Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 484
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Biochem ; 88: 811-837, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30388027

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.


Subject(s)
Botulinum Toxins/therapeutic use , Metalloendopeptidases/therapeutic use , Tetanus Toxin/therapeutic use , Animals , Botulinum Toxins/metabolism , Botulinum Toxins/toxicity , Humans , Metalloendopeptidases/metabolism , Metalloendopeptidases/toxicity , Protein Conformation , Protein Engineering , Tetanus Toxin/metabolism , Tetanus Toxin/toxicity
2.
EMBO J ; 43(16): 3358-3387, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977849

ABSTRACT

Tetanus neurotoxin (TeNT) causes spastic paralysis by inhibiting neurotransmission in spinal inhibitory interneurons. TeNT binds to the neuromuscular junction, leading to its internalisation into motor neurons and subsequent transcytosis into interneurons. While the extracellular matrix proteins nidogens are essential for TeNT binding, the molecular composition of its receptor complex remains unclear. Here, we show that the receptor-type protein tyrosine phosphatases LAR and PTPRδ interact with the nidogen-TeNT complex, enabling its neuronal uptake. Binding of LAR and PTPRδ to the toxin complex is mediated by their immunoglobulin and fibronectin III domains, which we harnessed to inhibit TeNT entry into motor neurons and protect mice from TeNT-induced paralysis. This function of LAR is independent of its role in regulating TrkB receptor activity, which augments axonal transport of TeNT. These findings reveal a multi-subunit receptor complex for TeNT and demonstrate a novel trafficking route for extracellular matrix proteins. Our study offers potential new avenues for developing therapeutics to prevent tetanus and dissecting the mechanisms controlling the targeting of physiological ligands to long-distance axonal transport in the nervous system.


Subject(s)
Membrane Glycoproteins , Motor Neurons , Tetanus Toxin , Animals , Mice , Tetanus Toxin/metabolism , Motor Neurons/metabolism , Membrane Glycoproteins/metabolism , Humans , Cell Adhesion Molecules/metabolism , Protein Binding , Receptor, trkB/metabolism , Axonal Transport , Receptor-Like Protein Tyrosine Phosphatases, Class 2
3.
Am J Pathol ; 194(9): 1752-1763, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38885925

ABSTRACT

Local tetanus develops when limited amounts of tetanus neurotoxin (TeNT) are released by Clostridium tetani generated from spores inside a necrotic wound. Within days, a spastic paralysis restricted to the muscles of the affected anatomical area develops. This paralysis follows the retrograde transport of TeNT inside the axons of motoneurons and its uptake by inhibitory interneurons with cleavage of a vesicle-associated membrane protein required for neurotransmitter release. Consequently, incontrollable excitation of motoneurons causes contractures of innervated muscles and leads to local spastic paralysis. Here, the initial events occurring close to the site of TeNT release were investigated in a mouse model of local tetanus. A peripheral flaccid paralysis was found to occur, before or concurrent to the spastic paralysis. At variance from the confined TeNT proteolytic activity taking place within motor neuron terminals, central protein cleavage was detected within inhibitory interneurons controlling motor neuron efferents innervating muscle groups distant from the site of TeNT release. These results indicate peripheral activity of TeNT in tetanus and explains why the spastic paralysis observed in local tetanus, although confined to single limbs, generally affects multiple muscles. The initial TeNT neuroparalytic activity can be detected by measuring the compound muscle action potential, providing a very early diagnosis and therapy, thus preventing the ensuing life-threatening generalized tetanus.


Subject(s)
Neuromuscular Junction , Paralysis , Tetanus Toxin , Tetanus , Animals , Tetanus/metabolism , Tetanus/complications , Tetanus Toxin/metabolism , Mice , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Neuromuscular Junction/drug effects , Paralysis/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Interneurons/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Female
4.
Int J Mol Sci ; 25(11)2024 May 26.
Article in English | MEDLINE | ID: mdl-38891974

