Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 12(8): e0182909, 2017.
Article in English | MEDLINE | ID: mdl-28800585

ABSTRACT

Clostridium tetani and Clostridium botulinum produce two of the most potent neurotoxins known, tetanus neurotoxin and botulinum neurotoxin, respectively. Extensive biochemical and genetic investigation has been devoted to identifying and characterizing various C. botulinum strains. Less effort has been focused on studying C. tetani likely because recently sequenced strains of C. tetani show much less genetic diversity than C. botulinum strains and because widespread vaccination efforts have reduced the public health threat from tetanus. Our aim was to acquire genomic data on the U.S. vaccine strain of C. tetani to better understand its genetic relationship to previously published genomic data from European vaccine strains. We performed high throughput genomic sequence analysis on two wild-type and two vaccine C. tetani strains. Comparative genomic analysis was performed using these and previously published genomic data for seven other C. tetani strains. Our analysis focused on single nucleotide polymorphisms (SNP) and four distinct constituents of the mobile genome (mobilome): a hypervariable flagellar glycosylation island region, five conserved bacteriophage insertion regions, variations in three CRISPR (clustered regularly interspaced short palindromic repeats)-Cas (CRISPR-associated) systems, and a single plasmid. Intact type IA and IB CRISPR/Cas systems were within 10 of 11 strains. A type IIIA CRISPR/Cas system was present in two strains. Phage infection histories derived from CRISPR-Cas sequences indicate C. tetani encounters phages common among commensal gut bacteria and soil-borne organisms consistent with C. tetani distribution in nature. All vaccine strains form a clade distinct from currently sequenced wild type strains when considering variations in these mobile elements. SNP, flagellar glycosylation island, prophage content and CRISPR/Cas phylogenic histories provide tentative evidence suggesting vaccine and wild type strains share a common ancestor.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Clostridium tetani/genetics , Genome, Bacterial , Phylogeny , Polymorphism, Single Nucleotide , Bacteriophages/genetics , Base Sequence , Chromosome Mapping , Clostridium tetani/classification , Clostridium tetani/pathogenicity , Genomic Islands , Glycosylation , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Plasmids/chemistry , Plasmids/metabolism , Sequence Analysis, DNA , Tetanus Toxin/biosynthesis , Tetanus Toxin/genetics , Tetanus Toxoid/genetics
2.
Clin Vaccine Immunol ; 15(9): 1374-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18667637

ABSTRACT

Antigenicities of several formalin-detoxified botulinum neurotoxin preparations were measured by inhibition and sandwich enzyme-linked immunosorbent assay (ELISA), and immunogenicity was studied in mice. The toxoids were derived primarily from the serotype A 150-kDa neurotoxin protein, while one toxoid was derived from the naturally occurring 900-kDa toxin-hemagglutinin complex. Antigenicity was severely compromised in two commercially available toxoids. A variety of new toxoids were synthesized in-house by optimizing formaldehyde reaction conditions. Three of the resulting toxoids were found to be antigenically identical to the native toxin, as measured by inhibition ELISA, in spite of showing a reduction of toxicity by more than 100,000-fold. Sandwich ELISAs indicated that the in-house toxoids were two- to threefold less antigenic than the neurotoxin compared to commercial toxoids, which were about 100-fold less antigenic. Mice were immunized twice, on day 0 and day 14. By day 28, relatively high toxin-specific immunoglobulin G (IgG) titers were detected in animals that had received any of the in-house toxoids, with greater than 99% being IgG1 and the remainder being IgG2. These immunized mice remained asymptomatic after being challenged with 50 to 1,000,000 50% lethal dose (LD(50)) units of the 900-kDa neurotoxin. In contrast, animals immunized with several different batches of commercially available toxoids did not develop measurable toxin-specific antibody titers. However, these mice survived neurotoxin challenges with 2 LD(50) units but died when challenged with 6 LD(50) units. Neutralizing titers measured from pools of sera generated with the in-house toxoid preparations ranged from 2.5 to 5 U/ml. In terms of predicting immunogenicity, inhibition ELISAs comparing each formalin toxoid to the parent toxin provided good insight for screening the new toxoids as well as for estimating their relative in vivo potencies. Inhibition ELISA data indicate that those toxoids that most closely resemble the native toxin are highly immunogenic and protective. The superior quality of these new toxoids makes them useful tools for continued use in ELISA development and for antitoxin production.


