here, we report the crystal structure of the TEM8 extracellular vWA domain at 1.7 Å resolution. The overall structure has a typical integrin fold and is similar to that of the previously published CMG2 structure. In addition, using structure-based mutagenesis, we demonstrate that the putative interface region of TEM8 with PA (consisting of residues 56, 57, and 154–160) is responsible for the PA-binding affinity differences between the two receptors. In particular, Leu56 was shown to be a key factor for the lower affinity of TEM8 towards PA compared with CMG2. Because of its high affinity for PA and low expression in normal tissues, an isolated extracellular vWA domain of the L56A TEM8 variant may serve as a potent antitoxin and a potential therapeutic treatment for anthrax infection. Moreover, as TEM8 is often over-expressed in tumor cells, our TEM8 crystal structure may provide new insights into how to design PA mutants that preferentially target tumor cells.
Collapse
|
20
|
Van Der Goot G, Young JA. Receptors of anthrax toxin and cell entry. Mol Aspects Med 2009; 30:406-12. [PMID: 19732789 PMCID: PMC2783407 DOI: 10.1016/j.mam.2009.08.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Accepted: 08/24/2009] [Indexed: 11/29/2022]
Abstract
Anthrax toxin-receptor interactions are critical for toxin delivery to the host cell cytoplasm. This review summarizes what is known about the molecular details of the protective antigen (PA) toxin subunit interaction with either the ANTXR1 and ANTXR2 cellular receptors, and how receptor-type can dictate the low pH threshold of PA pore formation. The roles played by cellular factors in regulating the endocytosis of toxin-receptor complexes is also discussed.
Collapse
Affiliation(s)
- Gisou Van Der Goot
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, SV-AI extension, Station 15, 1015 Lausanne, Switzerland,
| | - John A.T. Young
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037,
| |
Collapse
|
21
|
Anthrax toxin receptor 2 is expressed in murine and tumor vasculature and functions in endothelial proliferation and morphogenesis. Oncogene 2009; 29:789-801. [PMID: 19901963 PMCID: PMC2939496 DOI: 10.1038/onc.2009.383] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The Capillary Morphogenesis Gene 2 (CMG2) gene encodes an Anthrax toxin receptor (ANTXR2) but the normal physiological function is not known. ANTXR2/CMG2 was originally identified as a result of up-regulation during capillary morphogenesis of endothelial cells cultured in vitro. We explored the hypothesis that key steps of the angiogenic process are either dependent or are influenced by ANTXR2/CMG2 activity. We describe the expression pattern of ANTXR2/CMG2 in several murine tissues and in normal breast and breast tumors. Endothelial expression was found in all of the tissues analyzed, in cultured endothelial cells and in breast tumor vessels; however ANTXR2/CMG2 expression was not restricted to this cell type. To assess potential angiogenic function, we utilized RNA interference to achieve significant reduction of ANTXR2/CMG2 expression in cultured human umbilical venous endothelial cells. Reduced ANTXR2/CMG2 expression resulted in significant inhibition of proliferation and reduced capacity of endothelial cells to form capillary-like networks in vitro, while overexpression of ANTXR2/CMG2 in HUVEC increased proliferation and capillary-like network formation. Little change in migration of endothelial cells was observed upon knockdown or overexpression. We conclude that ANTXR2/CMG2 functions to promote endothelial proliferation and morphogenesis during sprouting angiogenesis, consistent with the endothelial expression of ANTXR2/CMG2 in several vascular beds.
