1
|
Cai S, Kumar R, Singh BR. Clostridial Neurotoxins: Structure, Function and Implications to Other Bacterial Toxins. Microorganisms 2021; 9:2206. [PMID: 34835332 PMCID: PMC8618262 DOI: 10.3390/microorganisms9112206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 01/20/2023] Open
Abstract
Gram-positive bacteria are ancient organisms. Many bacteria, including Gram-positive bacteria, produce toxins to manipulate the host, leading to various diseases. While the targets of Gram-positive bacterial toxins are diverse, many of those toxins use a similar mechanism to invade host cells and exert their functions. Clostridial neurotoxins produced by Clostridial tetani and Clostridial botulinum provide a classical example to illustrate the structure-function relationship of bacterial toxins. Here, we critically review the recent progress of the structure-function relationship of clostridial neurotoxins, including the diversity of the clostridial neurotoxins, the mode of actions, and the flexible structures required for the activation of toxins. The mechanism clostridial neurotoxins use for triggering their activity is shared with many other Gram-positive bacterial toxins, especially molten globule-type structures. This review also summarizes the implications of the molten globule-type flexible structures to other Gram-positive bacterial toxins. Understanding these highly dynamic flexible structures in solution and their role in the function of bacterial toxins not only fills in the missing link of the high-resolution structures from X-ray crystallography but also provides vital information for better designing antidotes against those toxins.
Collapse
Affiliation(s)
- Shuowei Cai
- Department of Chemistry and Biochemistry, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Raj Kumar
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA; (R.K.); (B.R.S.)
| | - Bal Ram Singh
- Botulinum Research Center, Institute of Advanced Sciences, Dartmouth, MA 02747, USA; (R.K.); (B.R.S.)
| |
Collapse
|
2
|
Harnessing the Membrane Translocation Properties of AB Toxins for Therapeutic Applications. Toxins (Basel) 2021; 13:toxins13010036. [PMID: 33418946 PMCID: PMC7825107 DOI: 10.3390/toxins13010036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/28/2020] [Accepted: 01/01/2021] [Indexed: 12/31/2022] Open
Abstract
Over the last few decades, proteins and peptides have become increasingly more common as FDA-approved drugs, despite their inefficient delivery due to their inability to cross the plasma membrane. In this context, bacterial two-component systems, termed AB toxins, use various protein-based membrane translocation mechanisms to deliver toxins into cells, and these mechanisms could provide new insights into the development of bio-based drug delivery systems. These toxins have great potential as therapies both because of their intrinsic properties as well as the modular characteristics of both subunits, which make them highly amenable to conjugation with various drug classes. This review focuses on the therapeutical approaches involving the internalization mechanisms of three representative AB toxins: botulinum toxin type A, anthrax toxin, and cholera toxin. We showcase several specific examples of the use of these toxins to develop new therapeutic strategies for numerous diseases and explain what makes these toxins promising tools in the development of drugs and drug delivery systems.
Collapse
|
3
|
The 25 kDa H CN Domain of Clostridial Neurotoxins Is Indispensable for Their Neurotoxicity. Toxins (Basel) 2020; 12:toxins12120743. [PMID: 33255952 PMCID: PMC7760224 DOI: 10.3390/toxins12120743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 11/17/2022] Open
Abstract
The extraordinarily potent clostridial neurotoxins (CNTs) comprise tetanus neurotoxin (TeNT) and the seven established botulinum neurotoxin serotypes (BoNT/A-G). They are composed of four structurally independent domains: the roles of the catalytically active light chain, the translocation domain HN, and the C-terminal receptor binding domain HCC are largely resolved, but that of the HCN domain sandwiched between HN and HCC has remained unclear. Here, mutants of BoNT/A, BoNT/B, and TeNT were generated by deleting their HCN domains or swapping HCN domains between each other. Both deletion and replacement of TeNT HCN domain by HCNA and HCNB reduced the biological activity similarly, by ~95%, whereas BoNT/A and B deletion mutants displayed >500-fold reduced activity in the mouse phrenic nerve hemidiaphragm assay. Swapping HCN domains between BoNT/A and B hardly impaired their biological activity, but substitution with HCNT did. Binding assays revealed that in the absence of HCN, not all receptor binding sites are equally well accessible. In conclusion, the presence of HCN is vital for CNTs to exert their neurotoxicity. Although structurally similar, the HCN domain of TeNT cannot equally substitute those of BoNT and vice versa, leaving the possibility that HCNT plays a different role in the intoxication mechanism of TeNT.
Collapse
|
4
|
Melkoumov A, St-Jean I, Banquy X, Leclair G, Leblond Chain J. GM1-Binding Conjugates To Improve Intestinal Permeability. Mol Pharm 2018; 16:60-70. [PMID: 30422668 DOI: 10.1021/acs.molpharmaceut.8b00776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Drugs and proteins with poor intestinal permeability have a limited oral bioavailability. To remediate this problem, a receptor-mediated endocytosis and transcytosis approach was explored. Indeed, the nontoxic β subunit of cholera toxin (CTB) can cross the intestinal barrier by binding to receptor GM1. In this study, we explored the use of GM1-binding peptides and CTB as potential covalent carriers of poorly permeable molecules. GM1-binding peptides (G23, P3) and CTB were conjugated to poorly permeable fluorescent probes such as fluorescein isothiocyanate (FITC) and albumin-FITC using triethylene glycol spacers and click chemistry. The affinity of the peptide conjugates with receptor GM1 was confirmed by isothermal titration calorimetry or microscale thermophoresis, and the results suggested the involvement of nonspecific interactions. Conjugating the model drugs to G23 and P3 improved the internalization into Caco-2 and T84 cells, although the process was not dependent on the amount of GM1 receptor. However, conjugation of bovine serum albumin FITC to CTB increased the internalization in the same cells in a GM1-dependent pathway. Peptide conjugates demonstrated a limited permeability through a Caco-2 monolayer, whereas G23 and CTB conjugates slightly enhanced permeability through a T84 cell monolayer compared to model drugs alone. Since CTB can improve the permeability of large macromolecules such as albumin, it is an interesting carrier for the improvement of oral bioavailability of various other macromolecules such as heparins, proteins, and siRNAs.