ABSTRACT

Tetanus disease, caused by C. tetani, starts with wounds or mucous layer contact. Prevented by vaccination, the lack of booster shots throughout life requires prophylactic treatment in case of accidents. The incidence of tetanus is high in underdeveloped countries, requiring the administration of antitetanus antibodies, usually derived from immunized horses or humans. Heterologous sera represent risks such as serum sickness. Human sera can carry unknown viruses. In the search for human monoclonal antibodies (mAbs) against TeNT (Tetanus Neurotoxin), we previously identified a panel of mAbs derived from B-cell sorting, selecting two nonrelated ones that binded to the C-terminal domain of TeNT (HCR/T), inhibiting its interaction with the cellular receptor ganglioside GT1b. Here, we present the results of cellular assays and molecular docking tools. TeNT internalization in neurons is prevented by more than 50% in neonatal rat spinal cord cells, determined by quantitative analysis of immunofluorescence punctate staining of Alexa Fluor 647 conjugated to TeNT. We also confirmed the mediator role of the Synaptic Vesicle Glycoprotein II (SV2) in TeNT endocytosis. The molecular docking assays to predict potential TeNT epitopes showed the binding of both antibodies to the HCR/T domain. A higher incidence was found between N1153 and W1297 when evaluating candidate residues for conformational epitope.


Subject(s)
Antibodies, Monoclonal , Endocytosis , Molecular Docking Simulation , Neurons , Tetanus Toxin , Animals , Rats , Neurons/metabolism , Humans , Antibodies, Monoclonal/immunology , Tetanus Toxin/immunology , Tetanus Toxin/metabolism , Tetanus/prevention & control , Tetanus/immunology , Epitopes/immunology , Gangliosides/immunology , Gangliosides/metabolism , Cells, Cultured , Computer Simulation , Metalloendopeptidases
5.
Appl Microbiol Biotechnol ; 107(23): 7197-7211, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37741939

ABSTRACT

Tetanus toxin (TeNT) and botulinum neurotoxins (BoNTs) are neuroprotein toxins, with the latter being the most toxic known protein. They are structurally similar and contain three functional domains: an N-terminal catalytic domain (light chain), an internal heavy-chain translocation domain (HN domain), and a C-terminal heavy chain receptor binding domain (Hc domain or RBD). In this study, fusion functional domain molecules consisting of the TeNT RBD (THc) and the BoNT/A RBD (AHc) (i.e., THc-Linker-AHc and AHc-Linker-THc) were designed, prepared, and identified. The interaction of each Hc domain and the ganglioside receptor (GT1b) or the receptor synaptic vesicle glycoprotein 2 (SV2) was explored in vitro. Their immune response characteristics and protective efficacy were investigated in animal models. The recombinant THc-linker-AHc and AHc-linker-THc proteins with the binding activity had the correct size and structure, thus representing novel subunit vaccines. THc-linker-AHc and AHc-linker-THc induced high levels of specific neutralizing antibodies, and showed strong immune protective efficacy against both toxins. The high antibody titers against the two novel fusion domain molecules and against individual THc and AHc suggested that the THc and AHc domains, as antigens in the fusion functional domain molecules, do not interact with each other and retain their full key epitopes responsible for inducing neutralizing antibodies. Thus, the recombinant THc-linker-AHc and AHc-linker-THc molecules are strong and effective bivalent biotoxin vaccines, protecting against two biotoxins simultaneously. Our experimental design will be valuable to develop recombinant double-RBD fusion molecules as potent bivalent subunit vaccines against bio-toxins. KEY POINTS: • Double-RBD fusion molecules from two toxins had the correct structure and activity. • THc-linker-AHc and AHc-linker-THc efficiently protected against both biotoxins. • Such bivalent biotoxin vaccines based on the RBD are a valuable experimental design.


Subject(s)
Botulinum Toxins, Type A , Tetanus Toxin , Animals , Tetanus Toxin/genetics , Tetanus Toxin/metabolism , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/metabolism , Protein Binding , Antibodies, Neutralizing , Vaccines, Subunit/genetics
6.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232926

ABSTRACT

Recent animal experiments suggested that centrally transported botulinum toxin type A (BoNT-A) might reduce an abnormal muscle tone, though with an unknown contribution to the dominant peripheral muscular effect observed clinically. Herein, we examined if late BoNT-A antispastic actions persist due to possible central toxin actions in rats. The early effect of intramuscular (i.m.) BoNT-A (5, 2 and 1 U/kg) on a reversible tetanus toxin (TeNT)-induced calf muscle spasm was examined 7 d post-TeNT and later during recovery from flaccid paralysis (TeNT reinjected on day 49 post-BoNT-A). Lumbar intrathecal (i.t.) BoNT-A-neutralizing antiserum was used to discriminate the transcytosis-dependent central toxin action of 5 U/kg BoNT-A. BoNT-A-truncated synaptosomal-associated protein 25 immunoreactivity was examined in the muscles and spinal cord at day 71 post-BoNT-A. All doses (5, 2 and 1 U/kg) induced similar antispastic actions in the early period (days 1-14) post-BoNT-A. After repeated TeNT, only the higher two doses prevented the muscle spasm and associated locomotor deficit. Central trans-synaptic activity contributed to the late antispastic effect of 5 U/kg BoNT-A. Ongoing BoNT-A enzymatic activity was present in both injected muscle and the spinal cord. These observations suggest that the treatment duration in sustained or intermittent muscular hyperactivity might be maintained by higher doses and combined peripheral and central BoNT-A action.


