1
|
Báez BB, Bacaglio CR, Prendergast JM, Rozés-Salvador V, Sheikh KA, Bianchet M, Farah MH, Schnaar RL, Bisbal M, Lopez PHH. Tumor necrosis factor α receptor 1A transduces the inhibitory effect on axon regeneration triggered by IgG anti-ganglioside GD1a antibodies. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167315. [PMID: 38897255 DOI: 10.1016/j.bbadis.2024.167315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Anti-ganglioside antibodies (anti-Gg Abs) have been linked to delayed/poor clinical recovery in both axonal and demyelinating forms of Guillain-Barrè Syndrome (GBS). In many instances, the incomplete recovery is attributed to the peripheral nervous system's failure to regenerate. The cross-linking of cell surface gangliosides by anti-Gg Abs triggers inhibition of nerve repair in both in vitro and in vivo axon regeneration paradigms. This mechanism involves the activation of the small GTPase RhoA, which negatively modulates the growth cone cytoskeleton. At present, the identity/es of the receptor/s responsible for transducing the signal that ultimately leads to RhoA activation remains poorly understood. The aim of this work was to identify the transducer molecule responsible for the inhibitory effect of anti-Gg Abs on nerve repair. Putative candidate molecules were identified through proteomic mass spectrometry of ganglioside affinity-captured proteins from rat cerebellar granule neurons (Prendergast et al., 2014). These candidates were evaluated using an in vitro model of neurite outgrowth with primary cultured dorsal root ganglion neurons (DRGn) and an in vivo model of axon regeneration. Using an shRNA-strategy to silence putative candidates on DRGn, we identified tumor necrosis factor receptor 1A protein (TNFR1A) as a transducer molecule for the inhibitory effect on neurite outgrowth from rat/mouse DRGn cultures of a well characterized mAb targeting the related gangliosides GD1a and GT1b. Interestingly, lack of TNFr1A expression on DRGn abolished the inhibitory effect on neurite outgrowth caused by anti-GD1a but not anti-GT1b specific mAbs, suggesting specificity of GD1a/transducer signaling. Similar results were obtained using primary DRGn cultures from TNFR1a-null mice, which did not activate RhoA after exposure to anti-GD1a mAbs. Generation of single point mutants at the stalk region of TNFR1A identified a critical amino acid for transducing GD1a signaling, suggesting a direct interaction. Finally, passive immunization with an anti-GD1a/GT1b mAb in an in vivo model of axon regeneration exhibited reduced inhibitory activity in TNFR1a-null mice compared to wild type mice. In conclusion, these findings identify TNFR1A as a novel transducer receptor for the inhibitory effect exerted by anti-GD1a Abs on nerve repair, representing a significant step forward toward understanding the factors contributing to poor clinical recovery in GBS associated with anti-Gg Abs.
Collapse
Affiliation(s)
- Bárbara B Báez
- Departamento de Química Biológica "Dr Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CIQUIBIC-CONICET-UNC, Argentina; Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Argentina
| | - Cristian R Bacaglio
- Departamento de Química Biológica "Dr Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CIQUIBIC-CONICET-UNC, Argentina; Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Argentina
| | - Jillian M Prendergast
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins Medicine, Baltimore, United States
| | - Victoria Rozés-Salvador
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Argentina
| | - Kazim A Sheikh
- Department of Neurology, University of Texas Medical School at Houston, Houston, United States
| | - Mario Bianchet
- Department of Biophysics & Biophysical Chemistry, School of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Mohamed H Farah
- Department of Neurology and Neuroscience, School of Medicine, Johns Hopkins Medicine, Baltimore, United States
| | - Ronald L Schnaar
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins Medicine, Baltimore, United States
| | - Mariano Bisbal
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Argentina
| | - Pablo H H Lopez
- Departamento de Química Biológica "Dr Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CIQUIBIC-CONICET-UNC, Argentina; Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
2
|
Xiong X, Qiu Y, Zheng J, Zhou L, Wang Q, Pang J, Zhang W, Chen H, Liu G, Han X. Generation and characterization of a monoclonal antibody against FGFR3 that protects mice from BoNT/A. Protein Expr Purif 2024; 213:106370. [PMID: 37709211 DOI: 10.1016/j.pep.2023.106370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/14/2023] [Accepted: 09/09/2023] [Indexed: 09/16/2023]
Abstract
Botulinum neurotoxin serotype A (BoNT/A) can cause flaccid paralysis of muscles, an illness fatal to human, by entering neurons and blocking neurotransmitter release. The process was mediated by three receptors. A specific monoclonal antibody anti-D23, designated as ML419, targeting the ectodomain (D23) of fibroblast growth factor receptor 3 (FGFR3), one of the three receptors, was screened and capable of disturbing the recognition of BoNT/A and FGFR3. ML419 was screened from 14 stable positive hybridoma cell lines, and was subcloned, sequenced, and classified as IgG2a(κ) subclass. ML419 binds the D23 domain of FGFR3 with high affinity (KD∼0.26 nM), and prevents the BoNT/A from entering Neuro-2a cells effectively. In vivo data showed that, 200 μg of ML419 could completely protect all the mice against with 5 MLD50 BoNT/A, while 100 μg of ML419 could protected 60% of the mice. Collectively, our results indicated that ML419 served as a good candidate for further development of therapeutics for BoNT/A.
Collapse
Affiliation(s)
| | - Yujin Qiu
- Academy of Military Medical Sciences, Beijing, PR China
| | - Jiahao Zheng
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Ling Zhou
- Department of Clinical Laboratory, The Affiliated Pudong Hospital, Fudan University, Shanghai, China
| | - Qingyang Wang
- Academy of Military Medical Sciences, Beijing, PR China
| | - Jinglun Pang
- College of Life Sciences, Inner Mongolia Agriculture University, Hohhot, Inner Mongolia, PR China
| | - Weicai Zhang
- Academy of Military Medical Sciences, Beijing, PR China
| | - Huipeng Chen
- Academy of Military Medical Sciences, Beijing, PR China.
| | - Gang Liu
- Academy of Military Medical Sciences, Beijing, PR China.
| | - Xiaodong Han
- College of Life Sciences, Inner Mongolia Agriculture University, Hohhot, Inner Mongolia, PR China.
| |
Collapse
|
3
|
Joensuu M, Syed P, Saber SH, Lanoue V, Wallis TP, Rae J, Blum A, Gormal RS, Small C, Sanders S, Jiang A, Mahrhold S, Krez N, Cousin MA, Cooper‐White R, Cooper‐White JJ, Collins BM, Parton RG, Balistreri G, Rummel A, Meunier FA. Presynaptic targeting of botulinum neurotoxin type A requires a tripartite PSG-Syt1-SV2 plasma membrane nanocluster for synaptic vesicle entry. EMBO J 2023; 42:e112095. [PMID: 37226896 PMCID: PMC10308369 DOI: 10.15252/embj.2022112095] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 04/18/2023] [Accepted: 04/28/2023] [Indexed: 05/26/2023] Open
Abstract
The unique nerve terminal targeting of botulinum neurotoxin type A (BoNT/A) is due to its capacity to bind two receptors on the neuronal plasma membrane: polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Whether and how PSGs and SV2 may coordinate other proteins for BoNT/A recruitment and internalization remains unknown. Here, we demonstrate that the targeted endocytosis of BoNT/A into synaptic vesicles (SVs) requires a tripartite surface nanocluster. Live-cell super-resolution imaging and electron microscopy of catalytically inactivated BoNT/A wildtype and receptor-binding-deficient mutants in cultured hippocampal neurons demonstrated that BoNT/A must bind coincidentally to a PSG and SV2 to target synaptic vesicles. We reveal that BoNT/A simultaneously interacts with a preassembled PSG-synaptotagmin-1 (Syt1) complex and SV2 on the neuronal plasma membrane, facilitating Syt1-SV2 nanoclustering that controls endocytic sorting of the toxin into synaptic vesicles. Syt1 CRISPRi knockdown suppressed BoNT/A- and BoNT/E-induced neurointoxication as quantified by SNAP-25 cleavage, suggesting that this tripartite nanocluster may be a unifying entry point for selected botulinum neurotoxins that hijack this for synaptic vesicle targeting.