Collapse
|
22
|
Ha SD, Ham B, Mogridge J, Saftig P, Lin S, Kim SO. Cathepsin B-mediated autophagy flux facilitates the anthrax toxin receptor 2-mediated delivery of anthrax lethal factor into the cytoplasm. J Biol Chem 2009; 285:2120-9. [PMID: 19858192 DOI: 10.1074/jbc.m109.065813] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anthrax lethal toxin (LeTx) is a virulence factor secreted by Bacillus anthracis and has direct cytotoxic effects on most cells once released into the cytoplasm. The cytoplasmic delivery of the proteolytically active component of LeTx, lethal factor (LF), is carried out by the transporter component, protective antigen, which interacts with either of two known surface receptors known as anthrax toxin receptor (ANTXR) 1 and 2. We found that the cytoplasmic delivery of LF by ANTXR2 was mediated by cathepsin B (CTSB) and required lysosomal fusion with LeTx-containing endosomes. Also, binding of protective antigen to ANXTR1 or -2 triggered autophagy, which facilitated the cytoplasmic delivery of ANTXR2-associated LF. We found that whereas cells treated with the membrane-permeable CTSB inhibitor CA074-Me- or CTSB-deficient cells had no defect in fusion of LC3-containing autophagic vacuoles with lysosomes, autophagic flux was significantly delayed. These results suggested that the ANTXR2-mediated cytoplasmic delivery of LF was enhanced by CTSB-dependent autophagic flux.
Collapse
Affiliation(s)
- Soon-Duck Ha
- Department of Microbiology and Immunology, Siebens-Drake Research Institute, University of Western Ontario, London, Ontario N6G2V4, Canada
| | | | | | | | | | | |
Collapse
|
23
|
Capillary morphogenesis protein-2 is the major receptor mediating lethality of anthrax toxin in vivo. Proc Natl Acad Sci U S A 2009; 106:12424-9. [PMID: 19617532 DOI: 10.1073/pnas.0905409106] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Anthrax toxin, a major virulence factor of Bacillus anthracis, gains entry into target cells by binding to either of 2 von Willebrand factor A domain-containing proteins, tumor endothelium marker-8 (TEM8) and capillary morphogenesis protein-2 (CMG2). The wide tissue expression of TEM8 and CMG2 suggest that both receptors could play a role in anthrax pathogenesis. To explore the roles of TEM8 and CMG2 in normal physiology, as well as in anthrax pathogenesis, we generated TEM8- and CMG2-null mice and TEM8/CMG2 double-null mice by deleting TEM8 and CMG2 transmembrane domains. TEM8 and CMG2 were found to be dispensable for mouse development and life, but both are essential in female reproduction in mice. We found that the lethality of anthrax toxin for mice is mostly mediated by CMG2 and that TEM8 plays only a minor role. This is likely because anthrax toxin has approximately 11-fold higher affinity for CMG2 than for TEM8. Finally, the CMG2-null mice are also shown to be highly resistant to B. anthracis spore infection, attesting to the importance of both anthrax toxin and CMG2 in anthrax infections.
Collapse
|
24
|
Li G, Qu Y, Cai C, Kong Y, Liu S, Zhang J, Zhao J, Fu L, Xu J, Chen W. The inhibition of the interaction between the anthrax toxin and its cellular receptor by an anti-receptor monoclonal antibody. Biochem Biophys Res Commun 2009; 385:591-5. [PMID: 19486894 DOI: 10.1016/j.bbrc.2009.05.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 05/26/2009] [Indexed: 11/25/2022]
Abstract
The high affinity binding of the anthrax protective antigen (PA) to one of its receptors, capillary morphogenesis protein 2 (CMG2), is essential for the intoxication process of anthrax toxin. To acquire novel research tools to study the PA-CMG2 interaction, we generated several anti-CMG2 monoclonal antibodies (MAbs). We demonstrated that one of the MAbs, 4B5, could inhibit PA-CMG2 binding and could also protect the sensitive cells against an anthrax lethal toxin challenge. We identified the epitope recognized by 4B5 and confirmed that the key residues of the epitope were the residues (119)YI-LK(125) of CMG2. Based on our results, we propose that 4B5 binds to the E122 pocket of CMG2 and interrupts the interaction between the pocket and the PA 2beta3-2beta4 loop. To our knowledge, this is the first report to illustrate that an anti-CMG2 antibody could inhibit the PA-CMG2 interaction and therefore interfere with the intoxication of anthrax toxin.