Subject(s)
Botulinum Toxins, Type A , Animals , Botulinum Toxins, Type A/pharmacology , Muscle Hypertonia/drug therapy , Rats , Spasm/drug therapy , Synaptosomal-Associated Protein 25/metabolism , Tetanus Toxin/metabolism , Tetanus Toxin/pharmacology
7.
Int J Mol Sci ; 23(8)2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35457172

ABSTRACT

Tetanus and Botulinum type B neurotoxins are bacterial metalloproteases that specifically cleave the vesicle-associated membrane protein VAMP at an identical peptide bond, resulting in inhibition of neuroexocytosis. The minute amounts of these neurotoxins commonly used in experimental animals are not detectable, nor is detection of their VAMP substrate sensitive enough. The immune detection of the cleaved substrate is much more sensitive, as we have previously shown for botulinum neurotoxin type A. Here, we describe the production in rabbit of a polyclonal antibody raised versus a peptide encompassing the 13 residues C-terminal with respect to the neurotoxin cleavage site. The antibody was affinity purified and found to recognize, with high specificity and selectivity, the novel N-terminus of VAMP that becomes exposed after cleavage by tetanus toxin and botulinum toxin type B. This antibody recognizes the neoepitope not only in native and denatured VAMP but also in cultured neurons and in neurons in vivo in neurotoxin-treated mice or rats, suggesting the great potential of this novel tool to elucidate tetanus and botulinum B toxin activity in vivo.


Subject(s)
Botulinum Toxins, Type A , Tetanus , Animals , Antibodies/metabolism , Mice , Neurotoxins/metabolism , Peptides/metabolism , Proteolysis , R-SNARE Proteins/chemistry , R-SNARE Proteins/metabolism , Rabbits , Rats , Tetanus Toxin/chemistry , Tetanus Toxin/metabolism
8.
J Neurochem ; 158(6): 1244-1253, 2021 09.
Article in English | MEDLINE | ID: mdl-33629408

ABSTRACT

Tetanus is a deadly but preventable disease caused by a protein neurotoxin produced by Clostridium tetani. Spores of C. tetani may contaminate a necrotic wound and germinate into a vegetative bacterium that releases a toxin, termed tetanus neurotoxin (TeNT). TeNT enters the general circulation, binds to peripheral motor neurons and sensory neurons, and is transported retroaxonally to the spinal cord. It then enters inhibitory interneurons and blocks the release of glycine or GABA causing a spastic paralysis. This review attempts to correlate the metalloprotease activity of TeNT and its trafficking and localization into the vertebrate body to the nature and sequence of appearance of the symptoms of tetanus.


Subject(s)
Brain/metabolism , Peripheral Nerves/metabolism , Spinal Cord/metabolism , Tetanus Toxin/metabolism , Tetanus/metabolism , Animals , Brain/microbiology , Humans , Neurotoxins/antagonists & inhibitors , Neurotoxins/metabolism , Peripheral Nerves/microbiology , Spinal Cord/microbiology , Tetanus/prevention & control , Tetanus Toxin/antagonists & inhibitors , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/metabolism
9.
Synapse ; 75(6): e22193, 2021 06.
Article in English | MEDLINE | ID: mdl-33141999