Collapse
Affiliation(s)
- Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
| | - Parnayan Syed
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Saber H Saber
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - James Rae
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Ailisa Blum
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Christopher Small
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Shanley Sanders
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Anmin Jiang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Stefan Mahrhold
- Institut für ToxikologieMedizinische Hochschule HannoverHannoverGermany
| | - Nadja Krez
- Institut für ToxikologieMedizinische Hochschule HannoverHannoverGermany
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, Hugh Robson BuildingUniversity of EdinburghEdinburghUK
- Muir Maxwell Epilepsy CentreUniversity of EdinburghEdinburghUK
- Simons Initiative for the Developing BrainUniversity of EdinburghEdinburghUK
| | - Ruby Cooper‐White
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
- School of Chemical EngineeringThe University of QueenslandBrisbaneQLDAustralia
| | - Justin J Cooper‐White
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
- School of Chemical EngineeringThe University of QueenslandBrisbaneQLDAustralia
- UQ Centre for Stem Cell Ageing and Regenerative EngineeringThe University of QueenslandBrisbaneQLDAustralia
| | - Brett M Collins
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Robert G Parton
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
- Centre for Microscopy and MicroanalysisThe University of QueenslandBrisbaneQLDAustralia
| | - Giuseppe Balistreri
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Department of Virology, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Andreas Rummel
- Institut für ToxikologieMedizinische Hochschule HannoverHannoverGermany
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
4
|
Liu Z, Lee PG, Krez N, Lam KH, Liu H, Przykopanski A, Chen P, Yao G, Zhang S, Tremblay JM, Perry K, Shoemaker CB, Rummel A, Dong M, Jin R. Structural basis for botulinum neurotoxin E recognition of synaptic vesicle protein 2. Nat Commun 2023; 14:2338. [PMID: 37095076 PMCID: PMC10125960 DOI: 10.1038/s41467-023-37860-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Botulinum neurotoxin E (BoNT/E) is one of the major causes of human botulism and paradoxically also a promising therapeutic agent. Here we determined the co-crystal structures of the receptor-binding domain of BoNT/E (HCE) in complex with its neuronal receptor synaptic vesicle glycoprotein 2A (SV2A) and a nanobody that serves as a ganglioside surrogate. These structures reveal that the protein-protein interactions between HCE and SV2 provide the crucial location and specificity information for HCE to recognize SV2A and SV2B, but not the closely related SV2C. At the same time, HCE exploits a separated sialic acid-binding pocket to mediate recognition of an N-glycan of SV2. Structure-based mutagenesis and functional studies demonstrate that both the protein-protein and protein-glycan associations are essential for SV2A-mediated cell entry of BoNT/E and for its potent neurotoxicity. Our studies establish the structural basis to understand the receptor-specificity of BoNT/E and to engineer BoNT/E variants for new clinical applications.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Pyung-Gang Lee
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Nadja Krez
- Institute of Toxicology, Hannover Medical School, Hannover, 30623, Germany
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Hao Liu
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Adina Przykopanski
- Institute of Toxicology, Hannover Medical School, Hannover, 30623, Germany
| | - Peng Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Sicai Zhang
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, IL, 60439, USA
| | | | - Andreas Rummel
- Institute of Toxicology, Hannover Medical School, Hannover, 30623, Germany
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
5
|
Cottone G, Chiodo L, Maragliano L, Popoff MR, Rasetti-Escargueil C, Lemichez E, Malliavin TE. In Silico Conformational Features of Botulinum Toxins A1 and E1 According to Intraluminal Acidification. Toxins (Basel) 2022; 14:toxins14090644. [PMID: 36136581 PMCID: PMC9500700 DOI: 10.3390/toxins14090644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/13/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Although botulinum neurotoxins (BoNTs) are among the most toxic compounds found in nature, their molecular mechanism of action is far from being elucidated. A key event is the conformational transition due to acidification of the interior of synaptic vesicles, leading to translocation of the BoNT catalytic domain into the neuronal cytosol. To investigate these conformational variations, homology modeling and atomistic simulations are combined to explore the internal dynamics of the sub-types BoNT/A1 (the most-used sub-type in medical applications) and BoNT/E1 (the most kinetically efficient sub-type). This first simulation study of di-chain BoNTs in closed and open states considers the effects of both neutral and acidic pH. The conformational mobility is driven by domain displacements of the ganglioside-binding site in the receptor binding domain, the translocation domain (HCNT) switch, and the belt α-helix, which present multiple conformations, depending on the primary sequence and the pH. Fluctuations of the belt α-helix are observed for closed conformations of the toxins and at acidic pH, while patches of more solvent-accessible residues appear under the same conditions in the core translocation domain HCNT. These findings suggest that, during translocation, the higher mobility of the belt could be transmitted to HCNT, leading to the favorable interaction of HCNT residues with the non-polar membrane environment.
Collapse
Affiliation(s)
- Grazia Cottone
- Department of Physics and Chemistry Emilio Segré, University of Palermo, Viale delle Scienze, 90128 Palermo, Italy
| | - Letizia Chiodo
- Department of Engineering, University Campus Bio-Medico of Rome, Via Á. del Portillo 21, 00128 Rome, Italy
| | - Luca Maragliano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Michel-Robert Popoff
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Inserm U1306, Unité des Toxines Bactériennes, 75015 Paris, France
| | - Christine Rasetti-Escargueil
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Inserm U1306, Unité des Toxines Bactériennes, 75015 Paris, France
| | - Emmanuel Lemichez
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Inserm U1306, Unité des Toxines Bactériennes, 75015 Paris, France
- Correspondence: (E.L.); (T.E.M.)
| | - Thérèse E. Malliavin
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Unité de Bioinformatique Structurale, 75015 Paris, France
- Laboratoire de Physique et Chimie Théoriques (LPCT), CNRS UMR7019, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France
- Laboratoire International Associé, CNRS and University of Illinois at Urbana-Champaign, 54506 Vandoeuvre-lès-Nancy, France
- Correspondence: (E.L.); (T.E.M.)
| |
Collapse
|
6
|
Crystal Structures of the Clostridium botulinum Neurotoxin A6 Cell Binding Domain Alone and in Complex with GD1a Reveal Significant Conformational Flexibility. Int J Mol Sci 2022; 23:ijms23179620. [PMID: 36077016 PMCID: PMC9456117 DOI: 10.3390/ijms23179620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/26/2022] Open
Abstract
Clostridium botulinum neurotoxin A (BoNT/A) targets the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, by cleaving synaptosomal-associated protein of 25 kDa size (SNAP-25). Cleavage of SNAP-25 results in flaccid paralysis due to repression of synaptic transmission at the neuromuscular junction. This activity has been exploited to treat a range of diseases associated with hypersecretion of neurotransmitters, with formulations of BoNT/A commercially available as therapeutics. Generally, BoNT activity is facilitated by three essential domains within the molecule, the cell binding domain (HC), the translocation domain (HN), and the catalytic domain (LC). The HC, which consists of an N-terminal (HCN) and a C-terminal (HCC) subdomain, is responsible for BoNT’s high target specificity where it forms a dual-receptor complex with synaptic vesicle protein 2 (SV2) and a ganglioside receptor on the surface of motor neurons. In this study, we have determined the crystal structure of botulinum neurotoxin A6 cell binding domain (HC/A6) in complex with GD1a and describe the interactions involved in ganglioside binding. We also present a new crystal form of wild type HC/A6 (crystal form II) where a large ‘hinge motion’ between the HCN and HCC subdomains is observed. These structures, along with a comparison to the previously determined wild type crystal structure of HC/A6 (crystal form I), reveals the degree of conformational flexibility exhibited by HC/A6.
Collapse
|
7
|
Gregory KS, Mojanaga OO, Liu SM, Acharya KR. Crystal Structures of Botulinum Neurotoxin Subtypes A4 and A5 Cell Binding Domains in Complex with Receptor Ganglioside. Toxins (Basel) 2022; 14:toxins14020129. [PMID: 35202156 PMCID: PMC8876736 DOI: 10.3390/toxins14020129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
Botulinum neurotoxins (BoNT) cause the potentially fatal neuroparalytic disease botulism that arises due to proteolysis of a SNARE protein. Each BoNT is comprised of three domains: a cell binding domain (HC), a translocation domain (HN), and a catalytic (Zn2+ endopeptidase) domain (LC). The HC is responsible for neuronal specificity by targeting both a protein and ganglioside receptor at the neuromuscular junction. Although highly toxic, some BoNTs are commercially available as therapeutics for the treatment of a range of neuromuscular conditions. Here we present the crystal structures of two BoNT cell binding domains, HC/A4 and HC/A5, in a complex with the oligosaccharide of ganglioside, GD1a and GM1b, respectively. These structures, along with a detailed comparison with the previously reported apo-structures, reveal the conformational changes that occur upon ganglioside binding and the interactions involved.
Collapse
Affiliation(s)
- Kyle S. Gregory
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK; (K.S.G.); (O.O.M.)
| | - Otsile O. Mojanaga
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK; (K.S.G.); (O.O.M.)
| | - Sai Man Liu
- Protein Sciences Department, Ipsen Bioinnovation Limited, 102 Park Drive, Milton Park, Abingdon OX14 4RY, UK;
| | - K. Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK; (K.S.G.); (O.O.M.)
- Correspondence: ; Tel.: +44-(0)1225-386238
| |
Collapse
|
8
|
Structural Analysis of Botulinum Neurotoxins Type B and E by Cryo-EM. Toxins (Basel) 2021; 14:toxins14010014. [PMID: 35050991 PMCID: PMC8781748 DOI: 10.3390/toxins14010014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are the causative agents of a potentially lethal paralytic disease targeting cholinergic nerve terminals. Multiple BoNT serotypes exist, with types A, B and E being the main cause of human botulism. Their extreme toxicity has been exploited for cosmetic and therapeutic uses to treat a wide range of neuromuscular disorders. Although naturally occurring BoNT types share a common end effect, their activity varies significantly based on the neuronal cell-surface receptors and intracellular SNARE substrates they target. These properties are the result of structural variations that have traditionally been studied using biophysical methods such as X-ray crystallography. Here, we determined the first structures of botulinum neurotoxins using single-particle cryogenic electron microscopy. The maps obtained at 3.6 and 3.7 Å for BoNT/B and /E, respectively, highlight the subtle structural dynamism between domains, and of the binding domain in particular. This study demonstrates how the recent advances made in the field of single-particle electron microscopy can be applied to bacterial toxins of clinical relevance and the botulinum neurotoxin family in particular.
Collapse
|
9
|
Mechanism of Ganglioside Receptor Recognition by Botulinum Neurotoxin Serotype E. Int J Mol Sci 2021; 22:ijms22158315. [PMID: 34361086 PMCID: PMC8346984 DOI: 10.3390/ijms22158315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
The botulinum neurotoxins are potent molecules that are not only responsible for the lethal paralytic disease botulism, but have also been harnessed for therapeutic uses in the treatment of an increasing number of chronic neurological and neuromuscular disorders, in addition to cosmetic applications. The toxins act at the cholinergic nerve terminals thanks to an efficient and specific mechanism of cell recognition which is based on a dual receptor system that involves gangliosides and protein receptors. Binding to surface-anchored gangliosides is the first essential step in this process. Here, we determined the X-ray crystal structure of the binding domain of BoNT/E, a toxin of clinical interest, in complex with its GD1a oligosaccharide receptor. Beyond confirmation of the conserved ganglioside binding site, we identified key interacting residues that are unique to BoNT/E and a significant rearrangement of loop 1228–1237 upon carbohydrate binding. These observations were also supported by thermodynamic measurements of the binding reaction and assessment of ganglioside selectivity by immobilised-receptor binding assays. These results provide a structural basis to understand the specificity of BoNT/E for complex gangliosides.
