Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Int J Mol Sci ; 24(6)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36982762

ABSTRACT

Botulinum neurotoxin subtype A4 (BoNT/A4) is ~1000-fold less potent than BoNT/A1. This study addresses the basis for low BoNT/A4 potency. Utilizing BoNT/A1-A4 and BoNT/A4-A1 Light Chain-Heavy Chain (LC-HC) chimeras, HC-A4 was responsible for low BoNT/A4 potency. Earlier studies showed BoNT/A1-receptor binding domain (Hcc) bound a ß-strand peptide (556-564) and glycan-N559 within Luminal Domain 4 (LD4) of SV2C, the BoNT/A protein receptor. Relative to BoNT/A1, the Hcc of BoNT/A4 possesses two amino acid variants (D1141 and N1142) within the ß-peptide binding interface and one amino acid variant (R1292) located near the SV2C glycan-N559. Introduction of BoNT/A4 ß-strand peptide variant (D1141 and N1142) into BoNT/A1 reduced toxin potency 30-fold, and additional introduction of the BoNT/A4 glycan-N559 variant (D1141, N1142, and R1292) further reduced toxin potency to approach BoNT/A4. While introduction of BoNT/A1 glycan-N559 variant (G1292) into BoNT/A4 did not alter toxin potency, additional introduction of BoNT/A1 ß-strand peptide variants (G1141, S1142, and G1292) resulted in potency approaching BoNT/A1 potency. Thus, outcomes from these functional and modeling studies indicate that in rodent models, disruption of Hcc -SV2C ß-peptide and -glycan-N559 interactions mediate low BoNT/A4 potency, while in human motor neurons, disruption of Hcc-SV2C ß-peptide alone mediates low BoNT/A4 potency, which link to a species-specific variation at SV2C563.


Subject(s)
Amino Acids , Humans , Protein Binding , Protein Domains
2.
Int J Mol Sci ; 24(5)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36901734

ABSTRACT

Targeting the botulinum neurotoxin light chain (LC) metalloprotease using small-molecule metal chelate inhibitors is a promising approach to counter the effects of the lethal toxin. However, to overcome the pitfalls associated with simple reversible metal chelate inhibitors, it is crucial to investigate alternative scaffolds/strategies. In conjunction with Atomwise Inc., in silico and in vitro screenings were conducted, yielding a number of leads, including a novel 9-hydroxy-4H-pyrido [1,2-a]pyrimidin-4-one (PPO) scaffold. From this structure, an additional series of 43 derivatives were synthesized and tested, resulting in a lead candidate with a Ki of 150 nM in a BoNT/A LC enzyme assay and 17 µM in a motor neuron cell-based assay. These data combined with structure-activity relationship (SAR) analysis and docking led to a bifunctional design strategy, which we termed "catch and anchor" for the covalent inhibition of BoNT/A LC. Kinetic evaluation was conducted on structures prepared from this catch and anchor campaign, providing kinact/Ki values, and rationale for inhibition seen. Covalent modification was validated through additional assays, including an FRET endpoint assay, mass spectrometry, and exhaustive enzyme dialysis. The data presented support the PPO scaffold as a novel candidate for targeted covalent inhibition of BoNT/A LC.


Subject(s)
Clostridium botulinum , Peptide Hydrolases , Renal Dialysis , Structure-Activity Relationship , Metalloproteases , Metals , Protease Inhibitors/pharmacology
3.
Int J Mol Sci ; 23(22)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36430554

ABSTRACT

The huge advances in genomics and molecular biology in the past two decades have made now an exciting time to study bacterial toxins, in particular, the most potent bacterial toxin known to humankind, botulinum neurotoxins (BoNTs) [...].


Subject(s)
Bacterial Toxins , Botulinum Toxins , Neurotoxins/toxicity , Clostridium/genetics , Botulinum Toxins/toxicity , Bacterial Toxins/genetics , Genomics
4.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681775

ABSTRACT

Botulinum neurotoxin serotype A (BoNT/A) is the most potent protein toxin to humans. BoNT/A light chain (LC/A) cleavage of the membrane-bound SNAP-25 has been well-characterized, but how LC/A traffics to the plasma membrane to target SNAP-25 is unknown. Of the eight BoNT/A subtypes (A1-A8), LC/A3 has a unique short duration of action and low potency that correlate to the intracellular steady state of LC/A, where LC/A1 is associated with the plasma membrane and LC/A3 is present in the cytosol. Steady-state and live imaging of LC/A3-A1 chimeras identified a two-step process where the LC/A N terminus bound intracellular vesicles, which facilitated an internal α-helical-rich domain to mediate LC/A plasma membrane association. The propensity of LC/A variants for membrane association correlated with enhanced BoNT/A potency. Understanding the basis for light chain intracellular localization provides insight to mechanisms underlying BoNT/A potency, which can be extended to applications as a human therapy.