ABSTRACT

In the aging process, the brain presents biochemical and morphological alterations. The neurons of the limbic system show reduced size dendrites, in addition to the loss of dendritic spines. These disturbances trigger a decrease in motor and cognitive function. Likewise, it is reported that during aging, in the brain, there is a significant decrease in neurotrophic factors, which are essential in promoting the survival and plasticity of neurons. The carboxyl-terminal fragment of the heavy chain of the tetanus toxin (Hc-TeTx) acts similarly to neurotrophic factors, inducing neuroprotection in different models of neuronal damage. The aim here, was to evaluate the effect of Hc-TeTx on the motor processes of elderly mice (18 months old), and its impact on the dendritic morphology and density of dendritic spines of neurons in the limbic system. The morphological analysis in the dendrites was evaluated employing Golgi-Cox staining. Hc-TeTx was administered (0.5 mg/kg) intraperitoneally for three days in 18-month-old mice. Locomotor activity was evaluated in a novel environment 30 days after the last administration of Hc-TeTx. Mice treated with Hc-TeTx showed significant changes in their motor behavior, and an increased dendritic spine density of pyramidal neurons in layers 3 and 5 of the prefrontal cortex in the hippocampus, and medium spiny neurons of the nucleus accumbens (NAcc). In conclusion, the Hc-TeTx improves the plasticity of the brain regions of the limbic system of aged mice. Therefore, it is proposed as a pharmacological alternative to prevent or delay brain damage during aging.


Subject(s)
Neurons , Tetanus Toxin , Animals , Dendrites/metabolism , Hippocampus/metabolism , Limbic System/metabolism , Mice , Motor Activity , Neurons/metabolism , Tetanus Toxin/metabolism , Tetanus Toxin/pharmacology , Tetanus Toxin/therapeutic use
10.
Molecules ; 26(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34208805

ABSTRACT

This article presents experimental evidence and computed molecular models of a potential interaction between receptor domain D5 of TrkB with the carboxyl-terminal domain of tetanus neurotoxin (Hc-TeNT). Computational simulations of a novel small cyclic oligopeptide are designed, synthesized, and tested for possible tetanus neurotoxin-D5 interaction. A hot spot of this protein-protein interaction is identified in analogy to the hitherto known crystal structures of the complex between neurotrophin and D5. Hc-TeNT activates the neurotrophin receptors, as well as its downstream signaling pathways, inducing neuroprotection in different stress cellular models. Based on these premises, we propose the Trk receptor family as potential proteic affinity receptors for TeNT. In vitro, Hc-TeNT binds to a synthetic TrkB-derived peptide and acts similar to an agonist ligand for TrkB, resulting in phosphorylation of the receptor. These properties are weakened by the mutagenesis of three residues of the predicted interaction region in Hc-TeNT. It also competes with Brain-derived neurotrophic factor, a native binder to human TrkB, for the binding to neural membranes, and for uptake in TrkB-positive vesicles. In addition, both molecules are located together In Vivo at neuromuscular junctions and in motor neurons.


Subject(s)
Membrane Glycoproteins/chemistry , Metalloendopeptidases/chemistry , Neuroprotective Agents/chemistry , Oligopeptides/chemistry , Receptor, trkB/chemistry , Tetanus Toxin/chemistry , Animals , Crystallography, X-Ray , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Metalloendopeptidases/metabolism , Metalloendopeptidases/pharmacology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Oligopeptides/metabolism , Oligopeptides/pharmacology , Protein Domains , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Receptor, trkB/pharmacology , Tetanus Toxin/metabolism , Tetanus Toxin/pharmacology
11.
Microb Cell Fact ; 19(1): 185, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33004043

ABSTRACT

BACKGROUND: Bacterial spores displaying heterologous antigens or enzymes have long been proposed as mucosal vaccines, functionalized probiotics or biocatalysts. Two main strategies have been developed to display heterologous molecules on the surface of Bacillus subtilis spores: (i) a recombinant approach, based on the construction of a gene fusion between a gene coding for a coat protein (carrier) and DNA coding for the protein to be displayed, and (ii) a non-recombinant approach, based on the spontaneous and stable adsorption of heterologous molecules on the spore surface. Both systems have advantages and drawbacks and the selection of one or the other depends on the protein to be displayed and on the final use of the activated spore. It has been recently shown that B. subtilis builds structurally and functionally different spores when grown at different temperatures; based on this finding B. subtilis spores prepared at 25, 37 or 42 °C were compared for their efficiency in displaying various model proteins by either the recombinant or the non-recombinant approach. RESULTS: Immune- and fluorescence-based assays were used to analyze the display of several model proteins on spores prepared at 25, 37 or 42 °C. Recombinant spores displayed different amounts of the same fusion protein in response to the temperature of spore production. In spores simultaneously displaying two fusion proteins, each of them was differentially displayed at the various temperatures. The display by the non-recombinant approach was only modestly affected by the temperature of spore production, with spores prepared at 37 or 42 °C slightly more efficient than 25 °C spores in adsorbing at least some of the model proteins tested. CONCLUSION: Our results indicate that the temperature of spore production allows control of the display of heterologous proteins on spores and, therefore, that the spore-display strategy can be optimized for the specific final use of the activated spores by selecting the display approach, the carrier protein and the temperature of spore production.