Collapse
|
10
|
Brier S, Rasetti-Escargueil C, Wijkhuisen A, Simon S, Marechal M, Lemichez E, Popoff MR. Characterization of a highly neutralizing single monoclonal antibody to botulinum neurotoxin type A. FASEB J 2021; 35:e21540. [PMID: 33817838 DOI: 10.1096/fj.202002492r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 01/15/2023]
Abstract
Compared to conventional antisera strategies, monoclonal antibodies (mAbs) represent an alternative and safer way to treat botulism, a fatal flaccid paralysis due to botulinum neurotoxins (BoNTs). In addition, mAbs offer the advantage to be produced in a reproducible manner. We previously identified a unique and potent mouse mAb (TA12) targeting BoNT/A1 with high affinity and neutralizing activity. In this study, we characterized the molecular basis of TA12 neutralization by combining Hydrogen/Deuterium eXchange Mass Spectrometry (HDX-MS) with site-directed mutagenesis and functional studies. We found that TA12 recognizes a conformational epitope located at the interface between the HCN and HCC subdomains of the BoNT/A1 receptor-binding domain (HC ). The TA12-binding interface shares common structural features with the ciA-C2 VHH epitope and lies on the face opposite recognized by ciA-C2- and the CR1/CR2-neutralizing mAbs. The single substitution of N1006 was sufficient to affect TA12 binding to HC confirming the position of the epitope. We further uncovered that the TA12 epitope overlaps with the BoNT/A1-binding site for both the neuronal cell surface receptor synaptic vesicle glycoprotein 2 isoform C (SV2C) and the GT1b ganglioside. Hence, TA12 potently blocks the entry of BoNT/A1 into neurons by interfering simultaneously with the binding of SV2C and to a lower extent GT1b. Our study reveals the unique neutralization mechanism of TA12 and emphasizes on the potential of using single mAbs for the treatment of botulism type A.
Collapse
Affiliation(s)
- Sébastien Brier
- Biological NMR Technological Platform, Institut Pasteur, CNRS UMR3528, Paris, France
| | | | - Anne Wijkhuisen
- Département Médicaments et Technologies pour la santé, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette, France
| | - Stéphanie Simon
- Département Médicaments et Technologies pour la santé, Université Paris-Saclay, CEA, INRAE, Gif-sur-Yvette, France
| | - Maud Marechal
- Institut Pasteur, Unité des Toxines Bactériennes, UMR CNRS 2001, Paris, France
| | - Emmanuel Lemichez
- Institut Pasteur, Unité des Toxines Bactériennes, UMR CNRS 2001, Paris, France
| | - Michel R Popoff
- Institut Pasteur, Unité des Toxines Bactériennes, UMR CNRS 2001, Paris, France
| |
Collapse
|
11
|
Characterization of clostridium botulinum neurotoxin serotype A (BoNT/A) and fibroblast growth factor receptor interactions using novel receptor dimerization assay. Sci Rep 2021; 11:7832. [PMID: 33837264 PMCID: PMC8035261 DOI: 10.1038/s41598-021-87331-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 03/24/2021] [Indexed: 01/03/2023] Open
Abstract
Clostridium botulinum neurotoxin serotype A (BoNT/A) is a potent neurotoxin that serves as an effective therapeutic for several neuromuscular disorders via induction of temporary muscular paralysis. Specific binding and internalization of BoNT/A into neuronal cells is mediated by its binding domain (HC/A), which binds to gangliosides, including GT1b, and protein cell surface receptors, including SV2. Previously, recombinant HC/A was also shown to bind to FGFR3. As FGFR dimerization is an indirect measure of ligand-receptor binding, an FCS & TIRF receptor dimerization assay was developed to measure rHC/A-induced dimerization of fluorescently tagged FGFR subtypes (FGFR1-3) in cells. rHC/A dimerized FGFR subtypes in the rank order FGFR3c (EC50 ≈ 27 nM) > FGFR2b (EC50 ≈ 70 nM) > FGFR1c (EC50 ≈ 163 nM); rHC/A dimerized FGFR3c with similar potency as the native FGFR3c ligand, FGF9 (EC50 ≈ 18 nM). Mutating the ganglioside binding site in HC/A, or removal of GT1b from the media, resulted in decreased dimerization. Interestingly, reduced dimerization was also observed with an SV2 mutant variant of HC/A. Overall, the results suggest that the FCS & TIRF receptor dimerization assay can assess FGFR dimerization with known and novel ligands and support a model wherein HC/A, either directly or indirectly, interacts with FGFRs and induces receptor dimerization.
Collapse
|
12
|
Structural Insights into Rational Design of Single-Domain Antibody-Based Antitoxins against Botulinum Neurotoxins. Cell Rep 2021; 30:2526-2539.e6. [PMID: 32101733 PMCID: PMC7138525 DOI: 10.1016/j.celrep.2020.01.107] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/23/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Botulinum neurotoxin (BoNT) is one of the most acutely lethal toxins known to humans, and effective treatment for BoNT intoxication is urgently needed. Single-domain antibodies (VHH) have been examined as a countermeasure for BoNT because of their high stability and ease of production. Here, we investigate the structures and the neutralization mechanisms for six unique VHHs targeting BoNT/A1 or BoNT/B1. These studies reveal diverse neutralizing mechanisms by which VHHs prevent host receptor binding or block transmembrane delivery of the BoNT protease domain. Guided by this knowledge, we design heterodimeric VHHs by connecting two neutralizing VHHs via a flexible spacer so they can bind simultaneously to the toxin. These bifunctional VHHs display much greater potency in a mouse co-intoxication model than similar heterodimers unable to bind simultaneously. Taken together, our studies offer insight into antibody neutralization of BoNTs and advance our ability to design multivalent anti-pathogen VHHs with improved therapeutic properties. Botulinum neurotoxins (BoNTs) are extremely toxic biothreats. Lam et al. report the crystal structures and neutralizing mechanisms of six unique antitoxin VHHs against BoNT/A1 and BoNT/B1, the two major human pathogenic BoNTs. They then develop a platform for structure-based rational design of bifunctional VHH heterodimers with superior antitoxin potencies.
Collapse
|
13
|
|
14
|
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
|
15
|
Production and characterization of a neutralizing antibody against botulinum neurotoxin A. J Immunol Methods 2020; 487:112871. [PMID: 33007319 DOI: 10.1016/j.jim.2020.112871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/09/2020] [Accepted: 09/24/2020] [Indexed: 11/20/2022]
Abstract
As a category A toxic, the botulinum toxin(BoNT) is responsible for human botulism with an estimated lethal dose of 1 ng/kg which greatly increases the potential risk of use as bioweapons. Therefore, the development of anti-BoNT antibodies is urgent. In this paper, the HC domain of BoNT/A was purified and immunized with Balb/c mice. Monoclonal antibodies were screened against BoNT/A from 55 stable positive hybridoma cell lines, and one with the strongest neutralizing activity, designated as ML06, was subcloned, sequenced, and classified as IgG1(κ) subclass. The mouse protection assays showed that ML06 can neutralize the toxin of BoNT/A effectively both in vitro and in vivo, in a dose-dependent manner. The therapeutic assays showed that only 20% of mice injected with 4 LD50 BoNT/A can survive another injection of ML06 after 4 h. The prophylaxis assays showed the residual ML06 from mice injected with ML06 two weeks ago can protect mice against 4 LD50 BoNT/A challenge completely. Collectively, our results indicated that ML06 served as a good candidate for further development of immune therapeutics for BoNT/A.
Collapse
|
16
|
Two VHH Antibodies Neutralize Botulinum Neurotoxin E1 by Blocking Its Membrane Translocation in Host Cells. Toxins (Basel) 2020; 12:toxins12100616. [PMID: 32992561 PMCID: PMC7599855 DOI: 10.3390/toxins12100616] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Botulinum neurotoxin serotype E (BoNT/E) is one of the major causes of human botulism, which is a life-threatening disease caused by flaccid paralysis of muscles. After receptor-mediated toxin internalization into motor neurons, the translocation domain (HN) of BoNT/E transforms into a protein channel upon vesicle acidification in endosomes and delivers its protease domain (LC) across membrane to enter the neuronal cytosol. It is believed that the rapid onset of BoNT/E intoxication compared to other BoNT serotypes is related to its swift internalization and translocation. We recently identified two neutralizing single-domain camelid antibodies (VHHs) against BoNT/E1 termed JLE-E5 and JLE-E9. Here, we report the crystal structures of these two VHHs bound to the LCHN domain of BoNT/E1. The structures reveal that these VHHs recognize two distinct epitopes that are partially overlapping with the putative transmembrane regions on HN, and therefore could physically block membrane association of BoNT/E1. This is confirmed by our in vitro studies, which show that these VHHs inhibit the structural change of BoNT/E1 at acidic pH and interfere with BoNT/E1 association with lipid vesicles. Therefore, these two VHHs neutralize BoNT/E1 by preventing the transmembrane delivery of LC. Furthermore, structure-based sequence analyses show that the 3-dimensional epitopes of these two VHHs are largely conserved across many BoNT/E subtypes, suggesting a broad-spectrum protection against the BoNT/E family. In summary, this work improves our understanding of the membrane translocation mechanism of BoNT/E and paves the way for developing VHHs as diagnostics or therapeutics for the treatment of BoNT/E intoxication.
Collapse
|
17
|
Davies JR, Britton A, Liu SM, Acharya KR. High-resolution crystal structures of the botulinum neurotoxin binding domains from subtypes A5 and A6. FEBS Open Bio 2020; 10:1474-1481. [PMID: 32654405 PMCID: PMC7396429 DOI: 10.1002/2211-5463.12931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 11/10/2022] Open
Abstract
Clostridium botulinum neurotoxins (BoNTs) cause flaccid paralysis through inhibition of acetylcholine release from motor neurons; however, at tiny doses, this property is exploited for use as a therapeutic. Each member of the BoNT family of proteins consists of three distinct domains: a binding domain that targets neuronal cell membranes (HC ), a translocation domain (HN ) and a catalytic domain (LC). Here, we present high-resolution crystal structures of the binding domains of BoNT subtypes/A5 (HC /A5) and/A6 (HC /A6). These structures show that the core fold identified in other subtypes is maintained, but with subtle differences at the expected receptor-binding sites.
Collapse
Affiliation(s)
- Jonathan R Davies
- Department of Biology and Biochemistry, Claverton Down, University of Bath, Bath, UK.,Department of Biochemistry and Biophysics, Stockholm University, Sweden
| | - Amy Britton
- Department of Biology and Biochemistry, Claverton Down, University of Bath, Bath, UK
| | - Sai Man Liu
- Ipsen Bioinnovation Limited, Milton Park, Abingdon, UK
| | - K Ravi Acharya
- Department of Biology and Biochemistry, Claverton Down, University of Bath, Bath, UK
| |
Collapse
|
18
|
Burstein R, Blumenfeld AM, Silberstein SD, Manack Adams A, Brin MF. Mechanism of Action of OnabotulinumtoxinA in Chronic Migraine: A Narrative Review. Headache 2020; 60:1259-1272. [PMID: 32602955 PMCID: PMC7496564 DOI: 10.1111/head.13849] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/20/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
Objective To review the literature on the mechanism of action of onabotulinumtoxinA in chronic migraine. Background OnabotulinumtoxinA is a chronic migraine preventive treatment that significantly reduces headache frequency. The traditional mechanism described for onabotulinumtoxinA – reducing muscle contractions – is insufficient to explain its efficacy in migraine, which is primarily a sensory neurological disease. Methods A narrative literature review on the mechanism of action of onabotulinumtoxinA in chronic migraine. Results Following injection into tissues, onabotulinumtoxinA inhibits soluble N‐ethylmaleimide‐sensitive fusion attachment protein receptor (SNARE)‐mediated vesicle trafficking by cleaving one of its essential proteins, soluble N‐ethylmaleimide‐sensitive fusion attachment protein (SNAP‐25), which occurs in both motor and sensory nerves. OnabotulinumtoxinA inhibits regulated exocytosis of motor and sensory neurochemicals and proteins, as well as membrane insertion of peripheral receptors that convey pain from the periphery to the brain, because both processes are SNARE dependent. OnabotulinumtoxinA can decrease exocytosis of pro‐inflammatory and excitatory neurotransmitters and neuropeptides such as substance P, calcitonin gene‐related peptide, and glutamate from primary afferent fibers that transmit nociceptive pain and participate in the development of peripheral and central sensitization. OnabotulinumtoxinA also decreases the insertion of pain‐sensitive ion channels such as transient receptor potential cation channel subfamily V member 1 (TRPV1) into the membranes of nociceptive neurons; this is likely enhanced in the sensitized neuron. For chronic migraine prevention, onabotulinumtoxinA is injected into 31‐39 sites in 7 muscles of the head and neck. Sensory nerve endings of neurons whose cell bodies are located in trigeminal and cervical ganglia are distributed throughout the injected muscles, and are overactive in people with migraine. Through inhibition of these sensory nerve endings, onabotulinumtoxinA reduces the number of pain signals that reach the brain and consequently prevents activation and sensitization of central neurons postulated to be involved in migraine chronification. Conclusion OnabotulinumtoxinA likely acts via sensory mechanisms to treat chronic migraine.
Collapse
Affiliation(s)
- Rami Burstein
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Andrew M Blumenfeld
- The Headache Center of Southern California, The Neurology Center, Carlsbad, CA, USA
| | - Stephen D Silberstein
- Department of Neurology Jefferson Headache Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Mitchell F Brin
- Allergan, Inc., Irvine, CA, USA.,University of California, Irvine, CA, USA
| |
Collapse
|
19
|
Yin L, Masuyer G, Zhang S, Zhang J, Miyashita SI, Burgin D, Lovelock L, Coker SF, Fu TM, Stenmark P, Dong M. Characterization of a membrane binding loop leads to engineering botulinum neurotoxin B with improved therapeutic efficacy. PLoS Biol 2020; 18:e3000618. [PMID: 32182233 PMCID: PMC7077807 DOI: 10.1371/journal.pbio.3000618] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/12/2020] [Indexed: 11/25/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are a family of bacterial toxins with seven major serotypes (BoNT/A–G). The ability of these toxins to target and bind to motor nerve terminals is a key factor determining their potency and efficacy. Among these toxins, BoNT/B is one of the two types approved for medical and cosmetic uses. Besides binding to well-established receptors, an extended loop in the C-terminal receptor-binding domain (HC) of BoNT/B (HC/B) has been proposed to also contribute to toxin binding to neurons by interacting with lipid membranes (termed lipid-binding loop [LBL]). Analogous loops exist in the HCs of BoNT/C, D, G, and a chimeric toxin DC. However, it has been challenging to detect and characterize binding of LBLs to lipid membranes. Here, using the nanodisc system and biolayer interferometry assays, we find that HC/DC, C, and G, but not HC/B and HC/D, are capable of binding to receptor-free lipids directly, with HC/DC having the highest level of binding. Mutagenesis studies demonstrate the critical role of consecutive aromatic residues at the tip of the LBL for binding of HC/DC to lipid membranes. Taking advantage of this insight, we then create a “gain-of-function” mutant HC/B by replacing two nonaromatic residues at the tip of its LBL with tryptophan. Cocrystallization studies confirm that these two tryptophan residues do not alter the structure of HC/B or the interactions with its receptors. Such a mutated HC/B gains the ability to bind receptor-free lipid membranes and shows enhanced binding to cultured neurons. Finally, full-length BoNT/B containing two tryptophan mutations in its LBL, together with two additional mutations (E1191M/S1199Y) that increase binding to human receptors, is produced and evaluated in mice in vivo using Digit Abduction Score assays. This mutant toxin shows enhanced efficacy in paralyzing local muscles at the injection site and lower systemic diffusion, thus extending both safety range and duration of paralysis compared with the control BoNT/B. These findings establish a mechanistic understanding of LBL–lipid interactions and create a modified BoNT/B with improved therapeutic efficacy. Botulinum neurotoxins are a family of bacterial toxins, some of which are approved for medical and cosmetic uses. This study shows that introducing aromatic residues to a lipid binding loop improved therapeutic efficacy of botulinum neurotoxin B by enhancing its ability to bind to lipid membranes at motor nerve terminals.
Collapse
Affiliation(s)
- Linxiang Yin
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Geoffrey Masuyer
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Sicai Zhang
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jie Zhang
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shin-Ichiro Miyashita
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | | | | | - Tian-min Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- * E-mail: (PS); (MD)
| | - Min Dong
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (PS); (MD)
| |
Collapse
|
20
|
Poulain B, Lemichez E, Popoff MR. Neuronal selectivity of botulinum neurotoxins. Toxicon 2020; 178:20-32. [PMID: 32094099 DOI: 10.1016/j.toxicon.2020.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Botulinum neurotoxins (BoNTs) are highly potent toxins responsible for a severe disease, called botulism. They are also efficient therapeutic tools with an increasing number of indications ranging from neuromuscular dysfunction to hypersecretion syndrome, pain release, depression as well as cosmetic application. BoNTs are known to mainly target the motor-neurons terminals and to induce flaccid paralysis. BoNTs recognize a specific double receptor on neuronal cells consisting of gangliosides and synaptic vesicle protein, SV2 or synaptotagmin. Using cultured neuronal cells, BoNTs have been established blocking the release of a wide variety of neurotransmitters. However, BoNTs are more potent in motor-neurons than in the other neuronal cell types. In in vivo models, BoNT/A impairs the cholinergic neuronal transmission at the motor-neurons but also at neurons controlling secretions and smooth muscle neurons, and blocks several neuronal pathways including excitatory, inhibitory, and sensitive neurons. However, only a few reports investigated the neuronal selectivity of BoNTs in vivo. In the intestinal wall, BoNT/A and BoNT/B target mainly the cholinergic neurons and to a lower extent the other non-cholinergic neurons including serotonergic, glutamatergic, GABAergic, and VIP-neurons. The in vivo effects induced by BoNTs on the non-cholinergic neurons remain to be precisely investigated. We report here a literature review of the neuronal selectivity of BoNTs.
Collapse
Affiliation(s)
- Bernard Poulain
- Université de Strasbourg, CNRS, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | | | | |
Collapse
|
21
|
Gregory KS, Liu SM, Acharya KR. Crystal structure of botulinum neurotoxin subtype A3 cell binding domain in complex with GD1a co-receptor ganglioside. FEBS Open Bio 2020; 10:298-305. [PMID: 31945264 PMCID: PMC7050238 DOI: 10.1002/2211-5463.12790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 01/20/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are one of the most toxic proteins known to humans. Their molecular structure is comprised of three essential domains—a cell binding domain (HC), translocation domain and catalytic domain (light chain) . The HC domain facilitates the highly specific binding of BoNTs to the neuronal membrane via a dual‐receptor complex involving a protein receptor and a ganglioside. Variation in activity/toxicity across subtypes of serotype A has been attributed to changes in protein and ganglioside interactions, and their implications are important in the design of novel BoNT‐based therapeutics. Here, we present the structure of BoNT/A3 cell binding domain (HC/A3) in complex with the ganglioside GD1a at 1.75 Å resolution. The structure revealed that six residues interact with the three outermost monosaccharides of GD1a through several key hydrogen bonding interactions. A detailed comparison of structures of HC/A3 with HC/A1 revealed subtle conformational differences at the ganglioside binding site upon carbohydrate binding.
Collapse
Affiliation(s)
- Kyle S Gregory
- Department of Biology and Biochemistry, Claverton Down, University of Bath, UK
| | | | - K Ravi Acharya
- Department of Biology and Biochemistry, Claverton Down, University of Bath, UK
| |
Collapse
|
22
|
Abstract
Botulinum neurotoxins (BoNTs) are a family of bacterial protein toxins produced by various Clostridium species. They are traditionally classified into seven major serotypes (BoNT/A-G). Recent progress in sequencing microbial genomes has led to an ever-growing number of subtypes, chimeric toxins, BoNT-like toxins, and remotely related BoNT homologs, constituting an expanding BoNT superfamily. Recent structural studies of BoNTs, BoNT progenitor toxin complexes, tetanus neurotoxin (TeNT), toxin-receptor complexes, and toxin-substrate complexes have provided mechanistic understandings of toxin functions and the molecular basis for their variations. The growing BoNT superfamily of toxins present a natural repertoire that can be explored to develop novel therapeutic toxins, and the structural understanding of their variations provides a knowledge basis for engineering toxins to improve therapeutic efficacy and expand their clinical applications.
Collapse
Affiliation(s)
- Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
23
|
Abstract
Botulinum neurotoxins (BoNTs) and tetanus neurotoxin (TeNT) are the most potent toxins known and cause botulism and tetanus, respectively. BoNTs are also widely utilized as therapeutic toxins. They contain three functional domains responsible for receptor-binding, membrane translocation, and proteolytic cleavage of host proteins required for synaptic vesicle exocytosis. These toxins also have distinct features: BoNTs exist within a progenitor toxin complex (PTC), which protects the toxin and facilitates its absorption in the gastrointestinal tract, whereas TeNT is uniquely transported retrogradely within motor neurons. Our increasing knowledge of these toxins has allowed the development of engineered toxins for medical uses. The discovery of new BoNTs and BoNT-like proteins provides additional tools to understand the evolution of the toxins and to engineer toxin-based therapeutics. This review summarizes the progress on our understanding of BoNTs and TeNT, focusing on the PTC, receptor recognition, new BoNT-like toxins, and therapeutic toxin engineering.
Collapse
Affiliation(s)
- Min Dong
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Geoffrey Masuyer
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden;
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden; .,Department of Experimental Medical Science, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
24
|
Abstract
Gangliosides comprise a varied family of glycosphingolipid structures bearing one or more sialic acid residues. They are found in all mammalian tissues but are most abundant in the brain, where they represent the quantitatively major class of sialoglycans. As prominent molecular determinants on cell surfaces, they function as molecular-recognition partners for diverse glycan-binding proteins ranging from bacterial toxins to endogenous cell-cell adhesion molecules. Gangliosides also regulate the activity of plasma membrane proteins, including protein tyrosine kinases, by lateral association in the same membranes in which they reside. Their roles in molecular recognition and membrane protein regulation implicate gangliosides in human physiology and pathology, including infectious diseases, diabetes, cancer, and neurodegeneration. The varied structures and biosynthetic pathways of gangliosides are presented here, along with representative examples of their biological functions in health and disease.
Collapse
|
25
|
Davies JR, Liu SM, Acharya KR. Variations in the Botulinum Neurotoxin Binding Domain and the Potential for Novel Therapeutics. Toxins (Basel) 2018; 10:toxins10100421. [PMID: 30347838 PMCID: PMC6215321 DOI: 10.3390/toxins10100421] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/11/2018] [Accepted: 10/18/2018] [Indexed: 01/23/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are categorised into immunologically distinct serotypes BoNT/A to /G). Each serotype can also be further divided into subtypes based on differences in amino acid sequence. BoNTs are ~150 kDa proteins comprised of three major functional domains: an N-terminal zinc metalloprotease light chain (LC), a translocation domain (HN), and a binding domain (HC). The HC is responsible for targeting the BoNT to the neuronal cell membrane, and each serotype has evolved to bind via different mechanisms to different target receptors. Most structural characterisations to date have focussed on the first identified subtype within each serotype (e.g., BoNT/A1). Subtype differences within BoNT serotypes can affect intoxication, displaying different botulism symptoms in vivo, and less emphasis has been placed on investigating these variants. This review outlines the receptors for each BoNT serotype and describes the basis for the highly specific targeting of neuronal cell membranes. Understanding receptor binding is of vital importance, not only for the generation of novel therapeutics but also for understanding how best to protect from intoxication.
Collapse
Affiliation(s)
- Jonathan R Davies
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK.
| | - Sai Man Liu
- Ipsen Bioinnovation Limited, Abingdon OX14 4RY, UK.
| | - K Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK.
| |
Collapse
|
26
|
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
|
27
|
Gustafsson R, Zhang S, Masuyer G, Dong M, Stenmark P. Crystal Structure of Botulinum Neurotoxin A2 in Complex with the Human Protein Receptor SV2C Reveals Plasticity in Receptor Binding. Toxins (Basel) 2018; 10:E153. [PMID: 29649119 PMCID: PMC5923319 DOI: 10.3390/toxins10040153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/28/2018] [Accepted: 04/10/2018] [Indexed: 11/16/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are a family of highly dangerous bacterial toxins, with seven major serotypes (BoNT/A-G). Members of BoNTs, BoNT/A1 and BoNT/B1, have been utilized to treat an increasing number of medical conditions. The clinical trials are ongoing for BoNT/A2, another subtype of BoNT/A, which showed promising therapeutic properties. Both BoNT/A1 and BoNT/A2 utilize three isoforms of synaptic vesicle protein SV2 (SV2A, B, and C) as their protein receptors. We here present a high resolution (2.0 Å) co-crystal structure of the BoNT/A2 receptor-binding domain in complex with the human SV2C luminal domain. The structure is similar to previously reported BoNT/A-SV2C complexes, but a shift of the receptor-binding segment in BoNT/A2 rotates SV2C in two dimensions giving insight into the dynamic behavior of the interaction. Small differences in key residues at the binding interface may influence the binding to different SV2 isoforms, which may contribute to the differences between BoNT/A1 and BoNT/A2 observed in the clinic.
Collapse
Affiliation(s)
- Robert Gustafsson
- Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden.
| | - Sicai Zhang
- Department of Urology, Boston Children's Hospital, Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.
| | - Geoffrey Masuyer
- Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden.
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden.
| |
Collapse
|
28
|
Davies JR, Hackett GS, Liu SM, Acharya KR. High resolution crystal structures of the receptor-binding domain of Clostridium botulinum neurotoxin serotypes A and FA. PeerJ 2018; 6:e4552. [PMID: 29576992 PMCID: PMC5866713 DOI: 10.7717/peerj.4552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/08/2018] [Indexed: 01/12/2023] Open
Abstract
The binding specificity of botulinum neurotoxins (BoNTs) is primarily a consequence of their ability to bind to multiple receptors at the same time. BoNTs consist of three distinct domains, a metalloprotease light chain (LC), a translocation domain (HN) and a receptor-binding domain (HC). Here we report the crystal structure of HC/FA, complementing an existing structure through the modelling of a previously unresolved loop which is important for receptor-binding. Our HC/FA structure also contains a previously unidentified disulphide bond, which we have also observed in one of two crystal forms of HC/A1. This may have implications for receptor-binding and future recombinant toxin production.
Collapse
Affiliation(s)
- Jonathan R Davies
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | | | - Sai Man Liu
- Ipsen Bioinnovation Limited, Abingdon, United Kingdom
| | - K Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| |
Collapse
|
29
|
Zuverink M, Barbieri JT. Protein Toxins That Utilize Gangliosides as Host Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:325-354. [PMID: 29747819 DOI: 10.1016/bs.pmbts.2017.11.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Subsets of protein toxins utilize gangliosides as host receptors. Gangliosides are preferred receptors due to their extracellular localization on the eukaryotic cell and due to their essential nature in host physiology. Glycosphingolipids, including gangliosides, are mediators of signal transduction within and between eukaryotic cells. Protein toxins possess AB structure-function organization, where the A domain encodes a catalytic function for the posttranslational modification of a host macromolecule, including proteins and nucleic acids, and a B domain, which encodes host receptor recognition, including proteins and glycosphingolipids, alone or in combination. Protein toxins use similar strategies to bind glycans by pockets and loops, generally employing hydrogen bonding and aromatic stacking to stabilize interactions with sugars. In some cases, glycan binding facilitates uptake, while in other cases, cross-linking or a second receptor is necessary to stimulate entry. The affinity that protein toxins have for host glycans is necessary for tissue targeting, but not always sufficient to cause disease. In addition to affinity for binding the glycan, the lipid moiety also plays an important role in productive uptake and tissue tropism. Upon endocytosis, the protein toxin must escape to another intracellular compartment or into cytosol to modify a host substrate, modulating host signaling, often resulting in cytotoxic or apoptotic events in the cell, and a unique morbidity for the organism. The study of protein toxins that utilize gangliosides as host receptors has illuminated numerous eukaryotic cellular processes, identified the basis for developing interventions to prevent disease through vaccines and control bacterial diseases through therapies. In addition, subsets of these protein toxins have been utilized as therapeutic agents to treat numerous human inflictions.
Collapse
|
30
|
Yu CH, Song DH, Choi JY, Joe HE, Jeong WH, Hur GH, Shin YK, Jeong ST. A mutated recombinant subunit vaccine protects mice and guinea pigs against botulinum type A intoxication. Hum Vaccin Immunother 2018; 14:329-336. [PMID: 29140753 PMCID: PMC5806659 DOI: 10.1080/21645515.2017.1405201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/17/2017] [Accepted: 11/09/2017] [Indexed: 02/08/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are the most potent toxins to mammals. A toxoid vaccine was previously used for prevention of botulinum intoxication; however, this vaccine is no longer available. Currently, no approved botulinum vaccines are available from the Food and Drug Administration (FDA). Recently, a recombinant host cell receptor-binding subunit created for use as a potential vaccine completed phase 2 clinical trials. The current study designed a vaccine candidate against BoNT type A (BoNT/A) using a structural design. Our vaccine candidate was the BoNT/A heavy chain C-terminal region (HCR) that contained the point mutation BA15 (R1269A) within the ganglioside-binding site. A Biacore affinity test showed that the affinity of BA15 for ganglioside GT1b was 100 times lower than that of the HCR. A SNAP25 cleavage assay revealed that immunized sera blocked SNAP25 cleavage of the BoNT/A toxin via BA15. In an in vivo experiment, mice and guinea pigs immunized with BA15 produced neutralizing antibodies that protected against 3,000 LD50 of BoNT/A. In conclusion, the results of both in vitro and in vivo assays showed that our BA15 vaccine candidate was similar to the recombinant host cell receptor-binding subunit vaccine. The inability of BA15to bind ganglioside shows that BA15 is a potential safe vaccine candidate.
Collapse
Affiliation(s)
- Chi Ho Yu
- Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Dong Hyun Song
- Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Jun Young Choi
- Abion R&D Institute, Hanhwa Biz-Metro, Guro-gu, Seoul, Republic of Korea
| | - Hae Eun Joe
- Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Woo Hyeon Jeong
- Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Gyeung Haeng Hur
- Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Young Kee Shin
- Department of Pharmacy, College of Pharmacy, Seoul National University, Dajeon, Seoul, Republic of Korea
| | - Seong Tae Jeong
- Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| |
Collapse
|
31
|
Davies JR, Rees J, Liu SM, Acharya KR. High resolution crystal structures of Clostridium botulinum neurotoxin A3 and A4 binding domains. J Struct Biol 2017; 202:113-117. [PMID: 29288126 DOI: 10.1016/j.jsb.2017.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/20/2017] [Accepted: 12/23/2017] [Indexed: 11/27/2022]
Abstract
Clostridium botulinum neurotoxins (BoNTs) cause the life-threatening condition, botulism. However, while they have the potential to cause serious harm, they are increasingly being utilised for therapeutic applications. BoNTs comprise of seven distinct serotypes termed BoNT/A through BoNT/G, with the most widely characterised being sub-serotype BoNT/A1. Each BoNT consists of three structurally distinct domains, a binding domain (HC), a translocation domain (HN), and a proteolytic domain (LC). The HC domain is responsible for the highly specific targeting of the neurotoxin to neuronal cell membranes. Here, we present two high-resolution structures of the binding domain of subtype BoNT/A3 (HC/A3) and BoNT/A4 (HC/A4) at 1.6 Å and 1.34 Å resolution, respectively. The structures of both proteins share a high degree of similarity to other known BoNT HC domains whilst containing some subtle differences, and are of benefit to research into therapeutic neurotoxins with novel characteristics.