Collapse
Affiliation(s)
- Alexandre Melkoumov
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Isabelle St-Jean
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Xavier Banquy
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Grégoire Leclair
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| | - Jeanne Leblond Chain
- Faculty of Pharmacy , Université de Montréal , H3C 3J7 Montréal , Québec , Canada
| |
Collapse
|
5
|
Variability of Botulinum Toxins: Challenges and Opportunities for the Future. Toxins (Basel) 2018; 10:toxins10090374. [PMID: 30217070 PMCID: PMC6162648 DOI: 10.3390/toxins10090374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/03/2018] [Accepted: 09/08/2018] [Indexed: 12/31/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are the most potent known toxins, and are therefore classified as extremely harmful biological weapons. However, BoNTs are therapeutic drugs that are widely used and have an increasing number of applications. BoNTs show a high diversity and are divided into multiple types and subtypes. Better understanding of the activity at the molecular and clinical levels of the natural BoNT variants as well as the development of BoNT-based chimeric molecules opens the door to novel medical applications such as silencing the sensory neurons at targeted areas and dermal restoration. This short review is focused on BoNTs’ variability and the opportunities or challenges posed for future clinical applications.
Collapse
|
6
|
Ghosal KJ, Patel K, Singh BR, Hale ML. Role of critical elements in botulinum neurotoxin complex in toxin routing across intestinal and bronchial barriers. PLoS One 2018; 13:e0199524. [PMID: 29975725 PMCID: PMC6033393 DOI: 10.1371/journal.pone.0199524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/08/2018] [Indexed: 11/18/2022] Open
Abstract
The highly potent botulinum neurotoxin serotype A (BoNT/A) inhibits neurotransmitter release at neuromuscular junctions resulting in flaccid muscle paralysis, respiratory arrest and death. In order to reach their neuronal cell targets, BoNT/A must cross epithelial cell barriers lining the intestines and airways. The toxin is produced as a large protein complex comprised of the neurotoxin and non-toxic neurotoxin-associated proteins (NAPs). Although NAPs are known to protect the toxin from harsh environments, their role in the movement of BoNT/A across epithelial barriers has not been fully characterized. In the current study, movement of the toxin across epithelial cells was examined macroscopically using a sensitive near infrared fluorescence transcytosis assay and microscopically using fluorescently labeled toxin and confocal microscopy. The studies show that the BoNT/A complex internalizes more rapidly than the pure toxin. The studies also show that one NAP protein, hemaglutinin 33 (Hn33), enhanced both the binding and movement of a deactivated recombinant botulinum neurotoxin A (DrBoNT) across epithelial cell monolayers and that the toxin associates with Hn33 on the cell surface. Collectively, the data demonstrate that, in addition to their protective role, NAPs and Hn33 play an important role in BoNT/A intoxication.
Collapse
Affiliation(s)
- Koyel J. Ghosal
- Department of Chemistry and Biochemistry, University of Massachusetts Dartmouth, North Dartmouth, Massachusetts, United States of America
| | - Kruti Patel
- Department of Chemistry and Biochemistry, University of Massachusetts Dartmouth, North Dartmouth, Massachusetts, United States of America
| | - Bal Ram Singh
- Department of Chemistry and Biochemistry, University of Massachusetts Dartmouth, North Dartmouth, Massachusetts, United States of America
| | - Martha L. Hale
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| |
Collapse
|
7
|
Garcia-Castillo MD, Chinnapen DJF, Lencer WI. Membrane Transport across Polarized Epithelia. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027912. [PMID: 28213463 DOI: 10.1101/cshperspect.a027912] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Polarized epithelial cells line diverse surfaces throughout the body forming selective barriers between the external environment and the internal milieu. To cross these epithelial barriers, large solutes and other cargoes must undergo transcytosis, an endocytic pathway unique to polarized cell types, and significant for the development of cell polarity, uptake of viral and bacterial pathogens, transepithelial signaling, and immunoglobulin transport. Here, we review recent advances in our knowledge of the transcytotic pathway for proteins and lipids. We also discuss briefly the promise of harnessing the molecules that undergo transcytosis as vehicles for clinical applications in drug delivery.
Collapse
Affiliation(s)
| | - Daniel J-F Chinnapen
- Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Digestive Diseases Center, Boston, Massachusetts 02155
| | - Wayne I Lencer
- Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Digestive Diseases Center, Boston, Massachusetts 02155
| |
Collapse
|
8
|
Yang NJ, Chiu IM. Bacterial Signaling to the Nervous System through Toxins and Metabolites. J Mol Biol 2017; 429:587-605. [PMID: 28065740 PMCID: PMC5325782 DOI: 10.1016/j.jmb.2016.12.023] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/21/2016] [Accepted: 12/29/2016] [Indexed: 12/31/2022]
Abstract
Mammalian hosts interface intimately with commensal and pathogenic bacteria. It is increasingly clear that molecular interactions between the nervous system and microbes contribute to health and disease. Both commensal and pathogenic bacteria are capable of producing molecules that act on neurons and affect essential aspects of host physiology. Here we highlight several classes of physiologically important molecular interactions that occur between bacteria and the nervous system. First, clostridial neurotoxins block neurotransmission to or from neurons by targeting the SNARE complex, causing the characteristic paralyses of botulism and tetanus during bacterial infection. Second, peripheral sensory neurons-olfactory chemosensory neurons and nociceptor sensory neurons-detect bacterial toxins, formyl peptides, and lipopolysaccharides through distinct molecular mechanisms to elicit smell and pain. Bacteria also damage the central nervous system through toxins that target the brain during infection. Finally, the gut microbiota produces molecules that act on enteric neurons to influence gastrointestinal motility, and metabolites that stimulate the "gut-brain axis" to alter neural circuits, autonomic function, and higher-order brain function and behavior. Furthering the mechanistic and molecular understanding of how bacteria affect the nervous system may uncover potential strategies for modulating neural function and treating neurological diseases.