Subject(s)
Bacillus subtilis/growth & development , Peptide Fragments/metabolism , Recombinant Fusion Proteins/metabolism , Spores, Bacterial/growth & development , Temperature , Tetanus Toxin/metabolism , Adsorption , Bacillus subtilis/genetics , Bacterial Proteins/metabolism , Green Fluorescent Proteins/metabolism , Peptide Fragments/genetics , Recombinant Fusion Proteins/genetics , Tetanus Toxin/genetics
12.
EMBO Rep ; 18(8): 1306-1317, 2017 08.
Article in English | MEDLINE | ID: mdl-28645943

ABSTRACT

The tetanus neurotoxin (TeNT) is a highly potent toxin produced by Clostridium tetani that inhibits neurotransmission of inhibitory interneurons, causing spastic paralysis in the tetanus disease. TeNT differs from the other clostridial neurotoxins by its unique ability to target the central nervous system by retrograde axonal transport. The crystal structure of the tetanus toxin reveals a "closed" domain arrangement stabilised by two disulphide bridges, and the molecular details of the toxin's interaction with its polysaccharide receptor. An integrative analysis combining X-ray crystallography, solution scattering and single particle electron cryo-microscopy reveals pH-mediated domain rearrangements that may give TeNT the ability to adapt to the multiple environments encountered during intoxication, and facilitate binding to distinct receptors.


Subject(s)
Tetanus Toxin/chemistry , Tetanus Toxin/metabolism , Animals , Crystallography, X-Ray , Humans , Hydrogen-Ion Concentration , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Microscopy, Electron , Neurotoxins/chemistry , Neurotoxins/metabolism , Protein Binding , Protein Domains , Synaptic Transmission/drug effects , Tetanus Toxin/pharmacology
13.
Mol Cell Proteomics ; 16(4 suppl 1): S161-S171, 2017 04.
Article in English | MEDLINE | ID: mdl-28179412

ABSTRACT

The human immune system consists of an intricate network of tightly controlled pathways, where proteases are essential instigators and executioners at multiple levels. Invading microbial pathogens also encode proteases that have evolved to manipulate and dysregulate host proteins, including host proteases during the course of disease. The identification of pathogen proteases as well as their substrates and mechanisms of action have empowered significant developments in therapeutics for infectious diseases. Yet for many pathogens, there remains a great deal to be discovered. Recently, proteomic techniques have been developed that can identify proteolytically processed proteins across the proteome. These "degradomics" approaches can identify human substrates of microbial proteases during infection in vivo and expose the molecular-level changes that occur in the human proteome during infection as an operational network to develop hypotheses for further research as well as new therapeutics. This Perspective Article reviews how proteases are utilized during infection by both the human host and invading bacterial pathogens, including archetypal virulence-associated microbial proteases, such as the Clostridia spp. botulinum and tetanus neurotoxins. We highlight the potential knowledge that degradomics studies of host-pathogen interactions would uncover, as well as how degradomics has been successfully applied in similar contexts, including use with a viral protease. We review how microbial proteases have been targeted in current therapeutic approaches and how microbial proteases have shaped and even contributed to human therapeutics beyond infectious disease. Finally, we discuss how, moving forward, degradomics research can greatly contribute to our understanding of how microbial pathogens cause disease in vivo and lead to the identification of novel substrates in vivo, and the development of improved therapeutics to counter these pathogens.


Subject(s)
Communicable Diseases/microbiology , Peptide Hydrolases/metabolism , Proteome/isolation & purification , Proteomics/methods , Clostridium botulinum/physiology , Clostridium tetani/physiology , Communicable Diseases/metabolism , Host-Pathogen Interactions , Humans , Proteolysis , Substrate Specificity , Tetanus Toxin/metabolism
14.
Curr Top Microbiol Immunol ; 406: 1-37, 2017.
Article in English | MEDLINE | ID: mdl-27921176

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 ß-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.