Collapse
Affiliation(s)
- Jonathan R Davies
- Department of Biology and Biochemistry, Claverton Down, University of Bath, Bath BA2 7AY, UK
| | - Jay Rees
- Department of Biology and Biochemistry, Claverton Down, University of Bath, Bath BA2 7AY, UK
| | - Sai Man Liu
- Ipsen Bioinnovation Limited, 102 Park Lane, Milton Park, Abingdon OX14 4RY, UK
| | - K Ravi Acharya
- Department of Biology and Biochemistry, Claverton Down, University of Bath, Bath BA2 7AY, UK.
| |
Collapse
|
32
|
Zhang S, Berntsson RPA, Tepp WH, Tao L, Johnson EA, Stenmark P, Dong M. Structural basis for the unique ganglioside and cell membrane recognition mechanism of botulinum neurotoxin DC. Nat Commun 2017; 8:1637. [PMID: 29158482 PMCID: PMC5696347 DOI: 10.1038/s41467-017-01534-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/25/2017] [Indexed: 12/15/2022] Open
Abstract
Botulinum neurotoxins (BoNTs), the most potent toxins known, are potential bioterrorism agents. It is well established that all seven serotypes of BoNTs (BoNT/A-G) require complex gangliosides as co-receptors. Here, we report that BoNT/DC, a presumed mosaic toxin between BoNT/D and BoNT/C1, binds and enters efficiently into neurons lacking complex gangliosides and shows no reduction in toxicity in mice deficient in complex gangliosides. The co-crystal structure of BoNT/DC with sialyl-Thomsen-Friedenreich antigen (Sialyl-T) suggests that BoNT/DC recognizes only the sialic acid, but not other moieties in gangliosides. Using liposome flotation assays, we demonstrate that an extended loop in BoNT/DC directly interacts with lipid membranes, and the co-occurring sialic acid binding and loop-membrane interactions mediate the recognition of gangliosides in membranes by BoNT/DC. These findings reveal a unique mechanism for cell membrane recognition and demonstrate that BoNT/DC can use a broad range of sialic acid-containing moieties as co-receptors.
Collapse
Affiliation(s)
- Sicai Zhang
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Ronnie P-A Berntsson
- Department of Biochemistry and Biophysics, Stockholm University, SE-106 91, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87, Umeå, Sweden
| | - William H Tepp
- Department of Bacteriology, University of Wisconsin, Madison, WI, 53706, USA
| | - Liang Tao
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Eric A Johnson
- Department of Bacteriology, University of Wisconsin, Madison, WI, 53706, USA
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, SE-106 91, Stockholm, Sweden.
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
Yao G, Lam KH, Weisemann J, Peng L, Krez N, Perry K, Shoemaker CB, Dong M, Rummel A, Jin R. A camelid single-domain antibody neutralizes botulinum neurotoxin A by blocking host receptor binding. Sci Rep 2017; 7:7438. [PMID: 28785006 PMCID: PMC5547058 DOI: 10.1038/s41598-017-07457-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/23/2017] [Indexed: 01/07/2023] Open
Abstract
Antibody treatment is currently the only available countermeasure for botulism, a fatal illness caused by flaccid paralysis of muscles due to botulinum neurotoxin (BoNT) intoxication. Among the seven major serotypes of BoNT/A-G, BoNT/A poses the most serious threat to humans because of its high potency and long duration of action. Prior to entering neurons and blocking neurotransmitter release, BoNT/A recognizes motoneurons via a dual-receptor binding process in which it engages both the neuron surface polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Previously, we identified a potent neutralizing antitoxin against BoNT/A1 termed ciA-C2, derived from a camelid heavy-chain-only antibody (VHH). In this study, we demonstrate that ciA-C2 prevents BoNT/A1 intoxication by inhibiting its binding to neuronal receptor SV2. Furthermore, we determined the crystal structure of ciA-C2 in complex with the receptor-binding domain of BoNT/A1 (HCA1) at 1.68 Å resolution. The structure revealed that ciA-C2 partially occupies the SV2-binding site on HCA1, causing direct interference of HCA1 interaction with both the N-glycan and peptide-moiety of SV2. Interestingly, this neutralization mechanism is similar to that of a monoclonal antibody in clinical trials, despite that ciA-C2 is more than 10-times smaller. Taken together, these results enlighten our understanding of BoNT/A1 interactions with its neuronal receptor, and further demonstrate that inhibiting toxin binding to the host receptor is an efficient countermeasure strategy.
Collapse
Affiliation(s)
- Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Jasmin Weisemann
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Lisheng Peng
- Department of Urology, Boston Children's Hospital, Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Nadja Krez
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, Illinois, USA
| | - Charles B Shoemaker
- Department of Infectious Diseases and Global Health, Tufts Clinical and Translational Science Institute, North Grafton, Massachusetts, USA
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas Rummel
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, California, USA.
| |
Collapse
|
34
|
Burns JR, Lambert GS, Baldwin MR. Insights into the Mechanisms by Which Clostridial Neurotoxins Discriminate between Gangliosides. Biochemistry 2017; 56:2571-2583. [DOI: 10.1021/acs.biochem.6b01246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Joshua R. Burns
- Department of Molecular Microbiology
and Immunology, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Gregory S. Lambert
- Department of Molecular Microbiology
and Immunology, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Michael R. Baldwin
- Department of Molecular Microbiology
and Immunology, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| |
Collapse
|
35
|
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
|
36
|
Benoit RM, Schärer MA, Wieser MM, Li X, Frey D, Kammerer RA. Crystal structure of the BoNT/A2 receptor-binding domain in complex with the luminal domain of its neuronal receptor SV2C. Sci Rep 2017; 7:43588. [PMID: 28252640 PMCID: PMC5333631 DOI: 10.1038/srep43588] [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: 11/08/2016] [Accepted: 01/25/2017] [Indexed: 11/09/2022] Open
Abstract
A detailed molecular understanding of botulinum neurotoxin (BoNT)/host-cell-receptor interactions is fundamental both for developing strategies against botulism and for generating improved BoNT variants for medical applications. The X-ray crystal structure of the receptor-binding domain (HC) of BoNT/A1 in complex with the luminal domain (LD) of its neuronal receptor SV2C revealed only few specific side-chain - side-chain interactions that are important for binding. Notably, two BoNT/A1 residues, Arg 1156 and Arg 1294, that are crucial for the interaction with SV2, are not conserved among subtypes. Because it has been suggested that differential receptor binding of subtypes might explain their differences in biological activity, we determined the crystal structure of BoNT/A2-HC in complex with SV2C-LD. Although only few side-chain interactions are conserved between the two BoNT/A subtypes, the overall binding mode of subtypes A1 and A2 is virtually identical. In the BoNT/A2-HC - SV2C complex structure, a missing cation-π stacking is compensated for by an additional salt bridge and an anion-π stacking interaction, which explains why the binding of BoNT/A subtypes to SV2C tolerates variable side chains. These findings suggest that motif extensions and a shallow binding cleft in BoNT/A-HC contribute to binding specificity.
Collapse
Affiliation(s)
- Roger M Benoit
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Martin A Schärer
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Mara M Wieser
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Xiaodan Li
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Daniel Frey
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Richard A Kammerer
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| |
Collapse
|
37
|
Shimabukuro J, Makyio H, Suzuki T, Nishikawa Y, Kawasaki M, Imamura A, Ishida H, Ando H, Kato R, Kiso M. Synthesis of seleno-fucose compounds and their application to the X-ray structural determination of carbohydrate-lectin complexes using single/multi-wavelength anomalous dispersion phasing. Bioorg Med Chem 2017; 25:1132-1142. [DOI: 10.1016/j.bmc.2016.12.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 12/18/2022]
|
38
|
Connan C, Popoff MR. Uptake of Clostridial Neurotoxins into Cells and Dissemination. Curr Top Microbiol Immunol 2017; 406:39-78. [PMID: 28879524 DOI: 10.1007/82_2017_50] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clostridial neurotoxins, botulinum neurotoxins (BoNT) and tetanus neurotoxin (TeNT), are potent toxins, which are responsible for severe neurological diseases in man and animals. BoNTs induce a flaccid paralysis (botulism) by inhibiting acetylcholine release at the neuromuscular junctions, whereas TeNT causes a spastic paralysis (tetanus) by blocking the neurotransmitter release (glycine, GABA) in inhibitory interneurons within the central nervous system. Clostridial neurotoxins recognize specific receptor(s) on the target neuronal cells and enter via a receptor-mediated endocytosis. They transit through an acidic compartment which allows the translocation of the catalytic chain into the cytosol, a prerequisite step for the intracellular activity of the neurotoxins. TeNT migrates to the central nervous system by using a motor neuron as transport cell. TeNT enters a neutral pH compartment and undergoes a retrograde axonal transport to the spinal cord or brain, where the whole undissociated toxin is delivered and interacts with target neurons. Botulism most often results from ingestion of food contaminated with BoNT. Thus, BoNT passes through the intestinal epithelial barrier mainly via a transcytotic mechanism and then diffuses or is transported to the neuromuscular junctions by the lymph or blood circulation. Indeed, clostridial neurotoxins are specific neurotoxins which transit through a transport cell to gain access to the target neuron, and use distinct trafficking pathways in both cell types.
Collapse
Affiliation(s)
- Chloé Connan
- Unité Des Bactéries Anaérobies et Toxines, Institut Pasteur, 25 Rue Du Dr Roux, 75724, Paris Cedex 15, France
| | - Michel R Popoff
- Unité Des Bactéries Anaérobies et Toxines, Institut Pasteur, 25 Rue Du Dr Roux, 75724, Paris Cedex 15, France.
| |
Collapse
|
39
|
Entry of Botulinum Neurotoxin Subtypes A1 and A2 into Neurons. Infect Immun 2016; 85:IAI.00795-16. [PMID: 27795365 DOI: 10.1128/iai.00795-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/05/2016] [Indexed: 11/20/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are the most toxic proteins for humans but also are common therapies for neurological diseases. BoNTs are dichain toxins, comprising an N-terminal catalytic domain (LC) disulfide bond linked to a C-terminal heavy chain (HC) which includes a translocation domain (HN) and a receptor binding domain (HC). Recently, the BoNT serotype A (BoNT/A) subtypes A1 and A2 were reported to possess similar potencies but different rates of cellular intoxication and pathology in a mouse model of botulism. The current study measured HCA1 and HCA2 entry into rat primary neurons and cultured Neuro2A cells. We found that there were two sequential steps during the association of BoNT/A with neurons. The initial step was ganglioside dependent, while the subsequent step involved association with synaptic vesicles. HCA1 and HCA2 entered the same population of synaptic vesicles and entered cells at similar rates. The primary difference was that HCA2 had a higher degree of receptor occupancy for cells and neurons than HcA1. Thus, HCA2 and HCA1 share receptors and entry pathway but differ in their affinity for receptor. The initial interaction of HCA1 and HCA2 with neurons may contribute to the unique pathologies of BoNT/A1 and BoNT/A2 in mouse models.
Collapse
|
40
|
Hamark C, Berntsson RPA, Masuyer G, Henriksson LM, Gustafsson R, Stenmark P, Widmalm G. Glycans Confer Specificity to the Recognition of Ganglioside Receptors by Botulinum Neurotoxin A. J Am Chem Soc 2016; 139:218-230. [PMID: 27958736 DOI: 10.1021/jacs.6b09534] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The highly poisonous botulinum neurotoxins, produced by the bacterium Clostridium botulinum, act on their hosts by a high-affinity association to two receptors on neuronal cell surfaces as the first step of invasion. The glycan motifs of gangliosides serve as initial coreceptors for these protein complexes, whereby a membrane protein receptor is bound. Herein we set out to characterize the carbohydrate minimal binding epitope of the botulinum neurotoxin serotype A. By means of ligand-based NMR spectroscopy, X-ray crystallography, computer simulations, and isothermal titration calorimetry, a screening of ganglioside analogues together with a detailed characterization of various carbohydrate ligand complexes with the toxin were accomplished. We show that the representation of the glycan epitope to the protein affects the details of binding. Notably, both branches of the oligosaccharide GD1a can associate to botulinum neurotoxin serotype A when expressed as individual trisaccharides. It is, however, the terminal branch of GD1a as well as this trisaccharide motif alone, corresponding to the sialyl-Thomsen-Friedenreich antigen, that represents the active ligand epitope, and these compounds bind to the neurotoxin with a high degree of predisposition but with low affinities. This finding does not correlate with the oligosaccharide moieties having a strong contribution to the total affinity, which was expected to be the case. We here propose that the glycan part of the ganglioside receptors mainly provides abundance and specificity, whereas the interaction with the membrane itself and protein receptor brings about the strong total binding of the toxin to the neuronal membrane.
Collapse
Affiliation(s)
- Christoffer Hamark
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| | - Ronnie P-A Berntsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| | - Geoffrey Masuyer
- Department of Biochemistry and Biophysics, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| | - Linda M Henriksson
- Department of Biochemistry and Biophysics, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| | - Robert Gustafsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| | - Göran Widmalm
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University , S-106 91 Stockholm, Sweden
| |
Collapse
|
41
|
Biological toxins of potential bioterrorism risk: Current status of detection and identification technology. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.05.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
42
|
Yao G, Zhang S, Mahrhold S, Lam KH, Stern D, Bagramyan K, Perry K, Kalkum M, Rummel A, Dong M, Jin R. N-linked glycosylation of SV2 is required for binding and uptake of botulinum neurotoxin A. Nat Struct Mol Biol 2016; 23:656-62. [PMID: 27294781 PMCID: PMC5033645 DOI: 10.1038/nsmb.3245] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/13/2016] [Indexed: 02/01/2023]
Abstract
Botulinum neurotoxin serotype A1 (BoNT/A1), a licensed drug widely used for medical and cosmetic applications, exerts its action by invading motoneurons. Here we report a 2.0-Å-resolution crystal structure of the BoNT/A1 receptor-binding domain in complex with its neuronal receptor, glycosylated human SV2C. We found that the neuronal tropism of BoNT/A1 requires recognition of both the peptide moiety and an N-linked glycan on SV2. This N-glycan-which is conserved in all SV2 isoforms across vertebrates-is essential for BoNT/A1 binding to neurons and for its potent neurotoxicity. The glycan-binding interface on SV2 is targeted by a human BoNT/A1-neutralizing antibody currently licensed as an antibotulism drug. Our studies reveal a new paradigm of host-pathogen interactions, in which pathogens exploit conserved host post-translational modifications, thereby achieving highly specific receptor binding while also tolerating genetic changes across multiple isoforms of receptors.
Collapse
Affiliation(s)
- Guorui Yao
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Sicai Zhang
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan Mahrhold
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Kwok-ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Daniel Stern
- Centre for Biological Threats and Special Pathogens - Biological Toxins (ZBS3), Robert Koch-Institut, Berlin, Germany
| | - Karine Bagramyan
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California USA
| | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, Illinois USA
| | - Markus Kalkum
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California USA
| | - Andreas Rummel
- Institut für Toxikologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Min Dong
- Department of Urology, Boston Children’s Hospital, Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| |
Collapse
|
43
|
Only the complex N559-glycan in the synaptic vesicle glycoprotein 2C mediates high affinity binding to botulinum neurotoxin serotype A1. Biochem J 2016; 473:2645-54. [PMID: 27313224 DOI: 10.1042/bcj20160439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/16/2016] [Indexed: 11/17/2022]
Abstract
The extraordinary potency of botulinum neurotoxins (BoNTs) is mediated by their high neurospecificity, targeting peripheral cholinergic motoneurons leading to flaccid paralysis and successive respiratory failure. Complex polysialo gangliosides accumulate BoNTs on the plasma membrane and facilitate subsequent binding to synaptic vesicle membrane proteins which results in toxin endocytosis. The luminal domain 4 (LD4) of the three synaptic vesicle glycoprotein 2 (SV2) isoforms A-C mediates uptake of the clinically most relevant serotype BoNT/A1. SV2C-LD4 exhibits the strongest protein-protein interaction and comprises five putative N-glycosylation sites (PNG sites). Here, we expressed human SV2C-LD4 fused to human IgG-Fc in prokaryotic and eukaryotic expression systems to analyse the effect of N-glycosylation of SV2C on the interaction with BoNT/A1. Mass spectrometric analysis of gSV2CLD-Fc demonstrates glycosylation of N534, N559 and N565, the latter two residing at the BoNT/A interface. Mutational analysis demonstrates that only the N559-glycan, but not N565-glycan increases affinity of BoNT/A for human gSV2C-LD4. The N559-glycan was characterised as a complex core-fucosylated type with a heterogeneity ranging up to tetra-antennary structure with bisecting N-acetylglucosamine which can establish extensive interactions with BoNT/A. The mutant gSV2CLD-Fc N559A displayed a 50-fold increased dissociation rate kd resulting in an overall 12-fold decreased binding affinity in surface plasmon resonance (SPR) experiments. The delayed dissociation might provide BoNT/A more time for endocytosis into synaptic vesicles. In conclusion, we show the importance of the complex N559-glycan of SV2C-LD4, adding a third anchor point beside a ganglioside and the SV2C-LD4 peptide, for BoNT/A neuronal cell surface binding and uptake.
Collapse
|
44
|
Botulinum Neurotoxin Serotype A Recognizes Its Protein Receptor SV2 by a Different Mechanism than Botulinum Neurotoxin B Synaptotagmin. Toxins (Basel) 2016; 8:toxins8050154. [PMID: 27196927 PMCID: PMC4885069 DOI: 10.3390/toxins8050154] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/28/2016] [Accepted: 05/09/2016] [Indexed: 11/23/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) exhibit extraordinary potency due to their exquisite neurospecificity, which is achieved by dual binding to complex polysialo-gangliosides and synaptic vesicle proteins. The luminal domain 4 (LD4) of the three synaptic vesicle glycoprotein 2 isoforms, SV2A‐C, identified as protein receptors for the most relevant serotype BoNT/A, binds within the 50 kDa cell binding domain HC of BoNT/A. Here, we deciphered the BoNT/A‐SV2 interactions in more detail. In pull down assays, the binding of HCA to SV2-LD4 isoforms decreases from SV2C >> SV2A > SV2B. A binding constant of 200 nM was determined for BoNT/A to rat SV2C-LD4 in GST pull down assay. A similar binding constant was determined by surface plasmon resonance for HCA to rat SV2C and to human SV2C, the latter being slightly lower due to the substitution L563F in LD4. At pH 5, as measured in acidic synaptic vesicles, the binding constant of HCA to hSV2C is increased more than 10-fold. Circular dichroism spectroscopy reveals that the quadrilateral helix of SV2C-LD4 already exists in solution prior to BoNT/A binding. Hence, the BoNT/A‐SV2C interaction is of different nature compared to BoNT/B‐Syt-II. In particular, the preexistence of the quadrilateral β-sheet helix of SV2 and its pH-dependent binding to BoNT/A via backbone–backbone interactions constitute major differences. Knowledge of the molecular details of BoNT/A‐SV2 interactions drives the development of high affinity peptides to counteract BoNT/A intoxications or to capture functional BoNT/A variants in innovative detection systems for botulism diagnostic.