Collapse
Affiliation(s)
- Nicole J Yang
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Nelms B, Dalomba NF, Lencer W. A targeted RNAi screen identifies factors affecting diverse stages of receptor-mediated transcytosis. J Cell Biol 2017; 216:511-525. [PMID: 28069747 PMCID: PMC5294788 DOI: 10.1083/jcb.201609035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/22/2016] [Accepted: 12/20/2016] [Indexed: 11/22/2022] Open
Abstract
Transcytosis plays an important role in establishing cell polarity and in mediating transport of large cargo across epithelial barriers, but its molecular basis is unclear. Nelms et al. present a new dataset of genes involved in receptor-mediated transcytosis and show that the apical and basolateral recycling and transcytotic pathways are genetically separable. Endosome transport by transcytosis is the primary mechanism by which proteins and other large cargo traverse epithelial barriers in normal tissue. Transcytosis is also essential for establishing and maintaining membrane polarity in epithelia and other polarized cells. To identify novel components of this pathway, we conducted a high-throughput RNA interference screen for factors necessary for the bidirectional transcytosis of IgG by the Fcγ receptor FcRn. This screen identified 23 genes whose suppression resulted in a reproducible decrease in FcRn-mediated transcytosis. Pulse-chase kinetic transport assays on four of the top-ranking genes (EXOC2, EXOC7, PARD6B, and LEPROT) revealed distinct effects on the apical and basolateral recycling and transcytotic pathways, demonstrating that these pathways are genetically separable. We also found a strong dependence on PARD6B for apical, but not basolateral, recycling, implicating this cell polarity gene in assembly or maintenance of the apical endosomal system. This dataset yields insights into how vesicular transport is adapted to the specialized functions of differentiated cell types and opens new research avenues into epithelial trafficking.
Collapse
Affiliation(s)
- Bradlee Nelms
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115.,Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138
| | - Natasha Furtado Dalomba
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Wayne Lencer
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115 .,Harvard Digestive Diseases Center, Boston, MA 02115
| |
Collapse
|
10
|
Retargeting the Clostridium botulinum C2 toxin to the neuronal cytosol. Sci Rep 2016; 6:23707. [PMID: 27025362 PMCID: PMC4812341 DOI: 10.1038/srep23707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/10/2016] [Indexed: 12/16/2022] Open
Abstract
Many biological toxins are known to attack specific cell types, delivering their enzymatic payloads to the cytosol. This process can be manipulated by molecular engineering of chimeric toxins. Using toxins with naturally unlinked components as a starting point is advantageous because it allows for the development of payloads separately from the binding/translocation components. Here the Clostridium botulinum C2 binding/translocation domain was retargeted to neural cell populations by deleting its non-specific binding domain and replacing it with a C. botulinum neurotoxin binding domain. This fusion protein was used to deliver fluorescently labeled payloads to Neuro-2a cells. Intracellular delivery was quantified by flow cytometry and found to be dependent on artificial enrichment of cells with the polysialoganglioside receptor GT1b. Visualization by confocal microscopy showed a dissociation of payloads from the early endosome indicating translocation of the chimeric toxin. The natural Clostridium botulinum C2 toxin was then delivered to human glioblastoma A172 and synchronized HeLa cells. In the presence of the fusion protein, native cytosolic enzymatic activity of the enzyme was observed and found to be GT1b-dependent. This retargeted toxin may enable delivery of therapeutics to peripheral neurons and be of use in addressing experimental questions about neural physiology.
Collapse
|
11
|
Inhibiting oral intoxication of botulinum neurotoxin A complex by carbohydrate receptor mimics. Toxicon 2015; 107:43-9. [PMID: 26272706 DOI: 10.1016/j.toxicon.2015.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
Botulinum neurotoxins (BoNTs) cause the disease botulism manifested by flaccid paralysis that could be fatal to humans and animals. Oral ingestion of the toxin with contaminated food is one of the most common routes for botulism. BoNT assembles with several auxiliary proteins to survive in the gastrointestinal tract and is subsequently transported through the intestinal epithelium into the general circulation. Several hemagglutinin proteins form a multi-protein complex (HA complex) that recognizes host glycans on the intestinal epithelial cell surface to facilitate BoNT absorption. Blocking carbohydrate binding to the HA complex could significantly inhibit the oral toxicity of BoNT. Here, we identify lactulose, a galactose-containing non-digestible sugar commonly used to treat constipation, as a prototype inhibitor against oral BoNT/A intoxication. As revealed by a crystal structure, lactulose binds to the HA complex at the same site where the host galactose-containing carbohydrate receptors bind. In vitro assays using intestinal Caco-2 cells demonstrated that lactulose inhibits HA from compromising the integrity of the epithelial cell monolayers and blocks the internalization of HA. Furthermore, co-administration of lactulose significantly protected mice against BoNT/A oral intoxication in vivo. Taken together, these data encourage the development of carbohydrate receptor mimics as a therapeutic intervention to prevent BoNT oral intoxication.