Subject(s)
Botulinum Toxins/immunology , Fixatives/pharmacology , Formaldehyde/pharmacology , Toxoids/immunology , Animals , Botulinum Antitoxin/blood , Botulinum Toxins/toxicity , Botulism/prevention & control , Enzyme-Linked Immunosorbent Assay , Immunization, Secondary , Immunoglobulin G/blood , Male , Mice , Neutralization Tests , Survival Analysis , Toxoids/toxicity
3.
Infect Immun ; 74(10): 5617-24, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16988237

ABSTRACT

Levels of botulinum neurotoxin (BoNT) proteolytic activity were compared using a cell-free assay and living neurons to measure extracellular and intracellular enzymatic activity. Within the cell-free reaction model, BoNT serotypes A and E (BoNT/A and BoNT/E, respectively) were reversibly inhibited by chelating Zn2+ with N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN). BoNT/E required relatively long incubation with TPEN to achieve total inhibition, whereas BoNT/A was inhibited immediately upon mixing. When naïve Zn2+-containing BoNTs were applied to cultured neurons, the cellular action of each BoNT was rapidly inhibited by subsequent addition of TPEN, which is membrane permeable. Excess Zn2+ added to the culture medium several hours after poisoning fully restored intracellular toxin activity. Unlike TPEN, EDTA irreversibly inhibited both BoNT/A and -E within the cell-free in vitro reaction. Excess Zn2+ did not reactivate the EDTA-treated toxins. However, application of EDTA-treated BoNT/A or -E to cultured neurons demonstrated normal toxin action in terms of both blocking neurotransmission and SNAP-25 proteolysis. Different concentrations of EDTA produced toxin preparations with incrementally reduced in vitro proteolytic activities, which, when applied to living neurons showed undiminished cellular potency. This suggests that EDTA renders the BoNT proteolytic domain conformationally inactive when tested with the cell-free reaction, but this change is corrected during entry into neurons. The effect of EDTA is unrelated to Zn2+ because TPEN could be applied to living cells before or after poisoning to produce rapid and reversible inhibition of both BoNTs. Therefore, bound Zn2+ is not required for toxin entry into neurons, and removal of Zn2+ from cytosolic BoNTs does not irreversibly alter toxin structure or function. We conclude that EDTA directly alters both BoNTs in a manner that is independent of Zn2+.


Subject(s)
Botulinum Toxins, Type A/toxicity , Botulinum Toxins/toxicity , Neurons/drug effects , Neurotoxins/toxicity , Animals , Biological Assay , Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins, Type A/antagonists & inhibitors , Cells, Cultured , Chelating Agents/pharmacology , Edetic Acid/pharmacology , Ethylenediamines/pharmacology , Mice , Mice, Inbred C57BL , Neurons/enzymology , Neurotoxins/antagonists & inhibitors , Peptide Hydrolases/metabolism , Zinc/pharmacology
4.
Biochemistry ; 43(2): 526-32, 2004 Jan 20.
Article in English | MEDLINE | ID: mdl-14717608

ABSTRACT

Botulinum neurotoxins (BoNTs) act within the synaptic terminal to block neurotransmitter release. The toxin enters the neuron by binding to neuronal membrane receptor(s), being taken up into an endosome-like compartment, and penetrating the endosome membrane via a pH-dependent translocation process. Once within the synaptic cytoplasm, BoNT serotypes A and E cleave separate sites on the C-terminus of the neuronal protein SNAP-25, one of the SNARE proteins required for synaptic vesicle fusion. In this study, we measured the effect of brief toxin exposure on SNAP-25 proteolysis in neuronal cell cultures as an indicator of toxin translocation. The results indicate that (1) uptake of both BoNT-A and -E is enhanced with synaptic activity induced by K+ depolarization in the presence of Ca2+ and (2) translocation of BoNT-A from the acidic endosomal compartment is slow relative to that of BoNT-E. Polyclonal antisera against each toxin protect cells when applied with the toxin during stimulation but has no effect when added immediately after toxin exposure, indicating that toxin endocytosis occurs with synaptic activity. Both serotypes cleave SNAP-25 at concentrations between 50 pM and 4 nM. IC50 values for SNAP-25 cleavage are approximately 0.5 nM for both serotypes. Inhibition of the pH-dependent translocation process by pretreating cultures with concanamycin A (Con A) prevents cleavage of SNAP-25 with IC50 values of approximately 25 nM. Addition of Con A at times up to 15 min after toxin exposure abrogated BoNT-A action; however, addition of Con A after 40 min was no longer protective. In contrast, Con A inhibited, but did not prevent, translocation of BoNT-E even when added immediately after toxin exposure, indicating that pH-dependent translocation of BoNT-E is rapid relative to that of BoNT-A. This study demonstrates that uptake of both BoNT-A and -E is enhanced with synaptic activity and that translocation of the toxin catalytic moiety into the cytosol occurs at different rates for these two serotypes.