Collapse
|
45
|
Rummel A. Two Feet on the Membrane: Uptake of Clostridial Neurotoxins. Curr Top Microbiol Immunol 2016; 406:1-37. [PMID: 27921176 DOI: 10.1007/82_2016_48] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The extraordinary potency of botulinum neurotoxins (BoNT) and tetanus neurotoxin (TeNT) is mediated by their high neurospecificity, targeting peripheral cholinergic motoneurons leading to flaccid and spastic paralysis, respectively, and successive respiratory failure. Complex polysialo gangliosides accumulate BoNT and TeNT on the plasma membrane. The ganglioside binding in BoNT/A, B, E, F, G, and TeNT occurs via a conserved ganglioside-binding pocket within the most carboxyl-terminal 25 kDa domain HCC, whereas BoNT/C, DC, and D display here two different ganglioside binding sites. This enrichment step facilitates subsequent binding of BoNT/A, B, DC, D, E, F, and G to the intraluminal domains of the synaptic vesicle glycoprotein 2 (SV2) isoforms A-C and synaptotagmin-I/-II, respectively. Whereas an induced α-helical 20-mer Syt peptide binds via side chain interactions to the tip of the HCC domain of BoNT/B, DC and G, the preexisting, quadrilateral β-sheet helix of SV2C-LD4 binds the clinically most relevant serotype BoNT/A mainly through backbone-backbone interactions at the interface of HCC and HCN. In addition, the conserved, complex N559-glycan branch of SV2C establishes extensive interactions with BoNT/A resulting in delayed dissociation providing BoNT/A more time for endocytosis into synaptic vesicles. An analogous interaction occurs between SV2A/B and BoNT/E. Altogether, the nature of BoNT-SV2 recognition clearly differs from BoNT-Syt. Subsequently, the synaptic vesicle is recycled and the bound neurotoxin is endocytosed. Acidification of the vesicle lumen triggers membrane insertion of the translocation domain, pore formation, and finally translocation of the enzymatically active light chain into the neuronal cytosol to halt release of neurotransmitters.
Collapse
Affiliation(s)
- Andreas Rummel
- Institut Für Toxikologie, Medizinische Hochschule Hannover, 30623, Hannover, Germany.
| |
Collapse
|
46
|
Blaum BS, Frank M, Walker RC, Neu U, Stehle T. Complement Factor H and Simian Virus 40 bind the GM1 ganglioside in distinct conformations. Glycobiology 2015; 26:532-9. [PMID: 26715202 DOI: 10.1093/glycob/cwv170] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/22/2015] [Indexed: 11/13/2022] Open
Abstract
Mammalian cell surfaces are decorated with a variety of glycan chains that orchestrate development and defense and are exploited by pathogens for cellular attachment and entry. While glycosidic linkages are, in principle, flexible, the conformational space that a given glycan can sample is subject to spatial and electrostatic restrictions imposed by its overall chemical structure. Here, we show how the glycan moiety of the GM1 ganglioside, a branched, monosialylated pentasaccharide that serves as a ligand for various proteins, undergoes differential conformational selection in its interactions with different lectins. Using STD NMR and X-ray crystallography, we found that the innate immune regulator complement Factor H (FH) binds a previously not reported GM1 conformation that is not compatible with the GM1-binding sites of other structurally characterized GM1-binding lectins such as the Simian Virus 40 (SV40) capsid. Molecular dynamics simulations of the free glycan in explicit solvent on the 10 μs timescale reveal that the FH-bound conformation nevertheless corresponds to a minimum in the Gibbs free energy plot. In contrast to the GM1 conformation recognized by SV40, the FH-bound GM1 conformation is associated with poor NOE restraints, explaining how it escaped(1)H-(1)H NOE-restrained modeling in the past and highlighting the necessity for ensemble representations of glycan structures.
Collapse
Affiliation(s)
- Bärbel S Blaum
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen 72076, Germany
| | - Martin Frank
- Biognos AB, Generatorsgatan 1, Gothenburg 41705, Sweden
| | - Ross C Walker
- San Diego Supercomputer Center and Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ursula Neu
- Max-Planck-Institute of Colloids and Interfaces, Potsdam 14476, Germany
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen 72076, Germany Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| |
Collapse
|
47
|
Membrane-Binding Mechanism of Clostridium perfringens Alpha-Toxin. Toxins (Basel) 2015; 7:5268-75. [PMID: 26633512 PMCID: PMC4690130 DOI: 10.3390/toxins7124880] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/17/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022] Open
Abstract
Clostridium perfringens alpha-toxin is a key mediator of gas gangrene, which is a life-threatening infection that manifests as fever, pain, edema, myonecrosis, and gas production. Alpha-toxin possesses phospholipase C and sphingomyelinase activities. The toxin is composed of an N-terminal domain (1-250 aa, N-domain), which is the catalytic site, and a C-terminal domain (251-370 aa, C-domain), which is the membrane-binding site. Immunization of mice with the C-domain of alpha-toxin prevents the gas gangrene caused by C. perfringens, whereas immunization with the N-domain has no effect. The central loop domain (55-93 aa), especially H….SW(84)Y(85)….G, plays an important role in the interaction with ganglioside GM1a. The toxin binds to lipid rafts in the presence of a GM1a/TrkA complex, and metabolites from phosphatidylcholine to diacylglycerol through the enzymatic activity of alpha-toxin itself. These membrane dynamics leads to the activation of endogenous PLCγ-1 via TrkA. In addition, treatment with alpha-toxin leads to the formation of diacylglycerol at membrane rafts in ganglioside-deficient DonQ cells; this in turn triggers endocytosis and cell death. This article summarizes the current the membrane-binding mechanism of alpha-toxin in detail.
Collapse
|
48
|
Rummel A. The long journey of botulinum neurotoxins into the synapse. Toxicon 2015; 107:9-24. [PMID: 26363288 DOI: 10.1016/j.toxicon.2015.09.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 08/27/2015] [Accepted: 09/08/2015] [Indexed: 01/09/2023]
Abstract
Botulinum neurotoxins (BoNT) cause the disease botulism, a flaccid paralysis of the muscle. They are also very effective, widely used medicines applied locally in sub-nanogram quantities. BoNTs are released together with several non-toxic, associated proteins as progenitor toxin complexes (PCT) by Clostridium botulinum to become highly potent oral poisons ingested via contaminated food. They block the neurotransmission in susceptible animals and humans already in nanogram quantities due to their specific ability to enter motoneurons and to cleave only selected neuronal proteins involved in neuroexocytosis. BoNTs have developed a sophisticated strategy to passage the gastrointestinal tract and to be absorbed in the intestine of the host to finally attack neurons. A non-toxic non-hemagglutinin (NTNHA) forms a binary complex with BoNT to protect it from gastrointestinal degradation. This binary M-PTC is one component of the bi-modular 14-subunit ∼760 kDa large progenitor toxin complex. The other component is the structurally and functionally independent dodecameric hemagglutinin (HA) complex which facilitates the absorption on the intestinal epithelium by glycan binding. Subsequent to its transcytosis the HA complex disrupts the tight junction of the intestinal barrier from the basolateral side by binding to E-cadherin. Now, the L-PTC can also enter the circulation by paracellular routes in much larger quantities. From here, the dissociated BoNTs reach the neuromuscular junction and accumulate via interaction with polysialo gangliosides, complex glycolipids, on motoneurons at the neuromuscular junction. Subsequently, additional specific binding to luminal segments of synaptic vesicles proteins like SV2 and synaptotagmin leads to their uptake. Finally, the neurotoxins shut down the synaptic vesicle cycle, which they had exploited before to enter their target cells, via specific cleavage of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins, which constitute the core components of the cellular membrane fusion machinery.
Collapse
Affiliation(s)
- Andreas Rummel
- Institut für Toxikologie, Medizinische Hochschule Hannover, 30623 Hannover, Germany.
| |
Collapse
|
49
|
Monoclonal Antibodies Targeting the Alpha-Exosite of Botulinum Neurotoxin Serotype/A Inhibit Catalytic Activity. PLoS One 2015; 10:e0135306. [PMID: 26275214 PMCID: PMC4537209 DOI: 10.1371/journal.pone.0135306] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 07/20/2015] [Indexed: 12/12/2022] Open
Abstract
The paralytic disease botulism is caused by botulinum neurotoxins (BoNT), multi-domain proteins containing a zinc endopeptidase that cleaves the cognate SNARE protein, thereby blocking acetylcholine neurotransmitter release. Antitoxins currently used to treat botulism neutralize circulating BoNT but cannot enter, bind to or neutralize BoNT that has already entered the neuron. The light chain endopeptidase domain (LC) of BoNT serotype A (BoNT/A) was targeted for generation of monoclonal antibodies (mAbs) that could reverse paralysis resulting from intoxication by BoNT/A. Single-chain variable fragment (scFv) libraries from immunized humans and mice were displayed on the surface of yeast, and 19 BoNT/A LC-specific mAbs were isolated by using fluorescence-activated cell sorting (FACS). Affinities of the mAbs for BoNT/A LC ranged from a KD value of 9.0×10−11 M to 3.53×10−8 M (mean KD 5.38×10−9 M and median KD 1.53×10−9 M), as determined by flow cytometry analysis. Eleven mAbs inhibited BoNT/A LC catalytic activity with IC50 values ranging from 8.3 ~73×10−9 M. The fine epitopes of selected mAbs were also mapped by alanine-scanning mutagenesis, revealing that the inhibitory mAbs bound the α-exosite region remote from the BoNT/A LC catalytic center. The results provide mAbs that could prove useful for intracellular reversal of paralysis post-intoxication and further define epitopes that could be targeted by small molecule inhibitors.
Collapse
|
50
|
Benoit RM, Frey D, Wieser MM, Thieltges KM, Jaussi R, Capitani G, Kammerer RA. Structure of the BoNT/A1--receptor complex. Toxicon 2015; 107:25-31. [PMID: 26260692 DOI: 10.1016/j.toxicon.2015.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 08/04/2015] [Indexed: 11/16/2022]
Abstract
Botulinum neurotoxin A causes botulism but is also used for medical and cosmetic applications. A detailed molecular understanding of BoNT/A--host receptor interactions is therefore fundamental for improving current clinical applications and for developing new medical strategies targeting human disorders. Towards this end, we recently solved an X-ray crystal structure of BoNT/A1 in complex with its neuronal protein receptor SV2C. Based on our findings, we discuss the potential implications for BoNT/A function.
Collapse
Affiliation(s)
- Roger M Benoit
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Daniel Frey
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Mara M Wieser
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Katherine M Thieltges
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Rolf Jaussi
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Guido Capitani
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Richard A Kammerer
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland.
| |
Collapse
|