Collapse
|
12
|
Kim DW, Lee SK, Ahnn J. Botulinum Toxin as a Pain Killer: Players and Actions in Antinociception. Toxins (Basel) 2015; 7:2435-53. [PMID: 26134255 PMCID: PMC4516922 DOI: 10.3390/toxins7072435] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 06/19/2015] [Accepted: 06/23/2015] [Indexed: 11/17/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) have been widely used to treat a variety of clinical ailments associated with pain. The inhibitory action of BoNTs on synaptic vesicle fusion blocks the releases of various pain-modulating neurotransmitters, including glutamate, substance P (SP), and calcitonin gene-related peptide (CGRP), as well as the addition of pain-sensing transmembrane receptors such as transient receptor potential (TRP) to neuronal plasma membrane. In addition, growing evidence suggests that the analgesic and anti-inflammatory effects of BoNTs are mediated through various molecular pathways. Recent studies have revealed that the detailed structural bases of BoNTs interact with their cellular receptors and SNAREs. In this review, we discuss the molecular and cellular mechanisms related to the efficacy of BoNTs in alleviating human pain and insights on engineering the toxins to extend therapeutic interventions related to nociception.
Collapse
Affiliation(s)
- Dong-Wan Kim
- Department of Life Science, School of Natural Science, Hanyang University, Seoul 133-791, Korea.
- BK21 PLUS Life Science for BioDefense Research (BDR) Team, Hanyang University, Seoul 133-791, Korea.
| | - Sun-Kyung Lee
- Department of Life Science, School of Natural Science, Hanyang University, Seoul 133-791, Korea.
- BK21 PLUS Life Science for BioDefense Research (BDR) Team, Hanyang University, Seoul 133-791, Korea.
- The Research Institute for Natural Science, Hanyang University, Seoul 133-791, Korea.
| | - Joohong Ahnn
- Department of Life Science, School of Natural Science, Hanyang University, Seoul 133-791, Korea.
- BK21 PLUS Life Science for BioDefense Research (BDR) Team, Hanyang University, Seoul 133-791, Korea.
- The Research Institute for Natural Science, Hanyang University, Seoul 133-791, Korea.
| |
Collapse
|
13
|
Lam KH, Jin R. Architecture of the botulinum neurotoxin complex: a molecular machine for protection and delivery. Curr Opin Struct Biol 2015; 31:89-95. [PMID: 25889616 DOI: 10.1016/j.sbi.2015.03.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/12/2015] [Accepted: 03/31/2015] [Indexed: 01/22/2023]
Abstract
Botulinum neurotoxins (BoNTs) are extremely poisonous protein toxins that cause the fatal paralytic disease botulism. They are naturally produced in bacteria with several nontoxic neurotoxin-associated proteins (NAPs) and together they form a progenitor toxin complex (PTC), the largest bacterial toxin complex known. In foodborne botulism, the PTC functions as a molecular machine that helps BoNT breach the host defense in the gut. Here, we discuss the substantial recent advance in elucidating the atomic structures and assembly of the 14-subunit PTC, including structures of BoNT and four NAPs. These structural studies shed light on the molecular mechanisms by which BoNT is protected against the acidic environment and proteolytic destruction in the gastrointestinal tract, and how it is delivered across the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
14
|
Botulinum toxin A complex exploits intestinal M cells to enter the host and exert neurotoxicity. Nat Commun 2015; 6:6255. [PMID: 25687350 PMCID: PMC4339894 DOI: 10.1038/ncomms7255] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 01/11/2015] [Indexed: 01/08/2023] Open
Abstract
To cause food-borne botulism, botulinum neurotoxin (BoNT) in the gastrointestinal lumen must traverse the intestinal epithelial barrier. However, the mechanism by which BoNT crosses the intestinal epithelial barrier remains unclear. BoNTs are produced along with one or more non-toxic components, with which they form progenitor toxin complexes (PTCs). Here we show that serotype A1 L-PTC, which has high oral toxicity and makes the predominant contribution to causing illness, breaches the intestinal epithelial barrier from microfold (M) cells via an interaction between haemagglutinin (HA), one of the non-toxic components, and glycoprotein 2 (GP2). HA strongly binds to GP2 expressed on M cells, which do not have thick mucus layers. Susceptibility to orally administered L-PTC is dramatically reduced in M-cell-depleted mice and GP2-deficient (Gp2−/−) mice. Our finding provides the basis for the development of novel antitoxin therapeutics and delivery systems for oral biologics. It is unclear how ingested botulinum neurotoxin invades the host to cause illness. Here, the authors show that the toxin complex containing neurotoxin, hemagglutinin (HA), and NTNHA proteins traverses the epithelial barrier via HA-glycoprotein 2 interaction and endocytosis by Peyer’s patch microfold cells.
Collapse
|
15
|
Lee K, Zhong X, Gu S, Kruel AM, Dorner MB, Perry K, Rummel A, Dong M, Jin R. Molecular basis for disruption of E-cadherin adhesion by botulinum neurotoxin A complex. Science 2014; 344:1405-10. [PMID: 24948737 PMCID: PMC4164303 DOI: 10.1126/science.1253823] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
How botulinum neurotoxins (BoNTs) cross the host intestinal epithelial barrier in foodborne botulism is poorly understood. Here, we present the crystal structure of a clostridial hemagglutinin (HA) complex of serotype BoNT/A bound to the cell adhesion protein E-cadherin at 2.4 angstroms. The HA complex recognizes E-cadherin with high specificity involving extensive intermolecular interactions and also binds to carbohydrates on the cell surface. Binding of the HA complex sequesters E-cadherin in the monomeric state, compromising the E-cadherin-mediated intercellular barrier and facilitating paracellular absorption of BoNT/A. We reconstituted the complete 14-subunit BoNT/A complex using recombinantly produced components and demonstrated that abolishing either E-cadherin- or carbohydrate-binding of the HA complex drastically reduces oral toxicity of BoNT/A complex in vivo. Together, these studies establish the molecular mechanism of how HAs contribute to the oral toxicity of BoNT/A.