Collapse
Affiliation(s)
- Guanlin Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rossi Paccani S, Benagiano M, Capitani N, Zornetta I, Ladant D, Montecucco C, D'Elios MM, Baldari CT. The adenylate cyclase toxins of Bacillus anthracis and Bordetella pertussis promote Th2 cell development by shaping T cell antigen receptor signaling. PLoS Pathog 2009; 5:e1000325. [PMID: 19266022 PMCID: PMC2643477 DOI: 10.1371/journal.ppat.1000325] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 02/03/2009] [Indexed: 01/28/2023] Open
Abstract
The adjuvanticity of bacterial adenylate cyclase toxins has been ascribed to their capacity, largely mediated by cAMP, to modulate APC activation, resulting in the expression of Th2–driving cytokines. On the other hand, cAMP has been demonstrated to induce a Th2 bias when present during T cell priming, suggesting that bacterial cAMP elevating toxins may directly affect the Th1/Th2 balance. Here we have investigated the effects on human CD4+ T cell differentiation of two adenylate cyclase toxins, Bacillus anthracis edema toxin (ET) and Bordetella pertussis CyaA, which differ in structure, mode of cell entry, and subcellular localization. We show that low concentrations of ET and CyaA, but not of their genetically detoxified adenylate cyclase defective counterparts, potently promote Th2 cell differentiation by inducing expression of the master Th2 transcription factors, c-maf and GATA-3. We also present evidence that the Th2–polarizing concentrations of ET and CyaA selectively inhibit TCR–dependent activation of Akt1, which is required for Th1 cell differentiation, while enhancing the activation of two TCR–signaling mediators, Vav1 and p38, implicated in Th2 cell differentiation. This is at variance from the immunosuppressive toxin concentrations, which interfere with the earliest step in TCR signaling, activation of the tyrosine kinase Lck, resulting in impaired CD3ζ phosphorylation and inhibition of TCR coupling to ZAP-70 and Erk activation. These results demonstrate that, notwithstanding their differences in their intracellular localization, which result in focalized cAMP production, both toxins directly affect the Th1/Th2 balance by interfering with the same steps in TCR signaling, and suggest that their adjuvanticity is likely to result from their combined effects on APC and CD4+ T cells. Furthermore, our results strongly support the key role of cAMP in the adjuvanticity of these toxins. Colonization by pathogens requires keeping at bay the host immune defenses, at least at the onset of infection. The adenylate cyclase (AC) toxins produced by many pathogenic bacteria assist in this crucial function by catalyzing the production of cAMP, which acts as a potent immunosuppressant. Nevertheless, at low concentrations, these toxins act as adjuvants, enhancing antibody responses to vaccination. We have investigated the molecular basis of the immunomodulatory activities of two AC toxins, Bacillus anthracis edema toxin and Bordetella pertussis CyaA. We show that high toxin concentrations inhibit activation of T lymphocytes, which orchestrate the adaptive immune response against pathogens, whereas low toxin concentrations promote differentiation of helper T lymphocytes to Th2 effectors, which are required for development of antibody-producing cells. Both the immunosuppressant and Th2–driving activities of the toxins are dependent on cAMP. The results demonstrate that, dependent on their concentration, the AC toxins of B. anthracis and B. pertussis evoke distinct responses on target T lymphocytes by differentially modulating antigen receptor signaling, resulting either in suppression of T cell activation or Th2 cell differentiation. These results are of relevance to the evolution of disease in infected individuals and provide novel mechanistic insight into the adjuvanticity of these toxins.