Subject(s)
Botulinum Toxins, Type A/metabolism , Botulinum Toxins/metabolism , Neurons/metabolism , Animals , Botulinum Antitoxin/pharmacology , Botulinum Toxins/pharmacology , Botulinum Toxins, Type A/pharmacology , Calcium/chemistry , Cells, Cultured , Concanavalin A/pharmacology , Fetus , Hydrolysis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Protein Transport , Spinal Cord/cytology , Spinal Cord/metabolism , Synaptosomal-Associated Protein 25
5.
J Protein Chem ; 22(5): 441-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14690246

ABSTRACT

Botulinum neurotoxin B (BoNT/B) produces muscle paralysis by cleaving synaptobrevin/vesicle-associated membrane protein (VAMP), an 18-kDa membrane-associated protein located on the surface of small synaptic vesicles. A capillary electrophoresis (CE) assay was developed to evaluate inhibitors of the proteolytic activity of BoNT/B with the objective of identifying suitable candidates for treatment of botulism. The assay was based on monitoring the cleavage of a peptide that corresponds to residues 44-94 of human VAMP-2 (V51) following reaction with the catalytic light chain (LC) of BoNT/B. Cleavage of V51 generated peptide fragments of 18 and 33 amino acids by scission of the bond between Q76 and F77. The fragments and parent peptide were clearly resolved by CE, allowing accurate quantification of the BoNT/B LC-mediated reaction rates. The results indicate that CE is suitable for assessing the enzymatic activity of BoNT/B LC.


Subject(s)
Botulinum Toxins/chemistry , Botulinum Toxins/metabolism , Electrophoresis, Capillary/methods , Botulinum Toxins, Type A , Freezing , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Osmolar Concentration , R-SNARE Proteins , Salts/pharmacology , Temperature , Zinc/pharmacology
6.
Neurochem Res ; 28(3-4): 477-82, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675134

ABSTRACT

Previous work had demonstrated that organomercurial-mediated modification of two cysteine residues in the vesicular acetylcholine transporter (VAChT) from Torpedo californica inhibits binding of vesamicol. The cysteines are protected by acetylcholine and vesamicol (Keller et al. 2000. J. Neurochem. 74:1739-1748). Modified "cysteine 1" is accessible to glutathione from the cytoplasmic surface, whereas modified "cysteine 2" is not. Different organomercurials and aqueous environments were used here to characterize diffusion pathway(s) leading to the cysteines. para-Chloromercuriphenylsulfonate modifies VAChT much more slowly than do more hydrophobic p-chloromercuribenzoate and phenylmercury chloride. Permeabilization of vesicles with cholate detergent increases the rate of modification by p-chloromercuriphenylsulfonate. Permeabilization does not affect the ability of glutathione to reverse modification by p-chloromercuriphenylsulfonate. Higher ionic strength causes about four-fold increase in the rate of modification. The results suggest that hydrophobic and electrostatic barriers inhibit modification of Torpedo VAChT by negatively charged organomercurials and glutathione cannot reach cysteine 2 from either side of the membrane.


Subject(s)
Carrier Proteins/chemistry , Cysteine/drug effects , Membrane Transport Proteins , Organomercury Compounds/pharmacology , Torpedo/metabolism , Vesicular Transport Proteins , 4-Chloromercuribenzenesulfonate/pharmacology , Animals , Diffusion , Ions , Osmolar Concentration , Permeability , Phenylmercury Compounds/pharmacology , Sodium Chloride/pharmacology , Synaptic Vesicles/metabolism , Vesicular Acetylcholine Transport Proteins , p-Chloromercuribenzoic Acid/pharmacology
7.
FEBS Lett ; 513(2-3): 163-8, 2002 Feb 27.
Article in English | MEDLINE | ID: mdl-11904143

ABSTRACT

A targeted delivery vehicle (DV) was developed for intracellular transport of emerging botulinum neurotoxin (BoNT) antagonists. The DV consisted of the isolated heavy chain (HC) of BoNT/A coupled to a 10-kDa amino dextran via the heterobifunctional linker 3-(2-pyridylthio)-propionyl hydrazide. The HC served to target BoNT-sensitive cells and promote internalization of the complex, while the dextran served as a platform to deliver model therapeutic molecules to the targeted cells. To determine the ability of this chimeric glycoprotein to enter neurons, dextran and HC were labeled independently with the fluorescent dyes Oregon green 488 and Cy3, respectively. Internalization of DV was monitored in primary cortical cells using laser confocal microscopy. Incubation of cells for 24 h with DV resulted in discrete punctate labeling of both soma and processes. The Cy3 and Oregon green 488 signals were generally co-localized, suggesting that the complex remained in the same intracellular compartment during the initial 24 h. The DV-associated fluorescence was reduced progressively by co-application of increasing concentrations of unlabeled BoNT/A holotoxin. The results suggest that the BoNT/A HC is able to mediate internalization of a coupled dextran, even though the latter bears no resemblance to the BoNT/A light chain (LC). The HC of BoNT/A thus offers promise as a selective carrier to deliver BoNT antagonists to the nerve terminal cytoplasm for inhibiting the proteolytic activity of internalized BoNT/A LC.


Subject(s)
Antitoxins/administration & dosage , Botulinum Toxins, Type A/antagonists & inhibitors , Drug Carriers/metabolism , Animals , Antitoxins/pharmacology , Binding, Competitive , Biological Transport , Cells, Cultured , Clostridium botulinum/chemistry , Coloring Agents/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...