Collapse
Affiliation(s)
- Kwangkook Lee
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Xiaofen Zhong
- Department of Microbiology and Immunobiology, Harvard Medical School, Division of Neuroscience, New England Primate Research Center, Southborough, MA 01772, USA
| | - Shenyan Gu
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Anna Magdalena Kruel
- Institut für Toxikologie, Medizinische Hochschule Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Martin B Dorner
- Centre for Biological Threats and Special Pathogens-Biological Toxins (ZBS3), Robert Koch-Institut, Nordufer 20, 13353 Berlin, Germany
| | - Kay Perry
- Northeastern Collaborative Access Team (NE-CAT) and Department of Chemistry and Chemical Biology, Cornell University, Building 436E, Argonne National Laboratory, 9700 S. Cass Avenue, Argonne, IL 60439, USA
| | - Andreas Rummel
- Institut für Toxikologie, Medizinische Hochschule Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Min Dong
- Department of Microbiology and Immunobiology, Harvard Medical School, Division of Neuroscience, New England Primate Research Center, Southborough, MA 01772, USA
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
16
|
Lee K, Lam KH, Kruel AM, Perry K, Rummel A, Jin R. High-resolution crystal structure of HA33 of botulinum neurotoxin type B progenitor toxin complex. Biochem Biophys Res Commun 2014; 446:568-73. [PMID: 24631690 DOI: 10.1016/j.bbrc.2014.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/03/2014] [Indexed: 12/13/2022]
Abstract
Botulinum neurotoxins (BoNTs) are produced as progenitor toxin complexes (PTCs) by Clostridium botulinum. The PTCs are composed of BoNT and non-toxic neurotoxin-associated proteins (NAPs), which serve to protect and deliver BoNT through the gastrointestinal tract in food borne botulism. HA33 is a key NAP component that specifically recognizes host carbohydrates and helps enrich PTC on the intestinal lumen preceding its transport across the epithelial barriers. Here, we report the crystal structure of HA33 of type B PTC (HA33/B) in complex with lactose at 1.46Å resolution. The structural comparisons among HA33 of serotypes A-D reveal two different HA33-glycan interaction modes. The glycan-binding pockets on HA33/A and B are more suitable to recognize galactose-containing glycans in comparison to the equivalent sites on HA33/C and D. On the contrary, HA33/C and D could potentially recognize Neu5Ac as an independent receptor, whereas HA33/A and B do not. These findings indicate that the different oral toxicity and host susceptibility observed among different BoNT serotypes could be partly determined by the serotype-specific interaction between HA33 and host carbohydrate receptors. Furthermore, we have identified a key structural water molecule that mediates the HA33/B-lactose interactions. It provides the structural basis for development of new receptor-mimicking compounds, which have enhanced binding affinity with HA33 through their water-displacing moiety.
Collapse
Affiliation(s)
- Kwangkook Lee
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | | | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, IL, USA
| | - Andreas Rummel
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
17
|
Yao G, Lee K, Gu S, Lam KH, Jin R. Botulinum neurotoxin A complex recognizes host carbohydrates through its hemagglutinin component. Toxins (Basel) 2014; 6:624-35. [PMID: 24525478 PMCID: PMC3942755 DOI: 10.3390/toxins6020624] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 01/15/2014] [Accepted: 02/05/2014] [Indexed: 11/16/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are potent bacterial toxins. The high oral toxicity of BoNTs is largely attributed to the progenitor toxin complex (PTC), which is assembled from BoNT and nontoxic neurotoxin-associated proteins (NAPs) that are produced together with BoNT in bacteria. Here, we performed ex vivo studies to examine binding of the highly homogeneous recombinant NAPs to mouse small intestine. We also carried out the first comprehensive glycan array screening with the hemagglutinin (HA) component of NAPs. Our data confirmed that intestinal binding of the PTC is partly mediated by the HA moiety through multivalent interactions between HA and host carbohydrates. The specific HA-carbohydrate recognition could be inhibited by receptor-mimicking saccharides.
Collapse
Affiliation(s)
- Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| | - Kwangkook Lee
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| | - Shenyan Gu
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
18
|
Lee K, Gu S, Jin L, Le TTN, Cheng LW, Strotmeier J, Kruel AM, Yao G, Perry K, Rummel A, Jin R. Structure of a bimodular botulinum neurotoxin complex provides insights into its oral toxicity. PLoS Pathog 2013; 9:e1003690. [PMID: 24130488 PMCID: PMC3795040 DOI: 10.1371/journal.ppat.1003690] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 07/11/2013] [Indexed: 11/24/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are produced by Clostridium botulinum and cause the fatal disease botulism, a flaccid paralysis of the muscle. BoNTs are released together with several auxiliary proteins as progenitor toxin complexes (PTCs) to become highly potent oral poisons. Here, we report the structure of a ∼760 kDa 14-subunit large PTC of serotype A (L-PTC/A) and reveal insight into its absorption mechanism. Using a combination of X-ray crystallography, electron microscopy, and functional studies, we found that L-PTC/A consists of two structurally and functionally independent sub-complexes. A hetero-dimeric 290 kDa complex protects BoNT, while a hetero-dodecameric 470 kDa complex facilitates its absorption in the harsh environment of the gastrointestinal tract. BoNT absorption is mediated by nine glycan-binding sites on the dodecameric sub-complex that forms multivalent interactions with carbohydrate receptors on intestinal epithelial cells. We identified monosaccharides that blocked oral BoNT intoxication in mice, which suggests a new strategy for the development of preventive countermeasures for BoNTs based on carbohydrate receptor mimicry. Food-borne botulinum neurotoxin (BoNT) poisoning results in fatal muscle paralysis. But how can BoNT–a large protein released by the bacteria clostridia–survive the hostile gastrointestinal (GI) tract to gain access to neurons that control muscle contraction? Here, we report the complete structure of a bimodular ∼760 kDa BoNT/A large progenitor toxin complex (L-PTC), which is composed of BoNT and four non-toxic bacterial proteins. The architecture of this bacterial machinery mimics an Apollo lunar module, whereby the “ascent stage” (a ∼290 kDa module) protects BoNT from destruction in the GI tract and the 3-arm “descent stage” (a ∼470 kDa module) mediates absorption of BoNT by binding to host carbohydrate receptors in the small intestine. This new finding has helped us identify the carbohydrate-binding sites and the monosaccharide IPTG as a prototypical oral inhibitor, which extends survival following lethal BoNT/A intoxication of mice. Hence, pre-treatment with small molecule inhibitors based on carbohydrate receptor mimicry can provide temporary protection against BoNT entry into the circulation.