Subject(s)
Botulinum Toxins/metabolism , Endocytosis , Tetanus Toxin/metabolism , Humans , Neurotransmitter Agents/metabolism , Protein Binding , Synaptic Vesicles/metabolism
15.
Curr Top Microbiol Immunol ; 406: 39-78, 2017.
Article in English | MEDLINE | ID: mdl-28879524

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.


Subject(s)
Botulinum Toxins/metabolism , Endocytosis , Neurotoxins/metabolism , Tetanus Toxin/metabolism , Animals , Biological Transport , Botulism/metabolism , Humans
16.
Cell Microbiol ; 19(2)2017 02.
Article in English | MEDLINE | ID: mdl-27404998

ABSTRACT

Botulinum and tetanus neurotoxins are the most toxic substances known and form the growing family of clostridial neurotoxins. They are composed of a metalloprotease light chain (L), linked via a disulfide bond to a heavy chain (H). H mediates the binding to nerve terminals and the membrane translocation of L into the cytosol where their substrates, the three SNARE proteins, are localised. L translocation is accompanied by unfolding, and it has to be reduced and reacquire the native fold to exert its neurotoxicity. The Thioredoxin reductase-Thioredoxin system is responsible for the reduction, but it is unknown whether the refolding of L is spontaneous or aided by host chaperones. Here we report that geldanamycin, a specific inhibitor of heat shock protein 90, hampers the refolding of L after membrane translocation and completely prevents the cleavage of SNAREs. We also found that geldanamycin strongly synergises with PX-12, an inhibitor of thioredoxin, suggesting that the processes of L chain refolding and interchain disulfide reduction are strictly coupled. Indeed we found that the heat shock protein 90 and the Thioredoxin reductase-Thioredoxin system physically interact on synaptic vesicle where they orchestrate a chaperone-redox machinery which is exploited by clostridial neurotoxins to deliver their catalytic part into the cytosol.


Subject(s)
Cytosol/metabolism , HSP90 Heat-Shock Proteins/metabolism , Protein Folding , Tetanus Toxin/metabolism , Protein Transport , Proteolysis , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism
17.
Nature ; 487(7406): 235-8, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22722837

ABSTRACT

It is generally accepted that the direct connection from the motor cortex to spinal motor neurons is responsible for dexterous hand movements in primates. However, the role of the 'phylogenetically older' indirect pathways from the motor cortex to motor neurons, mediated by spinal interneurons, remains elusive. Here we used a novel double-infection technique to interrupt the transmission through the propriospinal neurons (PNs), which act as a relay of the indirect pathway in macaque monkeys (Macaca fuscata and Macaca mulatta). The PNs were double infected by injection of a highly efficient retrograde gene-transfer vector into their target area and subsequent injection of adeno-associated viral vector at the location of cell somata. This method enabled reversible expression of green fluorescent protein (GFP)-tagged tetanus neurotoxin, thereby permitting the selective and temporal blockade of the motor cortex­PN­motor neuron pathway. This treatment impaired reach and grasp movements, revealing a critical role for the PN-mediated pathway in the control of hand dexterity. Anti-GFP immunohistochemistry visualized the cell bodies and axonal trajectories of the blocked PNs, which confirmed their anatomical connection to motor neurons. This pathway-selective and reversible technique for blocking neural transmission does not depend on cell-specific promoters or transgenic techniques, and is a new and powerful tool for functional dissection in system-level neuroscience studies.


Subject(s)
Hand/physiology , Motor Neurons/physiology , Neurosciences , Animals , Dependovirus/genetics , Green Fluorescent Proteins/metabolism , Macaca , Metalloendopeptidases/metabolism , Motor Cortex/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Tetanus Toxin/metabolism
18.
Biochim Biophys Acta ; 1858(3): 467-74, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26307528

ABSTRACT

Tetanus and botulinum neurotoxins are produced by anaerobic bacteria of the genus Clostridium and are the most poisonous toxins known, with 50% mouse lethal dose comprised within the range of 0.1-few nanograms per Kg, depending on the individual toxin. Botulinum neurotoxins are similarly toxic to humans and can therefore be considered for potential use in bioterrorism. At the same time, their neurospecificity and reversibility of action make them excellent therapeutics for a growing and heterogeneous number of human diseases that are characterized by a hyperactivity of peripheral nerve terminals. The complete crystallographic structure is available for some botulinum toxins, and reveals that they consist of four domains functionally related to the four steps of their mechanism of neuron intoxication: 1) binding to specific receptors of the presynaptic membrane; 2) internalization via endocytic vesicles; 3) translocation across the membrane of endocytic vesicles into the neuronal cytosol; 4) catalytic activity of the enzymatic moiety directed towards the SNARE proteins. Despite the many advances in understanding the structure-mechanism relationship of tetanus and botulinum neurotoxins, the molecular events involved in the translocation step have been only partially elucidated. Here we will review recent advances that have provided relevant insights on the process and discuss possible models that can be experimentally tested. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.