Collapse
Affiliation(s)
| | - Marisa Benagiano
- Department of Internal Medicine and Immunoallergology, University of Florence, Florence, Italy
| | - Nagaja Capitani
- Department of Evolutionary Biology, University of Siena, Siena, Italy
| | - Irene Zornetta
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Mario M. D'Elios
- Department of Internal Medicine and Immunoallergology, University of Florence, Florence, Italy
| | - Cosima T. Baldari
- Department of Evolutionary Biology, University of Siena, Siena, Italy
- * E-mail:
| |
Collapse
|
26
|
Frankel AE, Kuo SR, Dostal D, Watson L, Duesbery NS, Cheng CP, Cheng HJ, Leppla SH. Pathophysiology of anthrax. Front Biosci (Landmark Ed) 2009; 14:4516-24. [PMID: 19273366 DOI: 10.2741/3544] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Infection by Bacillus anthracis in animals and humans results from accidental or intentional exposure, by oral, cutaneous or pulmonary routes, to spores, which are normally present in the soil. Treatment includes administration of antibiotics, vaccination or treatment with antibody to the toxin. A better understanding of the molecular basis of the processes involved in the pathogenesis of anthrax namely, spore germination in macrophages and biological effects of the secreted toxins on heart and blood vessels will lead to improved management of infected animals and patients. Controlling germination will be feasible by inhibiting macrophage paralysis and cell death. On the other hand, the control of terminal hypotension might be achieved by inhibition of cardiomyocyte mitogen-activated protein kinase and stimulation of vessel cAMP.
Collapse
|
27
|
Efficient neutralization of antibody-resistant forms of anthrax toxin by a soluble receptor decoy inhibitor. Antimicrob Agents Chemother 2008; 53:1210-2. [PMID: 19075066 DOI: 10.1128/aac.01294-08] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A soluble receptor decoy inhibitor (RDI), comprised of the extracellular I domain of ANTXR2, is a candidate anthrax therapeutic. Here we show that RDI can effectively neutralize altered forms of the protective antigen toxin subunit that are resistant to 14B7 monoclonal antibody neutralization. These data highlight the potential of RDI to act as an adjunct to existing antibody-based therapies and indicate that inhibitors based on RDI might be useful as a stand-alone treatment against specifically engineered strains of Bacillus anthracis.
Collapse
|
28
|
Manayani DJ, Thomas D, Dryden KA, Reddy V, Siladi ME, Marlett JM, Rainey GJA, Pique ME, Scobie HM, Yeager M, Young JAT, Manchester M, Schneemann A. A viral nanoparticle with dual function as an anthrax antitoxin and vaccine. PLoS Pathog 2007; 3:1422-31. [PMID: 17922572 PMCID: PMC2000967 DOI: 10.1371/journal.ppat.0030142] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 08/13/2007] [Indexed: 11/19/2022] Open
Abstract
The recent use of Bacillus anthracis as a bioweapon has stimulated the search for novel antitoxins and vaccines that act rapidly and with minimal adverse effects. B. anthracis produces an AB-type toxin composed of the receptor-binding moiety protective antigen (PA) and the enzymatic moieties edema factor and lethal factor. PA is a key target for both antitoxin and vaccine development. We used the icosahedral insect virus Flock House virus as a platform to display 180 copies of the high affinity, PA-binding von Willebrand A domain of the ANTXR2 cellular receptor. The chimeric virus-like particles (VLPs) correctly displayed the receptor von Willebrand A domain on their surface and inhibited lethal toxin action in in vitro and in vivo models of anthrax intoxication. Moreover, VLPs complexed with PA elicited a potent toxin-neutralizing antibody response that protected rats from anthrax lethal toxin challenge after a single immunization without adjuvant. This recombinant VLP platform represents a novel and highly effective, dually-acting reagent for treatment and protection against anthrax. Anthrax is caused by the spore-forming, Gram-positive bacterium Bacillus anthracis. The toxic effects of B. anthracis are predominantly due to an AB-type toxin made up of the receptor-binding subunit protective antigen (PA) and two enzymatic subunits called lethal factor and edema factor. Protective immunity to B. anthracis infection is conferred by antibodies against PA, which is the primary component of the current anthrax vaccine. Although the vaccine is safe and effective, it requires multiple injections followed by annual boosters. The development of a well-characterized vaccine that induces immunity after a single injection is an important goal. We developed a reagent that combines the functions of an anthrax antitoxin and vaccine in a single compound. It is based on multivalent display of the anthrax toxin receptor, ANTXR2, on the surface of an insect virus. We demonstrate that the recombinant virus-like particles protect rats from anthrax intoxication and that they induce a potent immune response against lethal toxin when coated with PA. This immune response protected animals against lethal toxin challenge after a single administration without adjuvant. The PA-coated particles have significant advantages as an immunogen compared to monomeric PA and form the basis for development of an improved anthrax vaccine.
Collapse
Affiliation(s)
- Darly J Manayani
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - Diane Thomas
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - Kelly A Dryden
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - Vijay Reddy
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - Marc E Siladi
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - John M Marlett
- The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - G. Jonah A Rainey
- The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Michael E Pique
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - Heather M Scobie
- The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Mark Yeager
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California , United States of America
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California , United States of America
- Division of Cardiovascular Diseases, Scripps Clinic, La Jolla, California, United States of America
| | - John A. T Young
- The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Marianne Manchester
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California , United States of America
| | - Anette Schneemann
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California , United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
29
|
Yan M, Roehrl MH, Basar E, Wang JY. Selection and evaluation of the immunogenicity of protective antigen mutants as anthrax vaccine candidates. Vaccine 2007; 26:947-55. [PMID: 18192092 DOI: 10.1016/j.vaccine.2007.11.087] [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] [Received: 07/30/2007] [Revised: 11/28/2007] [Accepted: 11/29/2007] [Indexed: 11/17/2022]
Abstract
Protective antigen (PA) is a central component of anthrax toxin and a major antigen in anthrax vaccines. However, the use of native PA as a vaccine is not optimal. If administered to people who have been freshly exposed to anthrax, PA may actually aid in anthrax toxin formation and thus may pose a serious safety concern for postexposure vaccination applications. A non-functional PA mutant may be a much safer alternative. To identify an improved anthrax vaccine antigen, we examined four non-functional mutants of PA, each being impaired in a critical step of the cellular intoxication pathway of PA. These mutants were Rec(-) (unable to bind PA-receptors), SSSR (resistant to activation by furin), Oligo(-) (unable to form oligomers), and DNI (Dominant Negative Inhibitory, unable to form endosomal transmembrane pores). When tested in mice and after three doses of immunization, all four mutants were highly potent in eliciting PA-specific, toxin-neutralizing antibodies, with immunogenicity increasing in the order of PA<Rec(-)<SSSR<Oligo(-)<DNI. While the differences between Rec(-) or SSSR and PA were small and not statistically significant, DNI and Oligo(-) were significantly more immunogenic than wild-type PA. One year after immunization and compared with PA-immunized mice, DNI-immunized mice maintained significantly higher levels of anti-PA IgG with correspondingly higher titers of toxin-neutralizing activity. In contrast, Oligo(-)-immunized mice had high levels of anti-PA IgG but lower titers of toxin-neutralizing activity, suggesting that Oligo(-) mutation sites may overlap with critical protective epitopes of PA. Our study demonstrates that PA-based vaccines could be improved both in terms of safety and efficacy by strategic mutations that not only render PA non-functional but also simultaneously enhance its immunogenic potency. Recombinant PA mutants, particularly DNI, hold great promise as better and safer antigens than wild-type PA for use in postexposure vaccination.
Collapse
Affiliation(s)
- Ming Yan
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | | | | | | |
Collapse
|
30
|
Kuo SR, Willingham MC, Bour SH, Andreas EA, Park SK, Jackson C, Duesbery NS, Leppla SH, Tang WJ, Frankel AE. Anthrax toxin-induced shock in rats is associated with pulmonary edema and hemorrhage. Microb Pathog 2007; 44:467-72. [PMID: 18222626 DOI: 10.1016/j.micpath.2007.12.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 12/01/2007] [Accepted: 12/07/2007] [Indexed: 12/23/2022]
Abstract
Bacillus anthracis infections are frequently associated with severe and often irreversible hypotensive shock despite appropriate antibiotics and aggressive hemodynamic and pulmonary support. Based on the observations that the anthrax secreted proteins-protective antigen (PA), lethal factor (LF), and edema factor (EF) also produce shock and mortality in animal models, we chose to characterize further the clinical chemistries and microscopic pathology of toxin treated rats. Groups of three male Sprague Dawley rats received bolus intravenous infusions of PA/LF, PA/EF, LF, or EF alone and blood samples and tissues were collected and assayed for chemistries and tissue pathology. In PA/LF and PA/EF treated animals but not other groups, chemistries showed transaminasemia and elevated lactate dehydrogenase. PA/LF treated animals alone showed elevated hemoglobin and hematocrits; PA/EF treated animals alone showed lymphopenia. Pathology was remarkable for pulmonary edema in PA/LF treated rat lungs and pulmonary hemorrhage in PA/EF treated rat lungs. These results are consistent with our and others' previous findings that the morbidity and mortality associated with anthrax are not cytokine-mediated but due to a direct effect of the toxins on the cardiovascular system along with toxin-specific alterations in blood counts. PA/LF pathology matches that seen with acute cardiac failure, and PA/EF pathology coincides with direct vascular endothelial injury. These observations provide a rational basis for drug interventions to reduce the effect of these toxins on the heart and blood vessels.