Collapse
Affiliation(s)
- Kwangkook Lee
- Department of Physiology and Biophysics, University of California, Irvine, California, United States of America
| | - Shenyan Gu
- Department of Physiology and Biophysics, University of California, Irvine, California, United States of America
| | - Lei Jin
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Thi Tuc Nghi Le
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Luisa W. Cheng
- Foodborne Contaminants Research Unit, Western Regional Research Center, United States Department of Agriculture, Agricultural Research Service, Albany, California, United States of America
| | - Jasmin Strotmeier
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | | - Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, California, United States of America
| | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, Illinois, United States of America
| | - Andreas Rummel
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
- * E-mail: (AR); (RJ)
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, California, United States of America
- Neuroscience, Aging and Stem Cell Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- * E-mail: (AR); (RJ)
| |
Collapse
|
19
|
Miyashita SI, Sagane Y, Inui K, Hayashi S, Miyata K, Suzuki T, Ohyama T, Watanabe T, Niwa K. Botulinum toxin complex increases paracellular permeability in intestinal epithelial cells via activation of p38 mitogen-activated protein kinase. J Vet Med Sci 2013; 75:1637-42. [PMID: 23884081 PMCID: PMC3942962 DOI: 10.1292/jvms.13-0164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Clostridium botulinum produces a large toxin complex (L-TC) that increases paracellular
permeability in intestinal epithelial cells by a mechanism that remains unclear. Here, we
show that mitogen-activated protein kinases (MAPKs) are involved in this permeability
increase. Paracellular permeability was measured by FITC-dextran flux through a monolayer
of rat intestinal epithelial IEC-6 cells, and MAPK activation was estimated from western
blots. L-TC of C. botulinum serotype D strain 4947 increased paracellular
dextran flux and activated extracellular signal-regulated kinase (ERK), p38, but not c-Jun
N-terminal kinase (JNK) in IEC-6 cells. The permeability increase induced by L-TC was
abrogated by the p38 inhibitor SB203580. These results indicate that L-TC increases
paracellular permeability by activating p38, but not JNK and ERK.
Collapse
Affiliation(s)
- Shin-Ichiro Miyashita
- Department of Food and Cosmetic Science, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido 099-2493, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Botulinum neurotoxin (BoNT) is produced by Clostridium botulinum and associates with nontoxic neurotoxin-associated proteins to form high-molecular weight progenitor complexes (PCs). The PCs are required for the oral toxicity of BoNT in the context of food-borne botulism and are thought to protect BoNT from destruction in the gastrointestinal tract and aid in absorption from the gut lumen. The PC can differ in size and protein content depending on the C. botulinum strain. The oral toxicity of the BoNT PC increases as the size of the PC increases, but the molecular architecture of these large complexes and how they contribute to BoNT toxicity have not been elucidated. We have generated 2D images of PCs from strains producing BoNT serotypes A1, B, and E using negative stain electron microscopy and single-particle averaging. The BoNT/A1 and BoNT/B PCs were observed as ovoid-shaped bodies with three appendages, whereas the BoNT/E PC was observed as an ovoid body. Both the BoNT/A1 and BoNT/B PCs showed significant flexibility, and the BoNT/B PC was documented as a heterogeneous population of assembly/disassembly intermediates. We have also determined 3D structures for each serotype using the random conical tilt approach. Crystal structures of the individual proteins were placed into the BoNT/A1 and BoNT/B PC electron density maps to generate unique detailed models of the BoNT PCs. The structures highlight an effective platform that can be engineered for the development of mucosal vaccines and the intestinal absorption of oral biologics.