Subject(s)
Botulinum Toxins/metabolism , Cell Membrane/metabolism , Endocytosis , Presynaptic Terminals/metabolism , SNARE Proteins/metabolism , Tetanus Toxin/metabolism , Animals , Botulinum Toxins/chemistry , Cell Membrane/chemistry , Humans , Hydrogen-Ion Concentration , Mice , Presynaptic Terminals/chemistry , Protein Transport , SNARE Proteins/chemistry , Structure-Activity Relationship , Tetanus Toxin/chemistry
19.
Proc Natl Acad Sci U S A ; 111(32): E3343-52, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071179

ABSTRACT

Glial cells are an integral part of functional communication in the brain. Here we show that astrocytes contribute to the fast dynamics of neural circuits that underlie normal cognitive behaviors. In particular, we found that the selective expression of tetanus neurotoxin (TeNT) in astrocytes significantly reduced the duration of carbachol-induced gamma oscillations in hippocampal slices. These data prompted us to develop a novel transgenic mouse model, specifically with inducible tetanus toxin expression in astrocytes. In this in vivo model, we found evidence of a marked decrease in electroencephalographic (EEG) power in the gamma frequency range in awake-behaving mice, whereas neuronal synaptic activity remained intact. The reduction in cortical gamma oscillations was accompanied by impaired behavioral performance in the novel object recognition test, whereas other forms of memory, including working memory and fear conditioning, remained unchanged. These results support a key role for gamma oscillations in recognition memory. Both EEG alterations and behavioral deficits in novel object recognition were reversed by suppression of tetanus toxin expression. These data reveal an unexpected role for astrocytes as essential contributors to information processing and cognitive behavior.


Subject(s)
Astrocytes/physiology , Recognition, Psychology/physiology , Animals , Astrocytes/drug effects , Brain Waves/drug effects , Brain Waves/physiology , Calcium Signaling , Carbachol/pharmacology , Electroencephalography , Gene Expression , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Nerve Net/cytology , Nerve Net/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission , Tetanus Toxin/genetics , Tetanus Toxin/metabolism , Tissue Culture Techniques , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
20.
J Biol Chem ; 289(32): 22450-8, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24973217

ABSTRACT

Tetanus neurotoxin (TeNT) causes neuroparalytic disease by entering the neuronal soma to block the release of neurotransmitters. However, the mechanism by which TeNT translocates its enzymatic domain (light chain) across endosomal membranes remains unclear. We found that TeNT and a truncated protein devoid of the receptor binding domain (TeNT-LHN) associated with membranes enriched in acidic phospholipids in a pH-dependent manner. Thus, in contrast to diphtheria toxin, the formation of a membrane-competent state of TeNT requires the membrane interface and is modulated by the bilayer composition. Channel formation is further enhanced by tethering of TeNT to the membrane through ganglioside co-receptors prior to acidification. Thus, TeNT channel formation can be resolved into two sequential steps: 1) interaction of the receptor binding domain (heavy chain receptor binding domain) with ganglioside co-receptors orients the translocation domain (heavy chain translocation domain) as the lumen of the endosome is acidified and 2) low pH, in conjunction with acidic lipids within the membrane drives the conformational changes in TeNT necessary for channel formation.


Subject(s)
Metalloendopeptidases/metabolism , Metalloendopeptidases/toxicity , Neurons/drug effects , Neurons/metabolism , Tetanus Toxin/metabolism , Tetanus Toxin/toxicity , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Gangliosides/metabolism , Hydrogen-Ion Concentration , Ion Channels/drug effects , Ion Channels/metabolism , Liposomes/metabolism , Metalloendopeptidases/genetics , Models, Neurological , Mutagenesis, Site-Directed , Potassium/metabolism , Protein Structure, Secondary , Rats , Receptors, Cell Surface/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Tetanus Toxin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL