51
|
Djukic M, Brzuszkiewicz E, Fünfhaus A, Voss J, Gollnow K, Poppinga L, Liesegang H, Garcia-Gonzalez E, Genersch E, Daniel R. How to kill the honey bee larva: genomic potential and virulence mechanisms of Paenibacillus larvae. PLoS One 2014; 9:e90914. [PMID: 24599066 PMCID: PMC3944939 DOI: 10.1371/journal.pone.0090914] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 02/05/2014] [Indexed: 12/20/2022] Open
Abstract
Paenibacillus larvae, a Gram positive bacterial pathogen, causes American Foulbrood (AFB), which is the most serious infectious disease of honey bees. In order to investigate the genomic potential of P. larvae, two strains belonging to two different genotypes were sequenced and used for comparative genome analysis. The complete genome sequence of P. larvae strain DSM 25430 (genotype ERIC II) consisted of 4,056,006 bp and harbored 3,928 predicted protein-encoding genes. The draft genome sequence of P. larvae strain DSM 25719 (genotype ERIC I) comprised 4,579,589 bp and contained 4,868 protein-encoding genes. Both strains harbored a 9.7 kb plasmid and encoded a large number of virulence-associated proteins such as toxins and collagenases. In addition, genes encoding large multimodular enzymes producing nonribosomally peptides or polyketides were identified. In the genome of strain DSM 25719 seven toxin associated loci were identified and analyzed. Five of them encoded putatively functional toxins. The genome of strain DSM 25430 harbored several toxin loci that showed similarity to corresponding loci in the genome of strain DSM 25719, but were non-functional due to point mutations or disruption by transposases. Although both strains cause AFB, significant differences between the genomes were observed including genome size, number and composition of transposases, insertion elements, predicted phage regions, and strain-specific island-like regions. Transposases, integrases and recombinases are important drivers for genome plasticity. A total of 390 and 273 mobile elements were found in strain DSM 25430 and strain DSM 25719, respectively. Comparative genomics of both strains revealed acquisition of virulence factors by horizontal gene transfer and provided insights into evolution and pathogenicity.
Collapse
Affiliation(s)
- Marvin Djukic
- Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Elzbieta Brzuszkiewicz
- Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Anne Fünfhaus
- Department for Molecular Microbiology and Bee Diseases, Institute for Bee Research, Hohen Neuendorf, Germany
| | - Jörn Voss
- Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Kathleen Gollnow
- Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Lena Poppinga
- Department for Molecular Microbiology and Bee Diseases, Institute for Bee Research, Hohen Neuendorf, Germany
| | - Heiko Liesegang
- Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Eva Garcia-Gonzalez
- Department for Molecular Microbiology and Bee Diseases, Institute for Bee Research, Hohen Neuendorf, Germany
| | - Elke Genersch
- Department for Molecular Microbiology and Bee Diseases, Institute for Bee Research, Hohen Neuendorf, Germany
| | - Rolf Daniel
- Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
52
|
Clostridium perfringens epsilon toxin: a malevolent molecule for animals and man? Toxins (Basel) 2013; 5:2138-60. [PMID: 24284826 PMCID: PMC3847718 DOI: 10.3390/toxins5112138] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/30/2013] [Accepted: 10/31/2013] [Indexed: 12/27/2022] Open
Abstract
Clostridium perfringens is a prolific, toxin-producing anaerobe causing multiple diseases in humans and animals. One of these toxins is epsilon, a 33 kDa protein produced by Clostridium perfringens (types B and D) that induces fatal enteric disease of goats, sheep and cattle. Epsilon toxin (Etx) belongs to the aerolysin-like toxin family. It contains three distinct domains, is proteolytically-activated and forms oligomeric pores on cell surfaces via a lipid raft-associated protein(s). Vaccination controls Etx-induced disease in the field. However, therapeutic measures are currently lacking. This review initially introduces C. perfringens toxins, subsequently focusing upon the Etx and its biochemistry, disease characteristics in various animals that include laboratory models (in vitro and in vivo), and finally control mechanisms (vaccines and therapeutics).
Collapse
|
53
|
Ferrarezi MC, Curci VCLM, Cardoso TC. Cellular vacuolation and mitochondrial-associated factors induced by Clostridium perfringens epsilon toxin detected using acoustic flow cytometry. Anaerobe 2013; 24:55-9. [PMID: 24076036 DOI: 10.1016/j.anaerobe.2013.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 07/31/2013] [Accepted: 09/17/2013] [Indexed: 11/18/2022]
Abstract
Epsilon toxin (ETX) produced by Clostridium perfringens types B and D is a potent toxin that is responsible for fatal enterotoxaemia. In vitro, ETX, which is considered as a pore-forming toxin, forms a heptamer in Madin-Darby canine kidney (MDCK) cell membranes, which is considered to be a pre-pore stage. After binding of the ETX, vacuoles inside cell cytoplasm are produced. ETX causes decreased levels of essential coenzymes required for host cell energy. Here, we optimized and applied acoustic flow cytometry analysis in order to gain further insight into ETX-pathogenesis. Using acoustic flow cytometer analysis, which considered highly sensitive, ETX-exposed MDCK cells revealed mitochondrial membrane decreases followed by 25.48% and 45.45% of the exposed cells expressing the Bax and BCL-2 proteins at a pre-pore stage, respectively. These results together with high cytotoxicity and visualization of cell vacuoles, demonstrates that acoustic flow cytometry analysis potentially represents an effective tool to study ETX pathogenesis.
Collapse
Affiliation(s)
- Marina C Ferrarezi
- University of São Paulo State, Departamento de Apoio, Produção e Saúde Animal, Rua Clóvis Pestana, 793, Araçatuba 16050-680, São Paulo, Brazil
| | | | | |
Collapse
|
54
|
Los FCO, Randis TM, Aroian RV, Ratner AJ. Role of pore-forming toxins in bacterial infectious diseases. Microbiol Mol Biol Rev 2013; 77:173-207. [PMID: 23699254 PMCID: PMC3668673 DOI: 10.1128/mmbr.00052-12] [Citation(s) in RCA: 308] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pore-forming toxins (PFTs) are the most common bacterial cytotoxic proteins and are required for virulence in a large number of important pathogens, including Streptococcus pneumoniae, group A and B streptococci, Staphylococcus aureus, Escherichia coli, and Mycobacterium tuberculosis. PFTs generally disrupt host cell membranes, but they can have additional effects independent of pore formation. Substantial effort has been devoted to understanding the molecular mechanisms underlying the functions of certain model PFTs. Likewise, specific host pathways mediating survival and immune responses in the face of toxin-mediated cellular damage have been delineated. However, less is known about the overall functions of PFTs during infection in vivo. This review focuses on common themes in the area of PFT biology, with an emphasis on studies addressing the roles of PFTs in in vivo and ex vivo models of colonization or infection. Common functions of PFTs include disruption of epithelial barrier function and evasion of host immune responses, which contribute to bacterial growth and spreading. The widespread nature of PFTs make this group of toxins an attractive target for the development of new virulence-targeted therapies that may have broad activity against human pathogens.
Collapse
Affiliation(s)
| | - Tara M. Randis
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Raffi V. Aroian
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| | - Adam J. Ratner
- Department of Pediatrics, Columbia University, New York, New York, USA
| |
Collapse
|
55
|
Wioland L, Dupont JL, Bossu JL, Popoff MR, Poulain B. Attack of the nervous system by Clostridium perfringens Epsilon toxin: from disease to mode of action on neural cells. Toxicon 2013; 75:122-35. [PMID: 23632158 DOI: 10.1016/j.toxicon.2013.04.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 03/29/2013] [Accepted: 04/10/2013] [Indexed: 12/24/2022]
Abstract
Epsilon toxin (ET), produced by Clostridium perfringens types B and D, ranks among the four most potent poisonous substances known so far. ET-intoxication is responsible for enterotoxaemia in animals, mainly sheep and goats. This disease comprises several manifestations indicating the attack of the nervous system. This review aims to summarize the effects of ET on central nervous system. ET binds to endothelial cells of brain capillary vessels before passing through the blood-brain barrier. Therefore, it induces perivascular oedema and accumulates into brain. ET binding to different brain structures and to different component in the brain indicates regional susceptibility to the toxin. Histological examination has revealed nerve tissue and cellular lesions, which may be directly or indirectly caused by ET. The naturally occurring disease caused by ET-intoxication can be reproduced experimentally in rodents. In mice and rats, ET recognizes receptor at the surface of different neural cell types, including certain neurons (e.g. the granule cells in cerebellum) as well as oligodendrocytes, which are the glial cells responsible for the axons myelination. Moreover, ET induces release of glutamate and other transmitters, leading to firing of neural network. The precise mode of action of ET on neural cells remains to be determined.
Collapse
Affiliation(s)
- Laetitia Wioland
- Centre National de la Recherche Scientifique (CNRS) and Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR 3212, Strasbourg, France
| | | | | | | | | |
Collapse
|
56
|
Yan Q, Wang J, Yin Y, Yuan J. Breathing Polymersomes: CO
2
‐Tuning Membrane Permeability for Size‐Selective Release, Separation, and Reaction. Angew Chem Int Ed Engl 2013; 52:5070-3. [DOI: 10.1002/anie.201300397] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Indexed: 12/26/2022]
Affiliation(s)
- Qiang Yan
- Key Lab of Organic Optoelectronics & Engineering Department of Chemistry, Tsinghua University, Beijing 100084 (P.R. China)
| | - Jianbo Wang
- Computer Science School, China Women's University, Beijing 100084 (P.R. China)
| | - Yingwu Yin
- Key Lab of Organic Optoelectronics & Engineering Department of Chemistry, Tsinghua University, Beijing 100084 (P.R. China)
| | - Jinying Yuan
- Key Lab of Organic Optoelectronics & Engineering Department of Chemistry, Tsinghua University, Beijing 100084 (P.R. China)
| |
Collapse
|
57
|
Yan Q, Wang J, Yin Y, Yuan J. Breathing Polymersomes: CO2-Tuning Membrane Permeability for Size-Selective Release, Separation, and Reaction. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201300397] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
58
|
Affiliation(s)
| | - Sergey M. Bezrukov
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, U.S.A
| |
Collapse
|
59
|
Oligomerization of Clostridium perfringens epsilon toxin is dependent upon caveolins 1 and 2. PLoS One 2012; 7:e46866. [PMID: 23056496 PMCID: PMC3462777 DOI: 10.1371/journal.pone.0046866] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 09/06/2012] [Indexed: 01/27/2023] Open
Abstract
Evidence from multiple studies suggests that Clostridium perfringens ε-toxin is a pore-forming toxin, assembling into oligomeric complexes in the plasma membrane of sensitive cells. In a previous study, we used gene-trap mutagenesis to identify mammalian factors contributing to toxin activity, including caveolin-2 (CAV2). In this study, we demonstrate the importance of caveolin-2 and its interaction partner, caveolin-1 (CAV1), in ε-toxin-induced cytotoxicity. Using CAV2-specific shRNA in a toxin-sensitive human kidney cell line, ACHN, we confirmed that cells deficient in CAV2 exhibit increased resistance to ε-toxin. Similarly, using CAV1-specific shRNA, we demonstrate that cells deficient in CAV1 also exhibit increased resistance to the toxin. Immunoprecipitation of CAV1 and CAV2 from ε-toxin-treated ACHN cells demonstrated interaction of both CAV1 and -2 with the toxin. Furthermore, blue-native PAGE indicated that the toxin and caveolins were components of a 670 kDa protein complex. Although ε-toxin binding was only slightly perturbed in caveolin-deficient cells, oligomerization of the toxin was dramatically reduced in both CAV1- and CAV2-deficient cells. These results indicate that CAV1 and -2 potentiate ε-toxin induced cytotoxicity by promoting toxin oligomerization – an event which is requisite for pore formation and, by extension, cell death.
Collapse
|
60
|
Ivie SE, McClain MS. Identification of amino acids important for binding of Clostridium perfringens epsilon toxin to host cells and to HAVCR1. Biochemistry 2012; 51:7588-95. [PMID: 22938730 PMCID: PMC3534817 DOI: 10.1021/bi300690a] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Clostridium perfringens epsilon toxin belongs to the aerolysin-like family of pore-forming toxins and is one of the most potent bacterial toxins known. The epsilon toxin causes fatal enterotoxemia in sheep, goats, and possibly humans. Evidence indicates that the toxin binds to protein receptors including hepatitis A virus cellular receptor 1 (HAVCR1), but the region of the toxin responsible for cell binding has not been identified. In the present study, we identify amino acids within the epsilon toxin important for this cell interaction. Site-specific mutagenesis was used to investigate the role of a surface-accessible cluster of aromatic amino acids, and purified mutant proteins were tested in a series of cell-culture assays to assess cytotoxic activity and cell binding. When added to cells, four mutant proteins (Etx-Y29E, Etx-Y30E, Etx-Y36E and Etx-Y196E) were severely impaired in their ability to not only kill host cells, but also in their ability to permeabilize the plasma membrane. Circular dichroism spectroscopy and thermal stability studies revealed that the wild-type and mutant proteins were similarly folded. Additional experiments revealed that these mutant proteins were defective in binding to host cells and to HAVCR1. These data indicate that an amino acid motif including Y29, Y30, Y36, and Y196 is important for the ability of epsilon toxin to interact with cells and HAVCR1.
Collapse
Affiliation(s)
- Susan E. Ivie
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Mark S. McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
61
|
Harkness JM, Li J, McClane BA. Identification of a lambda toxin-negative Clostridium perfringens strain that processes and activates epsilon prototoxin intracellularly. Anaerobe 2012; 18:546-52. [PMID: 22982043 DOI: 10.1016/j.anaerobe.2012.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 07/23/2012] [Accepted: 09/03/2012] [Indexed: 01/06/2023]
Abstract
Clostridium perfringens type B and D strains produce epsilon toxin (ETX), which is one of the most potent clostridial toxins and is involved in enteritis and enterotoxemias of domestic animals. ETX is produced initially as an inactive prototoxin that is typically then secreted and processed by intestinal proteases or possibly, for some strains, lambda toxin. During the current work a unique C. perfringens strain was identified that intracellularly processes epsilon prototoxin to an active form capable of killing MDCK cells. This activated toxin is not secreted but instead is apparently released upon lysis of bacterial cells entering stationary phase. These findings broaden understanding of the pathogenesis of type B and D infections by identifying a new mechanism of ETX activation.
Collapse
Affiliation(s)
- Justine M Harkness
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
62
|
Fennessey CM, Ivie SE, McClain MS. Coenzyme depletion by members of the aerolysin family of pore-forming toxins leads to diminished ATP levels and cell death. MOLECULAR BIOSYSTEMS 2012; 8:2097-105. [PMID: 22688384 PMCID: PMC3759351 DOI: 10.1039/c2mb25142f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent studies demonstrated that a variety of bacterial pore-forming toxins induce cell death through a process of programmed necrosis characterized by the rapid depletion of cellular ATP. However, events leading to the necrosis and depletion of ATP are not thoroughly understood. We demonstrate that ATP-depletion induced by two pore-forming toxins, the Clostridium perfringens epsilon-toxin and the Aeromonas hydrophila aerolysin toxin, is associated with decreased mitochondrial membrane potential and opening of the mitochondrial permeability transition pore. To gain further insight into the toxin-induced metabolic changes contributing to necrosis and depletion of ATP, we analyzed the biochemical profiles of 251 distinct compounds by GC/MS or LC/MS/MS following exposure of a human kidney cell line to the epsilon-toxin. As expected, numerous biochemicals were seen to increase or decrease in response to epsilon-toxin. However, the pattern of these changes was consistent with the toxin-induced disruption of major energy-producing pathways in the cell including disruptions to the beta-oxidation of lipids. In particular, treatment with epsilon-toxin led to decreased levels of key coenzymes required for energy production including carnitine, NAD (and NADH), and coenzyme A. Independent biochemical assays confirmed that epsilon-toxin and aerolysin induced the rapid decrease of these coenzymes or their synthetic precursors. Incubation of cells with NADH or carnitine-enriched medium helped protect cells from toxin-induced ATP depletion and cell death. Collectively, these results demonstrate that members of the aerolysin family of pore-forming toxins lead to decreased levels of essential coenzymes required for energy production. The resulting loss of energy substrates is expected to contribute to dissipation of the mitochondrial membrane potential, opening of the mitochondrial permeability transition pore, and ultimately cell death.
Collapse
Affiliation(s)
- Christine M. Fennessey
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Susan E. Ivie
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Mark S. McClain
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| |
Collapse
|
63
|
Alexeev D, Kostrjukova E, Aliper A, Popenko A, Bazaleev N, Tyakht A, Selezneva O, Akopian T, Prichodko E, Kondratov I, Chukin M, Demina I, Galyamina M, Kamashev D, Vanyushkina A, Ladygina V, Levitskii S, Lazarev V, Govorun V. Application of Spiroplasma melliferum Proteogenomic Profiling for the Discovery of Virulence Factors and Pathogenicity Mechanisms in Host-associated Spiroplasmas. J Proteome Res 2011; 11:224-36. [DOI: 10.1021/pr2008626] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Dmitry Alexeev
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Moscow Institute of Physics and Technology - Bioinformatics Dolgoprudny,
Pervomayskaya 21 , Moscow 117303, Russian Federation
| | - Elena Kostrjukova
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Alexander Aliper
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Anna Popenko
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Nikolay Bazaleev
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Alexander Tyakht
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Oksana Selezneva
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Russian Research Centre Kurchatov Institute, pl. Akademika Kurchatova
1, Moscow 123182, Russian Federation
| | - Tatyana Akopian
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Elena Prichodko
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Ilya Kondratov
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Mikhail Chukin
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Irina Demina
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Maria Galyamina
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Dmitri Kamashev
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Russian Research Centre Kurchatov Institute, pl. Akademika Kurchatova
1, Moscow 123182, Russian Federation
| | - Anna Vanyushkina
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Russian Research Centre Kurchatov Institute, pl. Akademika Kurchatova
1, Moscow 123182, Russian Federation
| | - Valentina Ladygina
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
| | - Sergei Levitskii
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Russian Research Centre Kurchatov Institute, pl. Akademika Kurchatova
1, Moscow 123182, Russian Federation
| | - Vasily Lazarev
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Russian Research Centre Kurchatov Institute, pl. Akademika Kurchatova
1, Moscow 123182, Russian Federation
| | - Vadim Govorun
- Russian Institute of Physico-Chemical Medicine, Malaya Pirogovskaya 1a,
Moscow, Russian Federation
- Russian Research Centre Kurchatov Institute, pl. Akademika Kurchatova
1, Moscow 123182, Russian Federation
- M.M. Shemyakin–Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Ul. Miklukho-Maklaya,
16/10 , Moscow 117997, Russian Federation
| |
Collapse
|
64
|
Popoff MR. Multifaceted interactions of bacterial toxins with the gastrointestinal mucosa. Future Microbiol 2011; 6:763-97. [PMID: 21797691 DOI: 10.2217/fmb.11.58] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The digestive tract is one of the ecosystems that harbors the largest number and greatest variety of bacteria. Among them, certain bacteria have developed various strategies, including the synthesis of virulence factors such as toxins, to interact with the intestinal mucosa, and are responsible for various pathologies. A large variety of bacterial toxins of different sizes, structures and modes of action are able to interact with the gastrointestinal mucosa. Some toxins, termed enterotoxins, directly stimulate fluid secretion in enterocytes or cause their death, whereas other toxins pass through the intestinal barrier and disseminate by the general circulation to remote organs or tissues, where they are active. After recognition of a membrane receptor on target cells, toxins can act at the cell membrane by transducing a signal across the membrane in a hormone-like manner, by pore formation or by damaging membrane compounds. Other toxins can enter the cells and modify an intracellular target leading to a disregulation of certain physiological processes or disorganization of some structural architectures and cell death. Toxins are fascinating molecules, which mimic or interfere with eukaryotic physiological processes. Thereby, they have permitted the identification and characterization of new natural hormones or regulatory pathways. Besides use as protective antigens in vaccines, toxins offer multiple possibilities in pharmacology, such as immune modulation or specific delivery of a protein of interest into target cells.
Collapse
Affiliation(s)
- M R Popoff
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, 25 rue du Dr Roux, 757245 Paris cedex 15, France.
| |
Collapse
|
65
|
Chan QWT, Cornman RS, Birol I, Liao NY, Chan SK, Docking TR, Jackman SD, Taylor GA, Jones SJM, de Graaf DC, Evans JD, Foster LJ. Updated genome assembly and annotation of Paenibacillus larvae, the agent of American foulbrood disease of honey bees. BMC Genomics 2011; 12:450. [PMID: 21923906 PMCID: PMC3188533 DOI: 10.1186/1471-2164-12-450] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 09/16/2011] [Indexed: 01/13/2023] Open
Abstract
Abstract Results We used the Illumina GAIIx platform and conventional Sanger sequencing to generate a 182-fold sequence coverage of the P. larvae genome, and assembled the data using ABySS into a total of 388 contigs spanning 4.5 Mbp. Comparative genomics analysis against fully-sequenced soil bacteria P. JDR2 and P. vortex showed that regions of poor conservation may contain putative virulence factors. We used GLIMMER to predict 3568 gene models, and named them based on homology revealed by BLAST searches; proteases, hemolytic factors, toxins, and antibiotic resistance enzymes were identified in this way. Finally, mass spectrometry was used to provide experimental evidence that at least 35% of the genes are expressed at the protein level. Conclusions This update on the genome of P. larvae and annotation represents an immense advancement from what we had previously known about this species. We provide here a reliable resource that can be used to elucidate the mechanism of infection, and by extension, more effective methods to control and cure this widespread honey bee disease.
Collapse
Affiliation(s)
- Queenie W T Chan
- Department of Biochemistry Ž Molecular Biology, Centre for High-throughput Biology, University of British Columbia, 2125 East Mall, Vancouver, British Columbia, V6T 1Z4 Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Nagahama M, Itohayashi Y, Hara H, Higashihara M, Fukatani Y, Takagishi T, Oda M, Kobayashi K, Nakagawa I, Sakurai J. Cellular vacuolation induced by Clostridium perfringens epsilon-toxin. FEBS J 2011; 278:3395-407. [PMID: 21781280 DOI: 10.1111/j.1742-4658.2011.08263.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The epsilon-toxin of Clostridium perfringens forms a heptamer in the membranes of Madin-Darby canine kidney cells, leading to cell death. Here, we report that it caused the vacuolation of Madin-Darby canine kidney cells. The toxin induced vacuolation in a dose-dependent and time-dependent manner. The monomer of the toxin formed oligomers on lipid rafts in membranes of the cells. Methyl-β-cyclodextrin and poly(ethylene glycol) 4000 inhibited the vacuolation. Epsilon-toxin was internalized into the cells. Confocal microscopy revealed that the internalized toxin was transported from early endosomes (early endosome antigen 1 staining) to late endosomes and lysosomes (lysosomal-associated membrane protein 2 staining) and then distributed to the membranes of vacuoles. Furthermore, the vacuolation was inhibited by bafilomycin A1, a V-type ATPase inhibitor, and colchicine and nocodazole, microtubule-depolymerizing agents. The early endosomal marker green fluorescent protein-Rab5 and early endosome antigen 1 did not localize to vacuolar membranes. In contrast, the vacuolar membranes were specifically stained by the late endosomal and lysosomal marker green fluorescent protein-Rab7 and lysosomal-associated membrane protein 2. The vacuoles in the toxin-treated cells were stained with LysoTracker Red DND-99, a marker for late endosomes and lysosomes. A dominant negative mutant of Rab7 prevented the vacuolization, whereas a mutant form of Rab5 was less effective. These results demonstrate, for the first time, that: (a) oligomers of epsilon-toxin formed in lipid rafts are endocytosed; and (b) the vacuoles originating from late endosomes and lysosomes are formed by an oligomer of epsilon-toxin.
Collapse
Affiliation(s)
- Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Robertson SL, Li J, Uzal FA, McClane BA. Evidence for a prepore stage in the action of Clostridium perfringens epsilon toxin. PLoS One 2011; 6:e22053. [PMID: 21814565 PMCID: PMC3140917 DOI: 10.1371/journal.pone.0022053] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 06/16/2011] [Indexed: 11/25/2022] Open
Abstract
Clostridium perfringens epsilon toxin (ETX) rapidly kills MDCK II cells at 37°C, but not 4°C. The current study shows that, in MDCK II cells, ETX binds and forms an oligomeric complex equally well at 37°C and 4°C but only forms a pore at 37°C. However, the complex formed in MDCK cells treated with ETX at 4°C has the potential to form an active pore, since shifting those cells to 37°C results in rapid cytotoxicity. Those results suggested that the block in pore formation at 4°C involves temperature-related trapping of ETX in a prepore intermediate on the MDCK II cell plasma membrane surface. Evidence supporting this hypothesis was obtained when the ETX complex in MDCK II cells was shown to be more susceptible to pronase degradation when formed at 4°C vs. 37°C; this result is consistent with ETX complex formed at 4°C remaining present in an exposed prepore on the membrane surface, while the ETX prepore complex formed at 37°C is unaccessible to pronase because it has inserted into the plasma membrane to form an active pore. In addition, the ETX complex rapidly dissociated from MDCK II cells at 4°C, but not 37°C; this result is consistent with the ETX complex being resistant to dissociation at 37°C because it has inserted into membranes, while the ETX prepore readily dissociates from cells at 4°C because it remains on the membrane surface. These results support the identification of a prepore stage in ETX action and suggest a revised model for ETX cytotoxicity, i) ETX binds to an unidentified receptor, ii) ETX oligomerizes into a prepore on the membrane surface, and iii) the prepore inserts into membranes, in a temperature-sensitive manner, to form an active pore.
Collapse
Affiliation(s)
- Susan L. Robertson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Francisco A. Uzal
- California Animal Health and Food Safety Laboratory System, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Bruce A. McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
68
|
|
69
|
Bokori-Brown M, Savva CG, Fernandes da Costa SP, Naylor CE, Basak AK, Titball RW. Molecular basis of toxicity of Clostridium perfringens epsilon toxin. FEBS J 2011; 278:4589-601. [PMID: 21518257 DOI: 10.1111/j.1742-4658.2011.08140.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Clostridium perfringens ε-toxin is produced by toxinotypes B and D strains. The toxin is the aetiological agent of dysentery in newborn lambs but is also associated with enteritis and enterotoxaemia in goats, calves and foals. It is considered to be a potential biowarfare or bioterrorism agent by the US Government Centers for Disease Control and Prevention. The relatively inactive 32.9 kDa prototoxin is converted to active mature toxin by proteolytic cleavage, either by digestive proteases of the host, such as trypsin and chymotrypsin, or by C. perfringens λ-protease. In vivo, the toxin appears to target the brain and kidneys, but relatively few cell lines are susceptible to the toxin, and most work has been carried out using Madin-Darby canine kidney (MDCK) cells. The binding of ε-toxin to MDCK cells and rat synaptosomal membranes is associated with the formation of a stable, high molecular weight complex. The crystal structure of ε-toxin reveals similarity to aerolysin from Aeromonas hydrophila, parasporin-2 from Bacillus thuringiensis and a lectin from Laetiporus sulphureus. Like these toxins, ε-toxin appears to form heptameric pores in target cell membranes. The exquisite specificity of the toxin for specific cell types suggests that it binds to a receptor found only on these cells.
Collapse
Affiliation(s)
- Monika Bokori-Brown
- Biosciences, College of Life and Environmental Sciences, University of Exeter, UK
| | | | | | | | | | | |
Collapse
|
70
|
Ivie SE, Fennessey CM, Sheng J, Rubin DH, McClain MS. Gene-trap mutagenesis identifies mammalian genes contributing to intoxication by Clostridium perfringens ε-toxin. PLoS One 2011; 6:e17787. [PMID: 21412435 PMCID: PMC3055893 DOI: 10.1371/journal.pone.0017787] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 02/14/2011] [Indexed: 12/13/2022] Open
Abstract
The Clostridium perfringens ε-toxin is an extremely potent toxin associated with lethal toxemias in domesticated ruminants and may be toxic to humans. Intoxication results in fluid accumulation in various tissues, most notably in the brain and kidneys. Previous studies suggest that the toxin is a pore-forming toxin, leading to dysregulated ion homeostasis and ultimately cell death. However, mammalian host factors that likely contribute to ε-toxin-induced cytotoxicity are poorly understood. A library of insertional mutant Madin Darby canine kidney (MDCK) cells, which are highly susceptible to the lethal affects of ε-toxin, was used to select clones of cells resistant to ε-toxin-induced cytotoxicity. The genes mutated in 9 surviving resistant cell clones were identified. We focused additional experiments on one of the identified genes as a means of validating the experimental approach. Gene expression microarray analysis revealed that one of the identified genes, hepatitis A virus cellular receptor 1 (HAVCR1, KIM-1, TIM1), is more abundantly expressed in human kidney cell lines than it is expressed in human cells known to be resistant to ε-toxin. One human kidney cell line, ACHN, was found to be sensitive to the toxin and expresses a larger isoform of the HAVCR1 protein than the HAVCR1 protein expressed by other, toxin-resistant human kidney cell lines. RNA interference studies in MDCK and in ACHN cells confirmed that HAVCR1 contributes to ε-toxin-induced cytotoxicity. Additionally, ε-toxin was shown to bind to HAVCR1 in vitro. The results of this study indicate that HAVCR1 and the other genes identified through the use of gene-trap mutagenesis and RNA interference strategies represent important targets for investigation of the process by which ε-toxin induces cell death and new targets for potential therapeutic intervention.
Collapse
Affiliation(s)
- Susan E. Ivie
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Christine M. Fennessey
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Jinsong Sheng
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Donald H. Rubin
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Research Medicine, VA Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
| | - Mark S. McClain
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
71
|
Polymer partitioning and ion selectivity suggest asymmetrical shape for the membrane pore formed by epsilon toxin. Biophys J 2010; 99:782-9. [PMID: 20682255 DOI: 10.1016/j.bpj.2010.05.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 04/27/2010] [Accepted: 05/03/2010] [Indexed: 12/25/2022] Open
Abstract
Using poly-(ethylene glycol)s of different molecular weights, we probe the channels formed in planar lipid bilayers by epsilon toxin secreted by the anaerobic bacterium Clostridium perfringens. We find that the pore is highly asymmetric. The cutoff size of polymers entering the pore through its opening from the cis side, the side of toxin addition, is approximately 500 Da, whereas the cutoff size for the polymers entering from the trans side is approximately 2300 Da. Comparing these characteristic molecular weights with those reported earlier for OmpF porin and the alpha-Hemolysin channel, we estimate the radii of cis and trans openings as 0.4 nm and 1.0 nm, respectively. The simplest geometry corresponding to these findings is that of a truncated cone. The asymmetry of the pore is also confirmed by measurements of the reversal potential at oppositely directed salt gradients. The moderate anionic selectivity of the channel is salted-out more efficiently when the salt concentration is higher at the trans side of the pore.
Collapse
|
72
|
Shimada H, Kitada S. Mega assemblages of oligomeric aerolysin-like toxins stabilized by toxin-associating membrane proteins. J Biochem 2010; 149:103-15. [PMID: 20971989 DOI: 10.1093/jb/mvq124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Most β pore-forming toxins need to be oligomerized via receptors in order to form membrane pores. Though oligomerizing toxins frequently form SDS-resistant oligomers, it was questionable whether SDS-resistant oligomers reflected native functional toxin complexes. In order to elucidate the essence of the cytocidal assemblages, oligomers of aerolysin-like toxins, aerolysin, parasporin-2 and epsilon toxin, were examined with or without SDS. On Blue Native PAGE, each toxin, which had been solubilized from target cells with mild detergent, was a much larger complex (nearly 1 MDa) than the typical SDS-resistant oligomers (∼200 kDa). Size exclusion chromatography confirmed the huge toxin complexes. While a portion of the huge complexes were sensitive to proteases, SDS-resistant oligomers resist the proteolysis. Presumably the core toxin complexes remained intact while the cellular proteins were degraded. Moreover, intermediate complexes, which included no SDS-resistant oligomers, could be detected at lower temperatures. This study provides evidence for huge functional complexes of β pore-forming toxins and emphasizes their potential variance in composition.
Collapse
Affiliation(s)
- Hiroyasu Shimada
- Department of Bioscience and Bioinfomatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | | |
Collapse
|
73
|
Lonchamp E, Dupont JL, Wioland L, Courjaret R, Mbebi-Liegeois C, Jover E, Doussau F, Popoff MR, Bossu JL, de Barry J, Poulain B. Clostridium perfringens epsilon toxin targets granule cells in the mouse cerebellum and stimulates glutamate release. PLoS One 2010; 5:e13046. [PMID: 20941361 PMCID: PMC2948003 DOI: 10.1371/journal.pone.0013046] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 09/03/2010] [Indexed: 11/19/2022] Open
Abstract
Epsilon toxin (ET) produced by C. perfringens types B and D is a highly potent pore-forming toxin. ET-intoxicated animals express severe neurological disorders that are thought to result from the formation of vasogenic brain edemas and indirect neuronal excitotoxicity. The cerebellum is a predilection site for ET damage. ET has been proposed to bind to glial cells such as astrocytes and oligodendrocytes. However, the possibility that ET binds and attacks the neurons remains an open question. Using specific anti-ET mouse polyclonal antibodies and mouse brain slices preincubated with ET, we found that several brain structures were labeled, the cerebellum being a prominent one. In cerebellar slices, we analyzed the co-staining of ET with specific cell markers, and found that ET binds to the cell body of granule cells, oligodendrocytes, but not astrocytes or nerve endings. Identification of granule cells as neuronal ET targets was confirmed by the observation that ET induced intracellular Ca(2+) rises and glutamate release in primary cultures of granule cells. In cultured cerebellar slices, whole cell patch-clamp recordings of synaptic currents in Purkinje cells revealed that ET greatly stimulates both spontaneous excitatory and inhibitory activities. However, pharmacological dissection of these effects indicated that they were only a result of an increased granule cell firing activity and did not involve a direct action of the toxin on glutamatergic nerve terminals or inhibitory interneurons. Patch-clamp recordings of granule cell somata showed that ET causes a decrease in neuronal membrane resistance associated with pore-opening and depolarization of the neuronal membrane, which subsequently lead to the firing of the neuronal network and stimulation of glutamate release. This work demonstrates that a subset of neurons can be directly targeted by ET, suggesting that part of ET-induced neuronal damage observed in neuronal tissue is due to a direct effect of ET on neurons.
Collapse
Affiliation(s)
- Etienne Lonchamp
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Jean-Luc Dupont
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Laetitia Wioland
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Raphaël Courjaret
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Corinne Mbebi-Liegeois
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Emmanuel Jover
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Frédéric Doussau
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Michel R. Popoff
- Unité des Anaérobies et Leurs Toxines, Institut Pasteur à Paris, Paris, France
| | - Jean-Louis Bossu
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Jean de Barry
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Bernard Poulain
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| |
Collapse
|
74
|
Mathur DD, Deshmukh S, Kaushik H, Garg LC. Functional and structural characterization of soluble recombinant epsilon toxin of Clostridium perfringens D, causative agent of enterotoxaemia. Appl Microbiol Biotechnol 2010; 88:877-84. [PMID: 20676627 DOI: 10.1007/s00253-010-2785-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 06/20/2010] [Accepted: 07/18/2010] [Indexed: 10/19/2022]
Abstract
Clostridium perfringens types B and D are responsible for enterotoxaemia, one of the major causes of cattle mortality and is therefore of great economic concern. The epsilon toxin produced by the organism is the major antigenic determinant and has been directly implicated for the disease causation. In the present paper, we evaluated the biological activity of the recombinant epsilon toxin (rEtx) produced as soluble protein in Escherichia coli. The rEtx was purified to near homogeneity by a one-step anion-exchange chromatography. The immunological identity of purified rEtx was confirmed by Western blotting using a monoclonal antibody against the native toxin. The rEtx formed heptamer in the Madin-Darby canine kidney (MDCK) cells and synaptosomal membrane of mouse brain and was cytotoxic to the MDCK cells with a CT(50) of 30 ng/ml. The rEtx was highly stable and its thermostability profile related well with its biological activity. The rEtx was purified in large amounts and exhibited all the properties of native toxin and therefore can be used for the development of vaccine against the pathogen.
Collapse
Affiliation(s)
- Deepika Dayal Mathur
- Gene Regulation Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | | | | |
Collapse
|
75
|
Lewis M, Weaver CD, McClain MS. Identification of Small Molecule Inhibitors of Clostridium perfringens ε-Toxin Cytotoxicity Using a Cell-Based High-Throughput Screen. Toxins (Basel) 2010; 2:1825-1847. [PMID: 20721308 PMCID: PMC2922765 DOI: 10.3390/toxins2071825] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 06/23/2010] [Accepted: 07/06/2010] [Indexed: 11/16/2022] Open
Abstract
The Clostridium perfringens epsilon toxin, a select agent, is responsible for a severe, often fatal enterotoxemia characterized by edema in the heart, lungs, kidney, and brain. The toxin is believed to be an oligomeric pore-forming toxin. Currently, there is no effective therapy for countering the cytotoxic activity of the toxin in exposed individuals. Using a robust cell-based high-throughput screening (HTS) assay, we screened a 151,616-compound library for the ability to inhibit ε-toxin-induced cytotoxicity. Survival of MDCK cells exposed to the toxin was assessed by addition of resazurin to detect metabolic activity in surviving cells. The hit rate for this screen was 0.6%. Following a secondary screen of each hit in triplicate and assays to eliminate false positives, we focused on three structurally-distinct compounds: an N-cycloalkylbenzamide, a furo[2,3-b]quinoline, and a 6H-anthra[1,9-cd]isoxazol. None of the three compounds appeared to inhibit toxin binding to cells or the ability of the toxin to form oligomeric complexes. Additional assays demonstrated that two of the inhibitory compounds inhibited ε-toxin-induced permeabilization of MDCK cells to propidium iodide. Furthermore, the two compounds exhibited inhibitory effects on cells pre-treated with toxin. Structural analogs of one of the inhibitors identified through the high-throughput screen were analyzed and provided initial structure-activity data. These compounds should serve as the basis for further structure-activity refinement that may lead to the development of effective anti-ε-toxin therapeutics.
Collapse
Affiliation(s)
- Michelle Lewis
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Charles David Weaver
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Mark S. McClain
- Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
76
|
On the interaction of Clostridium perfringens enterotoxin with claudins. Toxins (Basel) 2010; 2:1336-56. [PMID: 22069641 PMCID: PMC3153257 DOI: 10.3390/toxins2061336] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 05/21/2010] [Accepted: 06/04/2010] [Indexed: 02/01/2023] Open
Abstract
Clostridium perfringens causes one of the most common foodborne illnesses, which is largely mediated by the Clostridium perfringens enterotoxin (CPE). The toxin consists of two functional domains. The N-terminal region mediates the cytotoxic effect through pore formation in the plasma membrane of the mammalian host cell. The C-terminal region (cCPE) binds to the second extracellular loop of a subset of claudins. Claudin-3 and claudin-4 have been shown to be receptors for CPE with very high affinity. The toxin binds with weak affinity to claudin-1 and -2 but contribution of these weak binding claudins to CPE-mediated disease is questionable. cCPE is not cytotoxic, however, it is a potent modulator of tight junctions. This review describes recent progress in the molecular characterization of the cCPE-claudin interaction using mutagenesis, in vitro binding assays and permeation studies. The results promote the development of recombinant cCPE-proteins and CPE-based peptidomimetics to modulate tight junctions for improved drug delivery or to treat tumors overexpressing claudins.
Collapse
|
77
|
Popoff MR, Poulain B. Bacterial toxins and the nervous system: neurotoxins and multipotential toxins interacting with neuronal cells. Toxins (Basel) 2010; 2:683-737. [PMID: 22069606 PMCID: PMC3153206 DOI: 10.3390/toxins2040683] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 03/18/2010] [Accepted: 04/07/2010] [Indexed: 12/13/2022] Open
Abstract
Toxins are potent molecules used by various bacteria to interact with a host organism. Some of them specifically act on neuronal cells (clostridial neurotoxins) leading to characteristics neurological affections. But many other toxins are multifunctional and recognize a wider range of cell types including neuronal cells. Various enterotoxins interact with the enteric nervous system, for example by stimulating afferent neurons or inducing neurotransmitter release from enterochromaffin cells which result either in vomiting, in amplification of the diarrhea, or in intestinal inflammation process. Other toxins can pass the blood brain barrier and directly act on specific neurons.
Collapse
Affiliation(s)
- Michel R. Popoff
- Neurotransmission et Sécrétion Neuroendocrine, CNRS UPR 2356 IFR 37 - Neurosciences, Centre de Neurochimie, 5, rue Blaise Pascal, F-67084 STRASBOURG cedex, France;
- Author to whom correspondence should be addressed;
| | | |
Collapse
|
78
|
Abstract
Clostridia produce the highest number of toxins of any type of bacteria and are involved in severe diseases in humans and other animals. Most of the clostridial toxins are pore-forming toxins responsible for gangrenes and gastrointestinal diseases. Among them, perfringolysin has been extensively studied and it is the paradigm of the cholesterol-dependent cytolysins, whereas Clostridium perfringens epsilon-toxin and Clostridium septicum alpha-toxin, which are related to aerolysin, are the prototypes of clostridial toxins that form small pores. Other toxins active on the cell surface possess an enzymatic activity, such as phospholipase C and collagenase, and are involved in the degradation of specific cell-membrane or extracellular-matrix components. Three groups of clostridial toxins have the ability to enter cells: large clostridial glucosylating toxins, binary toxins and neurotoxins. The binary and large clostridial glucosylating toxins alter the actin cytoskeleton by enzymatically modifying the actin monomers and the regulatory proteins from the Rho family, respectively. Clostridial neurotoxins proteolyse key components of neuroexocytosis. Botulinum neurotoxins inhibit neurotransmission at neuromuscular junctions, whereas tetanus toxin targets the inhibitory interneurons of the CNS. The high potency of clostridial toxins results from their specific targets, which have an essential cellular function, and from the type of modification that they induce. In addition, clostridial toxins are useful pharmacological and biological tools.
Collapse
Affiliation(s)
- Michel R Popoff
- Institut Pasteur, Bactéries Anaérobies et Toxines, 75724 Paris cedex 15, France.
| | | |
Collapse
|
79
|
Pelish TM, McClain MS. Dominant-negative inhibitors of the Clostridium perfringens epsilon-toxin. J Biol Chem 2009; 284:29446-53. [PMID: 19720828 PMCID: PMC2785577 DOI: 10.1074/jbc.m109.021782] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/24/2009] [Indexed: 01/27/2023] Open
Abstract
The Clostridium perfringens epsilon-toxin is responsible for a severe, often lethal intoxication. In this study, we characterized dominant-negative inhibitors of the epsilon-toxin. Site-specific mutations were introduced into the gene encoding epsilon-toxin, and recombinant proteins were expressed in Escherichia coli. Paired cysteine substitutions were introduced at locations predicted to form a disulfide bond. One cysteine in each mutant was introduced into the membrane insertion domain of the toxin; the second cysteine was introduced into the protein backbone. Mutant proteins with cysteine substitutions at amino acid positions I51/A114 and at V56/F118 lacked detectable cytotoxic activity in a MDCK cell assay. Cytotoxic activity could be reconstituted in both mutant proteins by incubation with dithiothreitol, indicating that the lack of cytotoxic activity was attributable to the formation of a disulfide bond. Fluorescent labeling of the cysteines also indicated that the introduced cysteines participated in a disulfide bond. When equimolar mixtures of wild-type epsilon-toxin and mutant proteins were added to MDCK cells, the I51C/A114C and V56C/F118C mutant proteins each inhibited the activity of wild-type epsilon-toxin. Further analysis of the inhibitory activity of the I51C/A114C and V56C/F118C mutant proteins indicated that these proteins inhibit the ability of the active toxin to form stable oligomeric complexes in the context of MDCK cells. These results provide further insight into the properties of dominant-negative inhibitors of oligomeric pore-forming toxins and provide the basis for developing new therapeutics for treating intoxication by epsilon-toxin.
Collapse
Affiliation(s)
- Teal M. Pelish
- From the Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Mark S. McClain
- From the Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
80
|
Knapp O, Maier E, Benz R, Geny B, Popoff MR. Identification of the channel-forming domain of Clostridium perfringens Epsilon-toxin (ETX). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2584-93. [PMID: 19835840 DOI: 10.1016/j.bbamem.2009.09.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 09/17/2009] [Accepted: 09/29/2009] [Indexed: 10/20/2022]
Abstract
Epsilon-toxin (ETX) is a potent toxin produced by Clostridium perfringens strains B and D. The bacteria are important pathogens in domestic animals and cause edema mediated by ETX. This toxin acts most likely by heptamer formation and rapid permeabilization of target cell membranes for monovalent anions and cations followed by a later entry of calcium. In this study, we compared the primary structure of ETX with that of the channel-forming stretches of a variety of binding components of A-B-types of toxins such as Anthrax protective antigen (PA), C2II of C2-toxin and Ib of Iota-toxin and found a remarkable homology to amino acids 151-180 of ETX. Site-directed mutagenesis of amino acids within the putative channel-forming domain resulted in changes of cytotoxicity and effects on channel characteristics in lipid bilayer experiments including changes of selectivity and partial channel block by methanethiosulfonate (MTS) reagents and antibodies against His(6)-tags from the trans-side of the lipid bilayer membranes.
Collapse
Affiliation(s)
- Oliver Knapp
- Department of Biotechnology, Biocenter, University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | | | | | | | | |
Collapse
|
81
|
Knapp O, Maier E, Mkaddem SB, Benz R, Bens M, Chenal A, Geny B, Vandewalle A, Popoff MR. Clostridium septicum alpha-toxin forms pores and induces rapid cell necrosis. Toxicon 2009; 55:61-72. [PMID: 19632260 DOI: 10.1016/j.toxicon.2009.06.037] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 06/25/2009] [Accepted: 06/29/2009] [Indexed: 11/25/2022]
Abstract
Alpha-toxin is the unique lethal virulent factor produced by Clostridium septicum, which causes traumatic or non-traumatic gas gangrene and necrotizing enterocolitis in humans. Here, we analyzed channel formation of the recombinant septicum alpha-toxin and characterized its activity on living cells. Recombinant septicum alpha-toxin induces the formation of ion-permeable channels with a single-channel conductance of about 175pS in 0.1M KCl in lipid bilayer membranes, which is typical for a large diffusion pore. Septicum alpha-toxin channels remained mostly in the open configuration, displayed no lipid specificity, and exhibited slight anion selectivity. Septicum alpha-toxin caused a rapid decrease in the transepithelial electrical resistance of MDCK cell monolayers grown on filters, and induced a rapid cell necrosis in a variety of cell lines, characterized by cell permeabilization to propidium iodide without DNA fragmentation and activation of caspase-3. Septicum alpha-toxin also induced a rapid K(+) efflux and ATP depletion. Incubation of the cells in K(+)-enriched medium delayed cell death caused by septicum alpha-toxin or epsilon-toxin, another potent pore-forming toxin, suggesting that the rapid loss of intracellular K(+) represents an early signal of pore-forming toxins-mediated cell necrosis.
Collapse
Affiliation(s)
- Oliver Knapp
- Institut Pasteur, Bactéries anaérobies et Toxines, 28 rue du Dr Roux, F-75724 Paris cedex 15, France
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Masson JB, Casanova D, Türkcan S, Voisinne G, Popoff MR, Vergassola M, Alexandrou A. Inferring maps of forces inside cell membrane microdomains. PHYSICAL REVIEW LETTERS 2009; 102:048103. [PMID: 19257479 DOI: 10.1103/physrevlett.102.048103] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Indexed: 05/27/2023]
Abstract
Mapping of the forces on biomolecules in cell membranes has spurred the development of effective labels, e.g., organic fluorophores and nanoparticles, to track trajectories of single biomolecules. Standard methods use particular statistics, namely the mean square displacement, to analyze the underlying dynamics. Here, we introduce general inference methods to fully exploit information in the experimental trajectories, providing sharp estimates of the forces and the diffusion coefficients in membrane microdomains. Rapid and reliable convergence of the inference scheme is demonstrated on trajectories generated numerically. The method is then applied to infer forces and potentials acting on the receptor of the toxin labeled by lanthanide-ion nanoparticles. Our scheme is applicable to any labeled biomolecule and results show its general relevance for membrane compartmentation.
Collapse
Affiliation(s)
- J-B Masson
- Institut Pasteur, CNRS URA 2171, Unit In Silico Genetics, 75724 Paris Cedex 15, France.
| | | | | | | | | | | | | |
Collapse
|
83
|
Manich M, Knapp O, Gibert M, Maier E, Jolivet-Reynaud C, Geny B, Benz R, Popoff MR. Clostridium perfringens delta toxin is sequence related to beta toxin, NetB, and Staphylococcus pore-forming toxins, but shows functional differences. PLoS One 2008; 3:e3764. [PMID: 19018299 PMCID: PMC2583947 DOI: 10.1371/journal.pone.0003764] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 10/19/2008] [Indexed: 11/18/2022] Open
Abstract
Clostridium perfringens produces numerous toxins, which are responsible for severe diseases in man and animals. Delta toxin is one of the three hemolysins released by a number of C. perfringens type C and possibly type B strains. Delta toxin was characterized to be cytotoxic for cells expressing the ganglioside GM2 in their membrane. Here we report the genetic characterization of Delta toxin and its pore forming activity in lipid bilayers. Delta toxin consists of 318 amino acids, its 28 N-terminal amino acids corresponding to a signal peptide. The secreted Delta toxin (290 amino acids; 32619 Da) is a basic protein (pI 9.1) which shows a significant homology with C. perfringens Beta toxin (43% identity), with C. perfringens NetB (40% identity) and, to a lesser extent, with Staphylococcus aureus alpha toxin and leukotoxins. Recombinant Delta toxin showed a preference for binding to GM2, in contrast to Beta toxin, which did not bind to gangliosides. It is hemolytic for sheep red blood cells and cytotoxic for HeLa cells. In artificial diphytanoyl phosphatidylcholine membranes, Delta and Beta toxin formed channels. Conductance of the channels formed by Delta toxin, with a value of about 100 pS to more than 1 nS in 1 M KCl and a membrane potential of 20 mV, was higher than those formed by Beta toxin and their distribution was broader. The results of zero-current membrane potential measurements and single channel experiments suggest that Delta toxin forms slightly anion-selective channels, whereas the Beta toxin channels showed a preference for cations under the same conditions. C. perfringens Delta toxin shows a significant sequence homolgy with C. perfringens Beta and NetB toxins, as well as with S. aureus alpha hemolysin and leukotoxins, but exhibits different channel properties in lipid bilayers. In contrast to Beta toxin, Delta toxin recognizes GM2 as receptor and forms anion-selective channels.
Collapse
Affiliation(s)
- Maria Manich
- Bactéries anaérobies et Toxines, Institut Pasteur, Paris, France
| | - Oliver Knapp
- Bactéries anaérobies et Toxines, Institut Pasteur, Paris, France
- Lehrstuhl für Biotechnologie, Theodor-Boveri-Institut (Biozentrum) der Universität Würzburg, Würzburg, Germany
| | - Maryse Gibert
- Bactéries anaérobies et Toxines, Institut Pasteur, Paris, France
| | - Elke Maier
- Lehrstuhl für Biotechnologie, Theodor-Boveri-Institut (Biozentrum) der Universität Würzburg, Würzburg, Germany
| | | | - Blandine Geny
- Bactéries anaérobies et Toxines, Institut Pasteur, Paris, France
| | - Roland Benz
- Lehrstuhl für Biotechnologie, Theodor-Boveri-Institut (Biozentrum) der Universität Würzburg, Würzburg, Germany
| | - Michel R. Popoff
- Bactéries anaérobies et Toxines, Institut Pasteur, Paris, France
- * E-mail:
| |
Collapse
|
84
|
Losada-Eaton DM, Uzal FA, Fernández Miyakawa ME. Clostridium perfringens epsilon toxin is absorbed from different intestinal segments of mice. Toxicon 2008; 51:1207-13. [PMID: 18457853 DOI: 10.1016/j.toxicon.2008.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2007] [Revised: 02/07/2008] [Accepted: 02/08/2008] [Indexed: 11/30/2022]
Abstract
Clostridium perfringens epsilon toxin is a potent toxin responsible for a rapidly fatal enterotoxaemia in several animal species. The pathogenesis of epsilon toxin includes toxicity to endothelial cells and neurons. Although epsilon toxin is absorbed from the gastrointestinal tract, the intestinal regions where the toxin is absorbed and the conditions favoring epsilon toxin absorption are unknown. The aim of this paper was to determine the toxicity of epsilon toxin absorbed from different gastrointestinal segments of mice and to evaluate the influence of the intestinal environment in the absorption of this toxin. Epsilon toxin diluted in one of several different saline solutions was surgically introduced into ligated stomach or intestinal segments of mice. Comparison of the toxicity of epsilon toxin injected in different sections of the gastrointestinal tract showed that this toxin can be absorbed from the small and the large intestine but not from the stomach of mice. The lethality of epsilon toxin was higher when this toxin was injected in the colon than in the small intestine. Low pH, and Na(+) and glucose added to the saline solution increased the toxicity of epsilon toxin injected into the small intestine. This study shows that absorption of epsilon toxin can occur in any intestinal segment of mice and that the physicochemical characteristics of the intestinal content can affect the absorption of this toxin.
Collapse
Affiliation(s)
- D M Losada-Eaton
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Universidad de Buenos Aires, Paraguay 2155, C1121ABG Buenos Aires, Argentina
| | | | | |
Collapse
|
85
|
Fernandez Miyakawa ME, Ibarra CA, Uzal FA. In vitro effects of Clostridium perfringens type D epsilon toxin on water and ion transport in ovine and caprine intestine. Anaerobe 2007; 9:145-9. [PMID: 16887702 DOI: 10.1016/s1075-9964(03)00069-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2002] [Revised: 04/11/2003] [Accepted: 05/20/2003] [Indexed: 11/21/2022]
Abstract
Clostridium perfringens type D produces enterotoxaemia in sheep, goats and other animals. The disease is caused by C. perfringens epsilon toxin, and while enterotoxaemia in goats is usually characterized by enterocolitis, the disease in sheep is characterized by systemic lesions (such as lung and brain oedema) with minor and inconsistent changes observed in the intestine. A possible explanation for these differences is that epsilon toxin is more promptly absorbed by sheep than goat intestine. In an attempt to clarify this, we examined the in vitro effects of epsilon toxin on sheep and goat intestine. Pieces of intestinal mucosa from recently slaughtered animals were mounted in a modified Ussing-type chamber where net water flux (J(w)), short-circuit current (I(sc)) and tissue conductance (G(t)) were simultaneously recorded. After 70 min of incubation with epsilon toxin a reduction in absorptive J(w) and an increase in I(sc) and G(t) were observed in colonic tissues of both sheep and goats, but no alterations were registered in the ileum of either species. These in vitro results show that epsilon toxin affects the transport function of the colonic mucosa but it does not seem to produce any transport alteration in the ileum mucosa.
Collapse
Affiliation(s)
- M E Fernandez Miyakawa
- Unidad de Salud Animal, Instituto Nacional de Tecnología Agropecuaria, Bariloche, Argentina
| | | | | |
Collapse
|
86
|
Chassin C, Bens M, de Barry J, Courjaret R, Bossu JL, Cluzeaud F, Ben Mkaddem S, Gibert M, Poulain B, Popoff MR, Vandewalle A. Pore-forming epsilon toxin causes membrane permeabilization and rapid ATP depletion-mediated cell death in renal collecting duct cells. Am J Physiol Renal Physiol 2007; 293:F927-37. [PMID: 17567938 DOI: 10.1152/ajprenal.00199.2007] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Clostridium perfringens epsilon toxin (ET) is a potent pore-forming cytotoxin causing fatal enterotoxemia in livestock. ET accumulates in brain and kidney, particularly in the renal distal-collecting ducts. ET binds and oligomerizes in detergent-resistant membranes (DRMs) microdomains and causes cell death. However, the causal linkage between membrane permeabilization and cell death is not clear. Here, we show that ET binds and forms 220-kDa insoluble complexes in plasma membrane DRMs of renal mpkCCD(cl4) collecting duct cells. Phosphatidylinositol-specific phospholipase C did not impair binding or the formation of ET complexes, suggesting that the receptor for ET is not GPI anchored. ET induced a dose-dependent fall in the transepithelial resistance and potential in confluent cells grown on filters, transiently stimulated Na+ absorption, and induced an inward ionic current and a sustained rise in [Ca2+]i. ET also induced rapid depletion of cellular ATP, and stimulated the AMP-activated protein kinase, a metabolic-sensing Ser/Thr kinase. ET also induced mitochondrial membrane permeabilization and mitochondrial-nuclear translocation of apoptosis-inducing factor, a potent caspase-independent cell death effector. Finally, ET induced cell necrosis characterized by a marked reduction in nucleus size without DNA fragmentation. DRM disruption by methyl-beta-cyclodextrin impaired ET oligomerization, and significantly reduced the influx of Na+ and [Ca2+]i, but did not impair ATP depletion and cell death caused by the toxin. These findings indicate that ET causes rapid necrosis of renal collecting duct cells and establish that ATP depletion-mediated cell death is not strictly correlated with the plasma membrane permeabilization and ion diffusion caused by the toxin.
Collapse
Affiliation(s)
- C Chassin
- Institut National de la Santé et de la Recherche Médicale U773, Centre de Recherche Biomédicale Bichat-Beaujon CRB3, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Soler-Jover A, Dorca J, Popoff MR, Gibert M, Saura J, Tusell JM, Serratosa J, Blasi J, Martín-Satué M. Distribution of Clostridium perfringens epsilon toxin in the brains of acutely intoxicated mice and its effect upon glial cells. Toxicon 2007; 50:530-40. [PMID: 17572464 DOI: 10.1016/j.toxicon.2007.04.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 04/24/2007] [Accepted: 04/25/2007] [Indexed: 10/23/2022]
Abstract
Epsilon toxin (epsilon-toxin), produced by Clostridium perfringens types B and D, causes fatal enterotoxaemia in livestock. The disease is principally manifested as severe and often fatal neurological disturbance. Oedema of several organs, including the brain, is also a clinical sign related to microvascular damage. Recombinant epsilon-toxin-green fluorescence protein (epsilon-toxin-GFP) and epsilon-prototoxin-GFP have already been characterised as useful tools to track their distribution in intravenously injected mice, by means of direct fluorescence microscopy detection. The results shown here, using an acutely intoxicated mouse model, strongly suggest that epsilon-toxin-GFP, but not epsilon-prototoxin-GFP, not only causes oedema but is also able to cross the blood-brain barrier and accumulate in brain tissue. In some brain areas, epsilon-toxin-GFP is found bound to glial cells, both astrocytes and microglia. Moreover, cytotoxicity assays, performed with mixed glial primary cultures, demonstrate the cytotoxic effect of epsilon-toxin upon both astrocytes and microglial cells.
Collapse
Affiliation(s)
- Alex Soler-Jover
- Laboratori de Neurobiologia Cellular i Molecular, Departament de Patologia i Terapèutica Experimental, Campus de Bellvitge, Universitat de Barcelona-IDIBELL, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
McClain MS, Cover TL. Functional analysis of neutralizing antibodies against Clostridium perfringens epsilon-toxin. Infect Immun 2007; 75:1785-93. [PMID: 17261609 PMCID: PMC1865726 DOI: 10.1128/iai.01643-06] [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] [Received: 10/11/2006] [Revised: 11/13/2006] [Accepted: 01/15/2007] [Indexed: 11/20/2022] Open
Abstract
The Clostridium perfringens epsilon-toxin causes a severe, often fatal illness (enterotoxemia) characterized by cardiac, pulmonary, kidney, and brain edema. In this study, we examined the activities of two neutralizing monoclonal antibodies against the C. perfringens epsilon-toxin. Both antibodies inhibited epsilon-toxin cytotoxicity towards cultured MDCK cells and inhibited the ability of the toxin to form pores in the plasma membranes of cells, as shown by staining cells with the membrane-impermeant dye 7-aminoactinomycin D. Using an antibody competition enzyme-linked immunosorbent assay (ELISA), a peptide array, and analysis of mutant toxins, we mapped the epitope recognized by one of the neutralizing monoclonal antibodies to amino acids 134 to 145. The antibody competition ELISA and analysis of mutant toxins suggest that the second neutralizing monoclonal antibody also recognizes an epitope in close proximity to this region. The region comprised of amino acids 134 to 145 overlaps an amphipathic loop corresponding to the putative membrane insertion domain of the toxin. Identifying the epitopes recognized by these neutralizing antibodies constitutes an important first step in the development of therapeutic agents that could be used to counter the effects of the epsilon-toxin.
Collapse
Affiliation(s)
- Mark S McClain
- Department of Medicine, Division of Infectious Diseases, A2200 Medical Center North, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | |
Collapse
|
89
|
Katayama H, Kusaka Y, Yokota H, Akao T, Kojima M, Nakamura O, Mekada E, Mizuki E. Parasporin-1, a Novel Cytotoxic Protein from Bacillus thuringiensis, Induces Ca2+ Influx and a Sustained Elevation of the Cytoplasmic Ca2+ Concentration in Toxin-sensitive Cells. J Biol Chem 2007; 282:7742-52. [PMID: 17204466 DOI: 10.1074/jbc.m611382200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Parasporin-1 is a novel non-insecticidal inclusion protein from Bacillus thuringiensis that is cytotoxic to specific mammalian cells. In this study, we investigated the effects of parasporin-1 on toxin-sensitive cell lines to elucidate the cytotoxic mechanism of parasporin-1. Parasporin-1 is not a membrane pore-forming toxin as evidenced by measurements of lactate dehydrogenase release, propidium iodide penetration, and membrane potential in parasporin-1-treated cells. Parasporin-1 decreased the level of cellular protein and DNA synthesis in parasporin-1-sensitive HeLa cells. The earliest change observed in cells treated with this toxin was a rapid elevation of the intracellular free-Ca(2+) concentration; increases in the intracellular Ca(2+) levels were observed 1-3 min following parasporin-1 treatment. Using four different cell lines, we found that the degree of cellular sensitivity to parasporin-1 was positively correlated with the size of the increase in the intracellular Ca(2+) concentration. The toxin-induced elevation of the intracellular Ca(2+) concentration was markedly decreased in low-Ca(2+) buffer and was not observed in Ca(2+)-free buffer. Accordingly, the cytotoxicity of parasporin-1 decreased in the low-Ca(2+) buffer and was restored by the addition of Ca(2+) to the extracellular medium. Suramin, which inhibits trimeric G-protein signaling, suppressed both the Ca(2+) influx and the cytotoxicity of parasporin-1. In parasporin-1-treated HeLa cells, degradation of pro-caspase-3 and poly(ADP-ribose) polymerase was observed. Furthermore, synthetic caspase inhibitors blocked the cytotoxic activity of parasporin-1. These results indicate that parasporin-1 activates apoptotic signaling in these cells as a result of the increased Ca(2+) level and that the Ca(2+) influx is the first step in the pathway that underlies parasporin-1 toxicity.
Collapse
Affiliation(s)
- Hideki Katayama
- Biotechnology and Food Research Institute, Fukuoka Industrial Technology Center, Aikawa, Kurume, Fukuoka 839-0861, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Kitada S, Abe Y, Shimada H, Kusaka Y, Matsuo Y, Katayama H, Okumura S, Akao T, Mizuki E, Kuge O, Sasaguri Y, Ohba M, Ito A. Cytocidal Actions of Parasporin-2, an Anti-tumor Crystal Toxin from Bacillus thuringiensis. J Biol Chem 2006; 281:26350-60. [PMID: 16809341 DOI: 10.1074/jbc.m602589200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Parasporin-2, a new crystal protein derived from noninsecticidal and nonhemolytic Bacillus thuringiensis, recognizes and kills human liver and colon cancer cells as well as some classes of human cultured cells. Here we report that a potent proteinase K-resistant parasporin-2 toxin shows specific binding to and a variety of cytocidal effects against human hepatocyte cancer cells. Cleavage of the N-terminal region of parasporin-2 was essential for the toxin activity, whereas C-terminal digestion was required for rapid cell injury. Protease-activated parasporin-2 induced remarkable morphological alterations, cell blebbing, cytoskeletal alterations, and mitochondrial and endoplasmic reticulum fragmentation. The plasma membrane permeability was increased immediately after the toxin treatment and most of the cytoplasmic proteins leaked from the cells, whereas mitochondrial and endoplasmic reticulum proteins remained in the intoxicated cells. Parasporin-2 selectively bound to cancer cells in slices of liver tumor tissues and susceptible human cultured cells and became localized in the plasma membrane until the cells were damaged. Thus, parasporin-2 acts as a cytolysin that permeabilizes the plasma membrane with target cell specificity and subsequently induces cell decay.
Collapse
Affiliation(s)
- Sakae Kitada
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka 812-8581, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Borrmann E, Schulze F, Cussler K, Hänel I, Diller R. Development of a cell culture assay for the quantitative determination of vaccination-induced antibodies in rabbit sera against Clostridium perfringens epsilon toxin and Clostridium novyi alpha toxin. Vet Microbiol 2005; 114:41-50. [PMID: 16361070 DOI: 10.1016/j.vetmic.2005.10.043] [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: 04/07/2005] [Revised: 10/27/2005] [Accepted: 10/28/2005] [Indexed: 11/16/2022]
Abstract
Cell culture assays are possible alternatives to replace in vivo neutralization tests currently required for potency testing of clostridial vaccines. Cell culture assays based on the MDCK cell line and the Vero cell line which are sensitive to the Clostridium (C.) perfringens type D epsilon toxin and Clostridium novyi type B alpha toxin, respectively, were developed, and the test conditions were standardized. The antibody titres of vaccinated rabbits measured in vitro were compared with the results of current test procedures recommended by European Pharmacopoeia. The correlation coefficients calculated were significant for all sera tested. The cell culture assays proved to be sensitive, specific, reproducible and reliable. Therefore, these cell culture assays could be suitable in vitro alternatives to the in vivo mouse neutralization experiments required for potency tests of clostridial vaccines, but further validation studies are necessary.
Collapse
Affiliation(s)
- E Borrmann
- Institute of Molecular Pathogenesis at the Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut, Naumburger Str. 96 a, D-07743 Jena, Germany.
| | | | | | | | | |
Collapse
|
92
|
Adamson RH, Ly JC, Fernandez-Miyakawa M, Ochi S, Sakurai J, Uzal F, Curry FE. Clostridium perfringens epsilon-toxin increases permeability of single perfused microvessels of rat mesentery. Infect Immun 2005; 73:4879-87. [PMID: 16041001 PMCID: PMC1201274 DOI: 10.1128/iai.73.8.4879-4887.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epsilon-toxin, the primary virulence factor of Clostridium perfringens type D, causes mortality in livestock, particularly sheep and goats, in which it induces an often-fatal enterotoxemia. It is believed to compromise the intestinal barrier and then enter the gut vasculature, from which it is carried systemically, causing widespread vascular endothelial damage and edema. Here we used single perfused venular microvessels in rat mesentery, which enabled direct observation of permeability properties of the in situ vascular wall during exposure to toxin. We determined the hydraulic conductivity (L(p)) of microvessels as a measure of the response to epsilon-toxin. We found that microvessels were highly sensitive to toxin. At 10 microg ml(-1) the L(p) increased irreversibly to more than 15 times the control value by 10 min. At 0.3 microg ml(-1) no increase in L(p) was observed for up to 90 min. The toxin-induced increase in L(p) was consistent with changes in ultrastructure of microvessels exposed to the toxin. Those microvessels exhibited gaps either between or through endothelial cells where perfusate had direct access to the basement membrane. Many endothelial cells appeared necrotic, highly attenuated, and with dense cytoplasm. We showed that epsilon-toxin, in a time- and dose-dependent manner, rapidly and irreversibly compromised the barrier function of venular microvessel endothelium. The results conformed to the hypothesis that epsilon-toxin interacts with vascular endothelial cells and increases the vessel wall permeability by direct damage of the endothelium.
Collapse
Affiliation(s)
- R H Adamson
- Department of Physiology and Membrane Biology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| | | | | | | | | | | | | |
Collapse
|
93
|
Czajkowsky DM, Iwamoto H, Szabo G, Cover TL, Shao Z. Mimicry of a host anion channel by a Helicobacter pylori pore-forming toxin. Biophys J 2005; 89:3093-101. [PMID: 16100263 PMCID: PMC1366806 DOI: 10.1529/biophysj.105.066746] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bacterial pore-forming toxins have traditionally been thought to function either by causing an essentially unrestricted flux of ions and molecules across a membrane or by effecting the transmembrane transport of an enzymatically active bacterial peptide. However, the Helicobacter pylori pore-forming toxin, VacA, does not appear to function by either of these mechanisms, even though at least some of its effects in cells are dependent on its pore-forming ability. Here we show that the VacA channel exhibits two of the most characteristic electrophysiological properties of a specific family of cellular channels, the ClC channels: an open probability dependent on the molar ratio of permeable ions and single channel events resolvable as two independent, voltage-dependent transitions. The sharing of such peculiar properties by VacA and host ClC channels, together with their similar magnitudes of conductance, ion selectivities, and localization within eukaryotic cells, suggests a novel mechanism of toxin action in which the VacA pore largely mimics the electrophysiological behavior of a host channel, differing only in the membrane potential at which it closes. As a result, VacA can perturb, but not necessarily abolish, the homeostatic ionic imbalance across a membrane and so change cellular physiology without necessarily jeopardizing vitality.
Collapse
Affiliation(s)
- Daniel M Czajkowsky
- Department of Molecular Physiology and Biological Physics, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
94
|
Puthoff DP, Sardesai N, Subramanyam S, Nemacheck JA, Williams CE. Hfr-2, a wheat cytolytic toxin-like gene, is up-regulated by virulent Hessian fly larval feedingdouble dagger. MOLECULAR PLANT PATHOLOGY 2005; 6:411-423. [PMID: 20565667 DOI: 10.1111/j.1364-3703.2005.00289.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
SUMMARY Both yield and grain-quality are dramatically decreased when susceptible wheat (Triticum aestivum) plants are infested by Hessian fly (Mayetiola destructor) larvae. Examination of the changes in wheat gene expression during infestation by virulent Hessian fly larvae has identified the up-regulation of a gene, Hessian fly responsive-2 (Hfr-2), which contains regions similar to genes encoding seed-specific agglutinin proteins from Amaranthus. Hfr-2, however, did not accumulate in developing seeds, as do other wheat seed storage proteins. Additionally, a separate region of the HFR-2 predicted amino acid sequence is similar to haemolytic proteins, from both mushroom and bacteria, that are able to form pores in cell membranes of mammalian red blood cells. The involvement of Hfr-2 in interactions with insects was supported by experiments demonstrating its up-regulation by both fall armyworm (Spodoptera frugiperda) and bird cherry-oat aphid (Rhopalosiphum padi) infestations but not by virus infection. Examination of wheat defence response pathways showed Hfr-2 up-regulation following methyl jasmonate treatment and only slight up-regulation in response to salicylic acid, abscisic acid and wounding treatments. Like related proteins, HFR-2 may normally function in defence against certain insects or pathogens. However, we propose that as virulent Hessian fly larvae manipulate the physiology of the susceptible host, the HFR-2 protein inserts in plant cell membranes at the feeding sites and by forming pores provides water, ions and other small nutritive molecules to the developing larvae.
Collapse
Affiliation(s)
- David P Puthoff
- USDA-ARS Crop Production and Pest Control Research Unit, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
95
|
Mantis NJ. Vaccines against the category B toxins: Staphylococcal enterotoxin B, epsilon toxin and ricin. Adv Drug Deliv Rev 2005; 57:1424-39. [PMID: 15935880 DOI: 10.1016/j.addr.2005.01.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2004] [Accepted: 01/25/2005] [Indexed: 01/15/2023]
Abstract
The threat of bioterrorism worldwide has accelerated the demand for the development of therapies and vaccines against the Category B toxins: staphylococcal enterotoxin B (SEB), epsilon toxin (ETX) produced by Clostridium perfringens types B and D, and ricin, a natural product of the castor bean. The diverse and unique nature of these toxins poses a challenge to vaccinologists. While formalin-inactivated toxins can successfully induce antibody-mediated protection in animals, their usefulness in humans is limited because of potential safety concerns. For this reason, research is now aimed at developing recombinant, attenuated vaccines based on a detailed understanding of the molecular mechanisms by which these toxins function. Vaccine development is further complicated by the fact that as bioterrorism agents, SEB, ETX and ricin would most likely be disseminated as aerosols or in food/water supplies. Our understanding of the mechanisms by which these toxins cross mucosal surfaces, and importance of mucosal immunity in preventing toxin uptake is only rudimentary.
Collapse
Affiliation(s)
- Nicholas J Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA.
| |
Collapse
|
96
|
Shimamoto S, Tamai E, Matsushita O, Minami J, Okabe A, Miyata S. Changes in ganglioside content affect the binding of Clostridium perfringens epsilon-toxin to detergent-resistant membranes of Madin-Darby canine kidney cells. Microbiol Immunol 2005; 49:245-53. [PMID: 15781998 DOI: 10.1111/j.1348-0421.2005.tb03726.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epsilon-toxin (ET) of Clostridium perfringens, which causes fatal enterotoxemia in ungulates, was previously shown to bind to and form a heptameric pore within the detergent-resistant membranes (DRMs) of MDCK cells. Depletion of cholesterol has also been shown to decrease the cytotoxicity of ET and its heptamerization. In this study, we investigated the effects of changes in sphingolipids, other DRM components of MDCK cells, on the cells' susceptibility to ET. Treatment with fumonisin B1 and PDMP, inhibitors of sphingolipid and glycosphingolipid syntheses, respectively, increased the susceptibility, while D609, a sphingomyelin synthesis inhibitor, had the opposite effect. The exogenous addition of ganglioside G(M1) dramatically decreased the ET binding, heptamerization and cytotoxicity. These effects were shown not to be due to ET binding to G(M1) or to denaturation of ET. We also found that the ET cytotoxicity towards MDCK cells decreased with an increase in culture time. In accordance with the resistance observed for prolonged cultured cells, G(M3), a major ganglioside component, increased and sialidase treatment increased their susceptibility. These results suggest that membrane-anchored sialic acid of G(M3) within DRMs inhibits ET binding, leading to prevention of the heptamerization of ET and cell death. It is also suggested that sialidase produced by this organism aids the targeting of ET to MDCK cells.
Collapse
Affiliation(s)
- Seiko Shimamoto
- Department of Microbiology, Faculty of Medicine, Kagawa University, Kita-gun, Japan
| | | | | | | | | | | |
Collapse
|
97
|
Smedley JG, Fisher DJ, Sayeed S, Chakrabarti G, McClane BA. The enteric toxins of Clostridium perfringens. Rev Physiol Biochem Pharmacol 2004; 152:183-204. [PMID: 15517462 DOI: 10.1007/s10254-004-0036-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The Gram-positive pathogen Clostridium perfringens is a major cause of human and veterinary enteric disease largely because this bacterium can produce several toxins when present inside the gastrointestinal tract. The enteric toxins of C. perfringens share two common features: (1) they are all single polypeptides of modest (approximately 25-35 kDa) size, although lacking in sequence homology, and (2) they generally act by forming pores or channels in plasma membranes of host cells. These enteric toxins include C. perfringens enterotoxin (CPE), which is responsible for the symptoms of a common human food poisoning and acts by forming pores after interacting with intestinal tight junction proteins. Two other C. perfringens enteric toxins, epsilon-toxin (a bioterrorism select agent) and beta-toxin, cause veterinary enterotoxemias when absorbed from the intestines; beta- and epsilon-toxins then apparently act by forming oligomeric pores in intestinal or extra-intestinal target tissues. The action of a newly discovered C. perfringens enteric toxin, beta2 toxin, has not yet been defined but precedent suggests it might also be a pore-former. Experience with other clostridial toxins certainly warrants continued research on these C. perfringens enteric toxins to develop their potential as therapeutic agents and tools for cellular biology.
Collapse
Affiliation(s)
- J G Smedley
- University of Pittsburgh School of Medicine, Department of Molecular Genetics and Biochemistry, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
98
|
Soler-Jover A, Blasi J, Gómez de Aranda I, Navarro P, Gibert M, Popoff MR, Martín-Satué M. Effect of epsilon toxin-GFP on MDCK cells and renal tubules in vivo. J Histochem Cytochem 2004; 52:931-42. [PMID: 15208360 DOI: 10.1369/jhc.4a6254.2004] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epsilon toxin (epsilon-toxin), produced by Clostridium perfringens types B and D, causes fatal enterotoxemia, also known as pulpy kidney disease, in livestock. Recombinant epsilon-toxin-green fluorescence protein (epsilon-toxin-GFP) and epsilon-prototoxin-GFP were successfully expressed in Escherichia coli. MTT assays on MDCK cells confirmed that recombinant epsilon-toxin-GFP retained the cytotoxicity of the native toxin. Direct fluorescence analysis of MDCK cells revealed a homogeneous peripheral pattern that was temperature sensitive and susceptible to detergent. epsilon-Toxin-GFP and epsilon-prototoxin-GFP bound to endothelia in various organs of injected mice, especially the brain. However, fluorescence mainly accumulated in kidneys. Mice injected with epsilon-toxin-GFP showed severe kidney alterations, including hemorrhagic medullae and selective degeneration of distal tubules. Moreover, experiments on kidney cryoslices demonstrated specific binding to distal tubule cells of a range of species. We demonstrate with new recombinant fluorescence tools that epsilon-toxin binds in vivo to endothelial cells and renal tubules, where it has a strong cytotoxic effect. Our binding experiments indicate that an epsilon-toxin receptor is expressed on renal distal tubules of mammalian species, including human.
Collapse
Affiliation(s)
- Alex Soler-Jover
- Department de Biologia Cellular i Anatomia Patològica, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
99
|
Cole AR, Gibert M, Popoff M, Moss DS, Titball RW, Basak AK. Clostridium perfringens ε-toxin shows structural similarity to the pore-forming toxin aerolysin. Nat Struct Mol Biol 2004; 11:797-8. [PMID: 15258571 DOI: 10.1038/nsmb804] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Accepted: 05/20/2004] [Indexed: 11/09/2022]
Abstract
Epsilon-toxin from Clostridium perfringens is a lethal toxin. Recent studies suggest that the toxin acts via an unusually potent pore-forming mechanism. Here we report the crystal structure of epsilon-toxin, which reveals structural similarity to aerolysin from Aeromonas hydrophila. Pore-forming toxins can change conformation between soluble and transmembrane states. By comparing the two toxins, we have identified regions important for this transformation.
Collapse
Affiliation(s)
- Ambrose R Cole
- Department of Crystallography, Birkbeck College, Malet Street, London WC1E 7HX, UK
| | | | | | | | | | | |
Collapse
|
100
|
Rings DM. Clostridial disease associated with neurologic signs: tetanus, botulism, and enterotoxemia. Vet Clin North Am Food Anim Pract 2004; 20:379-91, vii-viii. [PMID: 15203231 DOI: 10.1016/j.cvfa.2004.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clostridial infections are found worldwide in almost all species of animals and may involve a variety of body systems and present with a diversity of clinical signs. Most damage done through clostridial infections is due to the action of toxins released from the bacteria.Thus, disease caused by Clostridium spp should more correctly be called intoxication. Two prominent clostridial infections are associated with neurologic signs: Clostridium botulinum and C tetani. In both infections, the mechanism that is responsible for causing the problem is similar, despite the remarkable difference in clinical presentation. In addition, neurologic signs are described with C perfringens types C and D but are not the dominant feature of these diseases.
Collapse
Affiliation(s)
- D Michael Rings
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Tharp Street, Columbus, OH 43210, USA.
| |
Collapse
|