Collapse
|
21
|
Couesnon A, Molgó J, Connan C, Popoff MR. Preferential entry of botulinum neurotoxin A Hc domain through intestinal crypt cells and targeting to cholinergic neurons of the mouse intestine. PLoS Pathog 2012; 8:e1002583. [PMID: 22438808 PMCID: PMC3305446 DOI: 10.1371/journal.ppat.1002583] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 01/27/2012] [Indexed: 12/12/2022] Open
Abstract
Botulism, characterized by flaccid paralysis, commonly results from botulinum neurotoxin (BoNT) absorption across the epithelial barrier from the digestive tract and then dissemination through the blood circulation to target autonomic and motor nerve terminals. The trafficking pathway of BoNT/A passage through the intestinal barrier is not yet fully understood. We report that intralumenal administration of purified BoNT/A into mouse ileum segment impaired spontaneous muscle contractions and abolished the smooth muscle contractions evoked by electric field stimulation. Entry of BoNT/A into the mouse upper small intestine was monitored with fluorescent HcA (half C-terminal domain of heavy chain) which interacts with cell surface receptor(s). We show that HcA preferentially recognizes a subset of neuroendocrine intestinal crypt cells, which probably represent the entry site of the toxin through the intestinal barrier, then targets specific neurons in the submucosa and later (90–120 min) in the musculosa. HcA mainly binds to certain cholinergic neurons of both submucosal and myenteric plexuses, but also recognizes, although to a lower extent, other neuronal cells including glutamatergic and serotoninergic neurons in the submucosa. Intestinal cholinergic neuron targeting by HcA could account for the inhibition of intestinal peristaltism and secretion observed in botulism, but the consequences of the targeting to non-cholinergic neurons remains to be determined. Botulism is a severe and often fatal disease in man and animals characterized by flaccid paralysis. Clostridium botulinum produces a potent neurotoxin (botulinum neurotoxin) responsible for all the symptoms of botulism. Botulism is most often acquired by ingesting preformed botulinum neurotoxin in contaminated food or after intestinal colonization by C. botulinum under certain circumstances, such as in infant botulism, and toxin production in the intestine. The first step of the disease consists in the passage of the botulinum neurotoxin through the intestinal barrier, which is still poorly understood. We investigated the trafficking of the botulinum neurotoxin in a mouse intestinal loop model, using fluorescent HcA (half C-terminal domain of the heavy chain). We observed that HcA preferentially recognizes neuroendocrine intestinal crypt cells, which likely represent the entry site of the toxin through the intestinal barrier, then targets specific neurons, mainly cholinergic neurons, in the submucosa, and later (90–120 min) in the musculosa leading to local paralytic effects such as inhibition of intestinal peristaltism. These results represent an important advance in the understanding of the initial steps of botulism intoxication and can be the basis for the development of new specific countermeasures against botulism.
Collapse
Affiliation(s)
- Aurélie Couesnon
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, Paris, France
| | - Jordi Molgó
- CNRS, Institut de Neurobiologie Alfred Fessard – FRC2118, Laboratoire de Neurobiologie– et Développement UPR3294, Gif sur Yvette, France
| | - Chloé Connan
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, Paris, France
| | - Michel R. Popoff
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, Paris, France
- * E-mail:
| |
Collapse
|
22
|
Rapid immune responses to a botulinum neurotoxin Hc subunit vaccine through in vivo targeting to antigen-presenting cells. Infect Immun 2011; 79:3388-96. [PMID: 21576339 DOI: 10.1128/iai.00166-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The clostridial botulinum neurotoxins (BoNTs) are the most potent protein toxins known. The carboxyl-terminal fragment of the toxin heavy chain (Hc) has been intensively investigated as a BoNT vaccine immunogen. We sought to determine whether targeting Hc to antigen-presenting cells (APCs) could accelerate the immune responses to vaccination with BoNT serotype A (BoNT/A) Hc. To test this hypothesis, we targeted Hc to the Fc receptors for IgG (FcγRs) expressed by dendritic cells (DCs) and other APCs. Hc was expressed as a fusion protein with a recombinant ligand for human FcγRs (R4) to produce HcR4 or a similar ligand for murine FcγRs to produce HcmR4. HcR4, HcmR4, and Hc were produced as secreted proteins using baculovirus-mediated expression in SF9 insect cells. In vitro receptor binding assays showed that HcR4 effectively targets Hc to all classes of FcγRs. APCs loaded with HcR4 or HcmR4 are substantially more effective at stimulating Hc-reactive T cells than APCs loaded with nontargeted Hc. Mice immunized with a single dose of HcmR4 or HcR4 had earlier and markedly higher Hc-reactive antibody titers than mice immunized with nontargeted Hc. These results extend to BoNT neutralizing antibody titers, which are substantially higher in mice immunized with HcmR4 than in mice immunized with Hc. Our results demonstrate that targeting Hc to FcγRs augments the pace and magnitude of immune responses to Hc.
Collapse
|
23
|
Sugawara Y, Fujinaga Y. The botulinum toxin complex meets E-cadherin on the way to its destination. Cell Adh Migr 2011; 5:34-6. [PMID: 20935473 DOI: 10.4161/cam.5.1.13574] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Botulinum neurotoxin (BoNT) causes the disease botulism, which is characterized by flaccid paralysis, in humans and animals. The metalloprotease activity of BoNT inhibits neurotransmitter release at neuro-muscular junctions. In most cases, poisoning occurs when BoNT is ingested. Therefore, BoNT must pass through the epithelial barrier of the gastrointestinal tract to enter the systemic circulation and reach the target site. BoNT forms large protein complexes by associating with non-toxic components referred to as non-toxic non-hemagglutinin (NTNH) and hemagglutinin (HA). These proteins protect BoNT from the low pH and proteases in the digestive tract. We recently determined that HA has an unexpected function of disrupting the intercellular epithelial barrier by directly binding to E-cadherin. HA binds to E-cadherin and disrupts its function in a species-specific manner, and this interaction is essential to disrupt tight junctions. This activity is thought to facilitate the absorption of BoNT through the paracellular route of the intestinal epithelium in susceptible species.