Collapse
Affiliation(s)
- Shu-Ru Kuo
- Cancer Research Institute, Scott & White Memorial Hospital,Temple, TX 76502, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Anthrax toxin consists of three nontoxic proteins that self-assemble at the surface of receptor-bearing mammalian cells or in solution, yielding a series of toxic complexes. Two of the proteins, called Lethal Factor (LF) and Edema Factor (EF), are enzymes that act on cytosolic substrates. The third, termed Protective Antigen (PA), is a multifunctional protein that binds to receptors, orchestrates the assembly and internalization of the complexes, and delivers them to the endosome. There, the PA moiety forms a pore in the endosomal membrane and promotes translocation of LF and EF to the cytosol. Recent advances in understanding the entry process include insights into how PA recognizes its two known receptors and its ligands, LF and EF; how the PA:receptor interaction influences the pH-dependence of pore formation; and how the pore functions in promoting translocation of LF and EF across the endosomal membrane.
Collapse
Affiliation(s)
- John A T Young
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA.
| | | |
Collapse
|
32
|
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
|
33
|
Watson LE, Kuo SR, Katki K, Dang T, Park SK, Dostal DE, Tang WJ, Leppla SH, Frankel AE. Anthrax toxins induce shock in rats by depressed cardiac ventricular function. PLoS One 2007; 2:e466. [PMID: 17520025 PMCID: PMC1867860 DOI: 10.1371/journal.pone.0000466] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Accepted: 05/01/2007] [Indexed: 12/05/2022] Open
Abstract
Anthrax infections are frequently associated with severe and often irreversible hypotensive shock. The isolated toxic proteins of Bacillus anthracis produce a non-cytokine-mediated hypotension in rats by unknown mechanisms. These observations suggest the anthrax toxins have direct cardiovascular effects. Here, we characterize these effects. As a first step, we administered systemically anthrax lethal toxin (LeTx) and edema toxin (EdTx) to cohorts of three to twelve rats at different doses and determined the time of onset, degree of hypotension and mortality. We measured serum concentrations of the protective antigen (PA) toxin component at various time points after infusion. Peak serum levels of PA were in the µg/mL range with half-lives of 10–20 minutes. With doses that produced hypotension with delayed lethality, we then gave bolus intravenous infusions of toxins to groups of four to six instrumented rats and continuously monitored blood pressure by telemetry. Finally, the same doses used in the telemetry experiments were given to additional groups of four rats, and echocardiography was performed pretreatment and one, two, three and twenty-four hours post-treatment. LeTx and EdTx each produced hypotension. We observed a doubling of the velocity of propagation and 20% increases in left ventricular diastolic and systolic areas in LeTx-treated rats, but not in EdTx-treated rats. EdTx-but not LeTx-treated rats showed a significant increase in heart rate. These results indicate that LeTx reduced left ventricular systolic function and EdTx reduced preload. Uptake of toxins occurs readily into tissues with biological effects occurring within minutes to hours of serum toxin concentrations in the µg/mL range. LeTx and EdTx yield an irreversible shock with subsequent death. These findings should provide a basis for the rational design of drug interventions to reduce the dismal prognosis of systemic anthrax infections.
Collapse
Affiliation(s)
- Linley E. Watson
- Division of Cardiology, Scott and White Memorial Hospital, Scott, Sherwood and Brindley Foundation, Temple, Texas, United States of America
- Department of Medicine, Texas A&M University System, Health Science Center College of Medicine, Temple, Texas, United States of America
- Division of Molecular Cardiology, Texas A&M University System, Health Science Center College of Medicine, and Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Shu-ru Kuo
- Scott and White Cancer Research Institute, Temple, Texas, United States of America
| | - Khurshed Katki
- Department of Medicine, Texas A&M University System, Health Science Center College of Medicine, Temple, Texas, United States of America
| | - Tongyun Dang
- Scott and White Cancer Research Institute, Temple, Texas, United States of America
| | - Seong Kyu Park
- Scott and White Cancer Research Institute, Temple, Texas, United States of America
| | - David E. Dostal
- Department of Medicine, Texas A&M University System, Health Science Center College of Medicine, Temple, Texas, United States of America
- Division of Molecular Cardiology, Texas A&M University System, Health Science Center College of Medicine, and Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Wei-Jen Tang
- Ben May Institute for Cancer Research, The University of Chicago, Chicago, Illinois, United States of America
| | - Stephen H. Leppla
- Bacterial Toxins and Therapeutics Section, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Arthur E. Frankel
- Department of Medicine, Texas A&M University System, Health Science Center College of Medicine, Temple, Texas, United States of America
- Scott and White Cancer Research Institute, Temple, Texas, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
34
|
Nye SH, Wittenburg AL, Evans DL, O'Connor JA, Roman RJ, Jacob HJ. Rat survival to anthrax lethal toxin is likely controlled by a single gene. THE PHARMACOGENOMICS JOURNAL 2007; 8:16-22. [PMID: 17440430 DOI: 10.1038/sj.tpj.6500448] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We examined whether survival of different rat strains administered anthrax lethal toxin is genetically determined. A reproducible test population of first filial generation hybrid rats was bred based on the susceptibility of progenitors to anthrax lethal toxin and to maximize genetic diversity across the strains. These rats were then tested with varying doses of anthrax lethal toxin. We found that all 'sensitive' strains died within 2 h following systemic administration of 240 mug/kg lethal toxin, while one strain survived following a five times higher dose (1.4 mg/kg). The ability of lethal toxin to lyse macrophage cultures derived from the bone marrow of these strains corresponded with in vivo results. We conclude that a rat test population can detect strain differences in response to anthrax lethal toxin. Survival is influenced by the host genome background and is likely due to a single gene with a recessive mode of inheritance.
Collapse
Affiliation(s)
- S H Nye
- PhysioGenix Inc., 10437 Innovation Drive, Milwaukee, WI 53226, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Chen KH, Liu S, Bankston LA, Liddington RC, Leppla SH. Selection of anthrax toxin protective antigen variants that discriminate between the cellular receptors TEM8 and CMG2 and achieve targeting of tumor cells. J Biol Chem 2007; 282:9834-9845. [PMID: 17251181 PMCID: PMC2530824 DOI: 10.1074/jbc.m611142200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anthrax toxin, a three-component protein toxin secreted by Bacillus anthracis, assembles into toxic complexes at the surface of receptor-bearing eukaryotic cells. The protective antigen (PA) protein binds to receptors, either tumor endothelial cell marker 8 (TEM8) or CMG2 (capillary morphogenesis protein 2), and orchestrates the delivery of the lethal and edema factors into the cytosol. TEM8 is reported to be overexpressed during tumor angiogenesis, whereas CMG2 is more widely expressed in normal tissues. To extend prior work on targeting of tumor with modified anthrax toxins, we used phage display to select PA variants that preferentially bind to TEM8 as compared with CMG2. Substitutions were randomly introduced into residues 605-729 of PA, within the C-terminal domain 4 of PA, which is the principal region that contacts receptor. Candidates were characterized in cellular cytotoxicity assays with Chinese hamster ovary (CHO) cells expressing either TEM8 or CMG2. A PA mutant having the substitutions R659S and M662R had enhanced specificity toward TEM8-overexpressing CHO cells. This PA variant also displayed broad and potent tumoricidal activity to various human tumor cells, especially to HeLa and A549/ATCC cells. By contrast, the substitution N657Q significantly reduced toxicity to TEM8 but not CMG2-overexpressing CHO cells. Our results indicate that certain amino acid substitutions within PA domain 4 create anthrax toxins that selectively kill human tumor cells. The PA R659S/M662R protein may be useful as a therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Kuang-Hua Chen
- Laboratory of Bacterial Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892-3202
| | - Shihui Liu
- Laboratory of Bacterial Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892-3202
| | - Laurie A Bankston
- Infectious and Inflammatory Disease Center, Burnham Institute for Medical Research, La Jolla, California 92037
| | - Robert C Liddington
- Infectious and Inflammatory Disease Center, Burnham Institute for Medical Research, La Jolla, California 92037
| | - Stephen H Leppla
- Laboratory of Bacterial Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892-3202.
| |
Collapse
|
36
|
Scobie HM, Marlett JM, Rainey GJA, Lacy DB, Collier RJ, Young JA. Anthrax toxin receptor 2 determinants that dictate the pH threshold of toxin pore formation. PLoS One 2007; 2:e329. [PMID: 17389920 PMCID: PMC1824706 DOI: 10.1371/journal.pone.0000329] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 02/28/2007] [Indexed: 11/24/2022] Open
Abstract
The anthrax toxin receptors, ANTXR1 and ANTXR2, act as molecular clamps to prevent the protective antigen (PA) toxin subunit from forming pores until exposure to low pH. PA forms pores at pH ∼6.0 or below when it is bound to ANTXR1, but only at pH ∼5.0 or below when it is bound to ANTXR2. Here, structure-based mutagenesis was used to identify non-conserved ANTXR2 residues responsible for this striking 1.0 pH unit difference in pH threshold. Residues conserved between ANTXR2 and ANTXR1 that influence the ANTXR2-associated pH threshold of pore formation were also identified. All of these residues contact either PA domain 2 or the neighboring edge of PA domain 4. These results provide genetic evidence for receptor release of these regions of PA as being necessary for the protein rearrangements that accompany anthrax toxin pore formation.
Collapse
Affiliation(s)
- Heather M. Scobie
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John M. Marlett
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - G. Jonah A. Rainey
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - D. Borden Lacy
- Department of Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - R. John Collier
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - John A.T. Young
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
37
|
Scobie HM, Young JA. Divalent metal ion coordination by residue T118 of anthrax toxin receptor 2 is not essential for protective antigen binding. PLoS One 2006; 1:e99. [PMID: 17183731 PMCID: PMC1762376 DOI: 10.1371/journal.pone.0000099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Accepted: 11/21/2006] [Indexed: 11/30/2022] Open
Abstract
The protective antigen (PA) subunit of anthrax toxin interacts with the integrin-like I domains of either of two cellular receptors, ANTXR1 or ANTXR2. These I domains contain a metal ion-dependent adhesion site (MIDAS) made up of five non-consecutive amino acid residues that coordinate a divalent metal ion that is important for PA-binding. The MIDAS residues of integrin I domains shift depending upon whether the domain exists in a closed (ligand-unbound) or open (ligand-bound) conformation. Of relevance to this study, the MIDAS threonine residue coordinates the metal ion only in the open I domain conformation. Previously it was shown that the MIDAS threonine is essential for PA interaction with ANTXR1, a result consistent with the requirement that the I domain of that receptor adopts an open conformation for PA-binding [1]. Here we have tested the requirement for the MIDAS threonine of ANTXR2 for PA-binding. We show that the toxin can bind to a mutant receptor lacking the MIDAS threonine and that it can use that mutant receptor to intoxicate cultured cells. These findings suggest that an open-like configuration of the ANTXR2 MIDAS is not essential for the interaction with PA.
Collapse
Affiliation(s)
- Heather M. Scobie
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - John A.T. Young
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|