Collapse
Affiliation(s)
- Yo Sugawara
- International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | | |
Collapse
|
24
|
Sugawara Y, Matsumura T, Takegahara Y, Jin Y, Tsukasaki Y, Takeichi M, Fujinaga Y. Botulinum hemagglutinin disrupts the intercellular epithelial barrier by directly binding E-cadherin. ACTA ACUST UNITED AC 2010; 189:691-700. [PMID: 20457762 PMCID: PMC2872904 DOI: 10.1083/jcb.200910119] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Botulinum neurotoxin is produced by Clostridium botulinum and forms large protein complexes through associations with nontoxic components. We recently found that hemagglutinin (HA), one of the nontoxic components, disrupts the intercellular epithelial barrier; however, the mechanism underlying this phenomenon is not known. In this study, we identified epithelial cadherin (E-cadherin) as a target molecule for HA. HA directly binds E-cadherin and disrupts E-cadherin-mediated cell to cell adhesion. Although HA binds human, bovine, and mouse E-cadherin, it does not bind rat or chicken E-cadherin homologues. HA does not interact with other members of the classical cadherin family such as neural and vascular endothelial cadherin. Expression of rat E-cadherin but not mouse rescues Madin-Darby canine kidney cells from HA-induced tight junction (TJ) disruptions. These data demonstrate that botulinum HA directly binds E-cadherin and disrupts E-cadherin-mediated cell to cell adhesion in a species-specific manner and that the HA-E-cadherin interaction is essential for the disruption of TJ function.
Collapse
Affiliation(s)
- Yo Sugawara
- Laboratory for Infection Cell Biology, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Interaction of botulinum toxin with the epithelial barrier. J Biomed Biotechnol 2010; 2010:974943. [PMID: 20169001 PMCID: PMC2822237 DOI: 10.1155/2010/974943] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 12/24/2009] [Indexed: 11/17/2022] Open
Abstract
Botulinum neurotoxin (BoNT) is a protein toxin (approximately 150 kDa), which possesses a metalloprotease activity. Food-borne botulism is manifested when BoNT is absorbed from the digestive tract to the blood stream and enters the peripheral nerves, where the toxin cleaves core proteins of the neuroexocytosis apparatus and elicits the inhibition of neurotransmitter release. The initial obstacle to orally ingested BoNT entering the body is the epithelial barrier of the digestive tract. Recent cell biology and molecular biology studies are beginning to elucidate the mechanism by which this large protein toxin crosses the epithelial barrier. In this review, we provide an overview of the structural features of botulinum toxins (BoNT and BoNT complex) and the interaction of these toxins with the epithelial barrier.
Collapse
|
27
|
Abstract
Bacterial toxins damage the host at the site of bacterial infection or distant from the site. Bacterial toxins can be single proteins or oligomeric protein complexes that are organized with distinct AB structure-function properties. The A domain encodes a catalytic activity. ADP ribosylation of host proteins is the earliest post-translational modification determined to be performed by bacterial toxins; other modifications include glucosylation and proteolysis. Bacterial toxins also catalyze the non-covalent modification of host protein function or can modify host cell properties through direct protein-protein interactions. The B domain includes two functional domains: a receptor-binding domain, which defines the tropism of a toxin for a cell and a translocation domain that delivers the A domain across a lipid bilayer, either on the plasma membrane or the endosome. Bacterial toxins are often characterized based upon the secretion mechanism that delivers the toxin out of the bacterium, termed types I-VII. This review summarizes the major families of bacterial toxins and also describes the specific structure-function properties of the botulinum neurotoxins.
Collapse
Affiliation(s)
- James S Henkel
- Medical College of Wisconsin, Department of Microbiology and Molecular Genetics, Milwaukee, WI 53151, USA.
| | | | | |
Collapse
|
28
|
Stenmark P, Dupuy J, Imamura A, Kiso M, Stevens RC. Crystal structure of botulinum neurotoxin type A in complex with the cell surface co-receptor GT1b-insight into the toxin-neuron interaction. PLoS Pathog 2008; 4:e1000129. [PMID: 18704164 PMCID: PMC2493045 DOI: 10.1371/journal.ppat.1000129] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 07/18/2008] [Indexed: 11/18/2022] Open
Abstract
Botulinum neurotoxins have a very high affinity and specificity for their target cells requiring two different co-receptors located on the neuronal cell surface. Different toxin serotypes have different protein receptors; yet, most share a common ganglioside co-receptor, GT1b. We determined the crystal structure of the botulinum neurotoxin serotype A binding domain (residues 873–1297) alone and in complex with a GT1b analog at 1.7 Å and 1.6 Å, respectively. The ganglioside GT1b forms several key hydrogen bonds to conserved residues and binds in a shallow groove lined by Tryptophan 1266. GT1b binding does not induce any large structural changes in the toxin; therefore, it is unlikely that allosteric effects play a major role in the dual receptor recognition. Together with the previously published structures of botulinum neurotoxin serotype B in complex with its protein co-receptor, we can now generate a detailed model of botulinum neurotoxin's interaction with the neuronal cell surface. The two branches of the GT1b polysaccharide, together with the protein receptor site, impose strict geometric constraints on the mode of interaction with the membrane surface and strongly support a model where one end of the 100 Å long translocation domain helix bundle swing into contact with the membrane, initiating the membrane anchoring event. Botulinum neurotoxins are the most toxic substances known and are classified as a category A bioterrorism agent. Ongoing work on the development of countermeasures for the neurotoxin has been limited by an incomplete understanding of the means by which the toxin enters the cell. Our study provides a detailed look at how the toxin binds its ganglioside co-receptor on the cell surface. Together with earlier work this generates a detailed description of how the toxin binds its two co-receptors to position it for entrance into the neuronal cell. This structural data provides critical new insight about the action of the botulinum neurotoxins that can be applied toward the development of agents to block toxin uptake in the digestive system and/or inhibit the binding of the toxin at the neuromuscular junction.
Collapse
Affiliation(s)
- Pål Stenmark
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jérôme Dupuy
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Akihiro Imamura
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida, Sakyo-ku, Kyoto, Japan
| | - Makoto Kiso
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida, Sakyo-ku, Kyoto, Japan
| | - Raymond C. Stevens
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|