Subject(s)
Botulinum Toxins, Type A/metabolism , Cell Membrane/metabolism , Intracellular Membranes/metabolism , Animals , Botulinum Toxins, Type A/pharmacokinetics , Cell Membrane/drug effects , Female , Humans , Intracellular Membranes/drug effects , Mice , Mice, Inbred ICR , Protein Binding , Synaptosomal-Associated Protein 25/metabolism , Tumor Cells, Cultured
5.
J Am Chem Soc ; 139(21): 7264-7272, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28475321

ABSTRACT

Botulinum neurotoxin serotype A (BoNT/A) causes a debilitating and potentially fatal illness known as botulism. The toxin is also a bioterrorism threat, yet no pharmacological antagonist to counteract its effects has reached clinical approval. Existing strategies to negate BoNT/A intoxication have looked to antibodies, peptides, or organic small molecules as potential therapeutics. In this work, a departure from the traditional drug discovery mindset was pursued, in which the enzyme's susceptibility to metal ions was exploited. A screen of a series of metal salts showed marked inhibitory activity of group 11 and 12 metals against the BoNT/A light chain (LC) protease. Enzyme kinetics revealed that copper (I) and (II) cations displayed noncompetitive inhibition of the LC (Ki ≈ 1 µM), while mercury (II) cations were 10-fold more potent. Crystallographic and mutagenesis studies elucidated a key binding interaction between Cys165 on BoNT/A LC and the inhibitory metals. As potential copper prodrugs, ligand-copper complexes were examined in a cell-based model and were found to prevent BoNT/A cleavage of the endogenous protein substrate, SNAP-25, even at low µM concentrations of complexes. Further investigation of the complexes suggested a bioreductive mechanism causing intracellular release of copper, which directly inhibited the BoNT/A protease. In vivo experiments demonstrated that copper (II) dithiocarbamate and bis(thiosemicarbazone) complexes could delay BoNT/A-mediated lethality in a rodent model, indicating their potential for treating the harmful effects of BoNT/A intoxication. Our studies illustrate that metals can be therapeutically viable enzyme inhibitors; moreover, enzymes that share homology with BoNT LCs may be similarly targeted with metals.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Coordination Complexes/pharmacology , Copper/pharmacology , Protease Inhibitors/pharmacology , Thiocarbamates/pharmacology , Thiosemicarbazones/pharmacology , Animals , Botulinum Toxins, Type A/metabolism , Botulinum Toxins, Type A/toxicity , Coordination Complexes/chemistry , Copper/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Induced Pluripotent Stem Cells/drug effects , Kinetics , Mice , Neurons/cytology , Neurons/drug effects , Protease Inhibitors/chemistry , Structure-Activity Relationship , Thiocarbamates/chemistry , Thiosemicarbazones/chemistry
6.
J Infect Dis ; 213(3): 379-85, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26068781

ABSTRACT

Botulism is a potentially fatal paralytic disease caused by the action of botulinum neurotoxin (BoNT) on nerve cells. There are 7 known serotypes (A-G) of BoNT and up to 40 genetic variants. Clostridium botulinum strain IBCA10-7060 was recently reported to produce BoNT serotype B (BoNT/B) and a novel BoNT, designated as BoNT/H. The BoNT gene (bont) sequence of BoNT/H was compared to known bont sequences. Genetic analysis suggested that BoNT/H has a hybrid-like structure containing regions of similarity to the structures of BoNT/A1 and BoNT/F5. This novel BoNT was serologically characterized by the mouse neutralization assay and a neuronal cell-based assay. The toxic effects of this hybrid-like BoNT were completely eliminated by existing serotype A antitoxins, including those contained in multivalent therapeutic antitoxin products that are the mainstay of human botulism treatment.


Subject(s)
Botulinum Antitoxin/pharmacology , Botulinum Toxins/chemistry , Botulinum Toxins/classification , Animals , Biological Assay , Humans , Mice
7.
J Am Chem Soc ; 138(17): 5568-75, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27070533

ABSTRACT

Botulium neurotoxins (BoNTs) are among the most lethal toxins known to man. They are comprised of seven serotypes with BoNT/A being the most deadly; yet, there is no approved therapeutic for their intoxication or one that has even advanced to clinical trials. Botulinum neurotoxicity is ultimately governed through light chain (LC) protease SNARE protein cleavage leading to a loss of neurotransmitter release. Pharmacological attempts to ablate BoNT/A intoxication have sought to either nullify cellular toxin entry or critical biochemical junctions found within its intricate mechanism of action. In these regards, reports have surfaced of nonpeptidic small molecule inhibitors, but few have demonstrated efficacy in neutralizing cellular toxicity, a key prerequisite before rodent lethality studies can be initiated. On the basis of a lead discovered in our BoNT/A cellular assay campaign, we investigated a family of N-hydroxysuccinimide inhibitors grounded upon structure activity relationship (SAR) fundamentals. Molecules stemming from this SAR exercise were theorized to be protease inhibitors. However, this proposition was overturned on the basis of extensive kinetic analysis. Unexpectedly, inhibitor data pointed to thioredoxin reductase (TrxR), an essential component required for BoNT protease translocation. Also unforeseen was the inhibitors' mechanism of action against TrxR, which was found to be brokered through a suicide-mechanism utilizing quinone methide as the inactivating element. This new series of TrxR inhibitors provides an alternative means to negate the etiological agent responsible for BoNT intoxication, the LC protease.


Subject(s)
Botulinum Toxins, Type A/toxicity , Enzyme Inhibitors/pharmacology , Synaptosomal-Associated Protein 25/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Botulinum Toxins, Type A/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Humans , Structure-Activity Relationship , Substrate Specificity
8.
Biochim Biophys Acta ; 1834(12): 2722-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24096023

ABSTRACT

Clostridium botulinum neurotoxins (BoNTs) cause the life-threatening disease botulism through the inhibition of neurotransmitter release by cleaving essential SNARE proteins. There are seven serologically distinctive types of BoNTs and many subtypes within a serotype have been identified. BoNT/A5 is a recently discovered subtype of type A botulinum neurotoxin which possesses a very high degree of sequence similarity and identity to the well-studied A1 subtype. In the present study, we examined the endopeptidase activity of these two BoNT/A subtypes and our results revealed significant differences in substrate binding and cleavage efficiency between subtype A5 and A1. Distinctive hydrolysis efficiency was observed between the two toxins during cleavage of the native substrate SNAP-25 versus a shortened peptide mimic. N-terminal truncation studies demonstrated that a key region of the SNAP-25, including the amino acid residues at 151 through 154 located in the remote binding region of the substrate, contributed to the differential catalytic properties between A1 and A5. Elevated binding affinity of the peptide substrate resulted from including these important residues and enhanced BoNT/A5's hydrolysis efficiency. In addition, mutations of these amino acid residues affect the proteolytic performance of the two toxins in different ways. This study provides a better understanding of the biological activity of these toxins, their performance characteristics in the Endopep-MS assay to detect BoNT in clinical samples and foods, and is useful for the development of peptide substrates.


Subject(s)
Botulinum Toxins, Type A/chemistry , Clostridium botulinum/enzymology , Synaptosomal-Associated Protein 25/chemistry , Botulinum Toxins, Type A/genetics , Catalysis , Hydrolysis , Mutation , Protein Binding , Synaptosomal-Associated Protein 25/genetics
9.
Appl Environ Microbiol ; 80(23): 7415-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239905

ABSTRACT

Clostridium botulinum subtype A4 neurotoxin (BoNT/A4) is naturally expressed in the dual-toxin-producing C. botulinum strain 657Ba at 100× lower titers than BoNT/B. In this study, we describe purification of recombinant BoNT/A4 (rBoNT/A4) expressed in a nonsporulating and nontoxigenic C. botulinum expression host strain. The rBoNT/A4 copurified with nontoxic toxin complex components provided in trans by the expression host and was proteolytically cleaved to the active dichain form. Activity of the recombinant BoNT/A4 in mice and in human neuronal cells was about 1,000-fold lower than that of BoNT/A1, and the recombinant BoNT/A4 was effectively neutralized by botulism heptavalent antitoxin. A previous report using recombinant truncated BoNT/A4 light chain (LC) expressed in Escherichia coli has indicated reduced stability and activity of BoNT/A4 LC compared to BoNT/A1 LC, which was surmounted by introduction of a single-amino-acid substitution, I264R. In order to determine whether this mutation would also affect the holotoxin activity of BoNT/A4, a recombinant full-length BoNT/A4 carrying this mutation as well as a second mutation predicted to increase solubility (L260F) was produced in the clostridial expression system. Comparative analyses of the in vitro, cellular, and in vivo activities of rBoNT/A4 and rBoNT/A4-L260F I264R showed 1,000-fold-lower activity than BoNT/A1 in both the mutated and nonmutated BoNT/A4. This indicates that these mutations do not alter the activity of BoNT/A4 holotoxin. In summary, a recombinant BoNT from a dual-toxin-producing strain was expressed and purified in an endogenous clostridial expression system, allowing analysis of this toxin.


Subject(s)
Botulinum Toxins, Type A/toxicity , Clostridium botulinum/metabolism , Gene Expression , Glycosides/toxicity , Neurons/drug effects , Triterpenes/toxicity , Amino Acid Substitution , Animals , Antitoxins/metabolism , Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/isolation & purification , Cells, Cultured , Clostridium botulinum/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Glycosides/chemistry , Glycosides/genetics , Glycosides/isolation & purification , Humans , Mice , Neutralization Tests , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/toxicity , Triterpenes/chemistry , Triterpenes/isolation & purification
10.
Curr Top Microbiol Immunol ; 364: 257-85, 2013.
Article in English | MEDLINE | ID: mdl-23239357

ABSTRACT

Botulinum neurotoxins (BoNTs) are the most potent human toxins known and the causative agent of botulism, and are widely used as valuable pharmaceuticals. The BoNTs are modular proteins consisting of a heavy chain and a light chain linked by a disulfide bond. Intoxication of neuronal cells by BoNTs is a multi-step process including specific cell binding, endocytosis, conformational change in the endosome, translocation of the enzymatic light chain into the cells cytosol, and SNARE target cleavage. The quantitative and reliable potency determination of fully functional BoNTs produced as active pharmaceutical ingredient (API) requires an assay that considers all steps in the intoxication pathway. The in vivo mouse bioassay has for years been the 'gold standard' assay used for this purpose, but it requires the use of large numbers of mice and thus causes associated costs and ethical concerns. Cell-based assays are currently the only in vitro alternative that detect fully functional BoNTs in a single assay and have been utilized for years for research purposes. Within the last 5 years, several cell-based BoNT detection assays have been developed that are able to quantitatively determine BoNT potency with similar or greater sensitivity than the mouse bioassay. These assays now offer an alternative method for BoNT potency determination. Such quantitative and reliable BoNT potency determination is a crucial step in basic research, in the development of pharmaceutical BoNTs, and in the quantitative detection of neutralizing antibodies.


Subject(s)
Animal Use Alternatives , Biological Assay/methods , Botulinum Toxins/isolation & purification , Botulism/diagnosis , Immunoassay/methods , Neurotoxins/isolation & purification , Animals , Antibodies, Neutralizing/immunology , Botulinum Toxins/chemistry , Botulinum Toxins/immunology , Botulism/immunology , Cell Line, Tumor , Clostridium botulinum/chemistry , Clostridium botulinum/immunology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Humans , Mice , Neurons/chemistry , Neurons/immunology , Neurotoxins/chemistry , Neurotoxins/immunology , Neurotoxins/toxicity , Protein Transport , SNARE Proteins/chemistry
11.
Bioorg Med Chem ; 22(3): 1208-17, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24360826

ABSTRACT

Botulinum neurotoxin A (BoNT/A) is the most potent toxin known. Unfortunately, it is also a potential bioweapon in terrorism, which is without an approved therapeutic treatment once cellular intoxication takes place. Previously, we reported how hydroxamic acid prodrug carbamates increased cellular uptake, which translated to successful inhibition of this neurotoxin. Building upon this research, we detail BoNT/A protease molecular modeling studies accompanied by the construction of small library of hydroxamic acids based on 2,4-dichlorocinnamic hydroxamic acid scaffold and their carbamate prodrug derivatization along with the evaluation of these molecules in both enzymatic and cellular models.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Hydroxamic Acids/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Protease Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Chemistry Techniques, Synthetic , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Models, Molecular , Prodrugs/chemical synthesis , Protease Inhibitors/chemistry , Small Molecule Libraries/chemical synthesis
12.
mBio ; 15(3): e0310623, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38347673

ABSTRACT

Botulinum neurotoxins (BoNTs) are a class of toxins produced by Clostridium botulinum (C. botulinum) and other species of Clostridia. BoNT/X is a putative novel botulinum neurotoxin identified through genome sequencing and capable of SNARE cleavage, but its neurotoxic potential in humans and vertebrates remained unclear. The C. botulinum strain producing BoNT/X, Strain 111, encodes both a plasmid-borne bont/b2 as well as the chromosomal putative bont/x. This study utilized C. botulinum Strain 111 from Japan as well as recombinantly produced full-length BoNT/X to more fully analyze this putative pathogenic toxin. We confirmed production of full-length, catalytically active native BoNT/X by C. botulinum Strain 111, produced as a disulfide-bonded dichain polypeptide similar to other BoNTs. Both the purified native and the recombinant BoNT/X had high enzymatic activity in vitro but displayed very low potency in human-induced pluripotent stem cell-derived neuronal cells and in mice. Intraperitoneal injection of up to 50 µg of native BoNT/X in mice did not result in botulism; however, mild local paralysis was observed after injection of 2 µg into the gastrocnemius muscle. We further demonstrate that the lack of toxicity by BoNT/X is due to inefficient neuronal cell association and entry, which can be rescued by replacing the receptor binding domain of BoNT/X with that of BoNT/A. These data demonstrate that BoNT/X is not a potent vertebrate neurotoxin like the classical seven serotypes of BoNTs. IMPORTANCE: The family of botulinum neurotoxins comprises the most potent toxins known to humankind. New members of this family of protein toxins as well as more distantly related homologs are being identified. The discovery of BoNT/X via bioinformatic screen in 2017 as a putative new BoNT serotype raised concern about its potential as a pathogenic agent with no available countermeasures. This study for the first time assessed both recombinantly produced and native purified BoNT/X for its vertebrate neurotoxicity.


Subject(s)
Botulism , Clostridium botulinum , Humans , Animals , Mice , Neurotoxins/chemistry , Neurotoxins/genetics , Neurotoxins/metabolism , Clostridium botulinum/genetics , Plasmids , Neurons/metabolism
13.
Infect Immun ; 81(10): 3894-902, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23918782

ABSTRACT

Botulinum neurotoxins (BoNTs) are synthesized by Clostridium botulinum and exist as seven immunologically distinct serotypes designated A through G. For most serotypes, several subtypes have now been described based on nominal differences in the amino acid sequences. BoNT/A1 is the most well-characterized subtype of the BoNT/A serotype, and many of its properties, including its potency, its prevalence as a food poison, and its utility as a pharmaceutical, have been thoroughly studied. In contrast, much remains unknown of the other BoNT/A subtypes. In this study, BoNT/A subtype 1 (BoNT/A1) to BoNT/A5 were characterized utilizing a mouse bioassay, an in vitro cleavage assay, and several neuronal cell-based assays. The data indicate that BoNT/A1 to -5 have distinct in vitro and in vivo toxicological properties and that, unlike those for BoNT/A1, the neuronal and mouse results for BoNT/A2 to -5 do not correlate with their enzymatic activity. These results indicate that BoNT/A1 to -5 have distinct characteristics, which are of importance for a greater understanding of botulism and for pharmaceutical applications.


Subject(s)
Botulinum Toxins, Type A/classification , Botulinum Toxins, Type A/toxicity , Neurons/drug effects , Animals , Biological Assay , Cells, Cultured , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/metabolism , Rats
14.
Bioorg Med Chem ; 21(5): 1344-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23340139

ABSTRACT

Botulinum neurotoxins (BoNTs) are the most lethal biotoxins known to mankind and are responsible for the neuroparalytic disease botulism. Current treatments for botulinum poisoning are all protein based and thus have a limited window of treatment opportunity. Inhibition of the BoNT light chain protease (LC) has emerged as a therapeutic strategy for the treatment of botulism as it may provide an effective post exposure remedy. Using a combination of crystallographic and modeling studies a series of hydroxamates derived from 1-adamantylacetohydroxamic acid (3a) were prepared. From this group of compounds, an improved potency of about 17-fold was observed for two derivatives. Detailed mechanistic studies on these structures revealed a competitive inhibition model, with a K(i)=27 nM, which makes these compounds some of the most potent small molecule, non-peptidic BoNT/A LC inhibitors reported to date.


Subject(s)
Adamantane/chemistry , Botulinum Toxins, Type A/antagonists & inhibitors , Hydroxamic Acids/chemistry , Protease Inhibitors/chemical synthesis , Animals , Binding Sites , Botulinum Toxins, Type A/metabolism , Catalytic Domain , Cells, Cultured , Crystallography, X-Ray , Drug Evaluation, Preclinical , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/toxicity , Kinetics , Molecular Docking Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/toxicity , Rats , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Structure-Activity Relationship
15.
Toxins (Basel) ; 15(9)2023 09 08.
Article in English | MEDLINE | ID: mdl-37755989

ABSTRACT

Vaccines are one of the most effective strategies to prevent pathogen-induced illness in humans. The earliest vaccines were based on live inoculations with low doses of live or related pathogens, which carried a relatively high risk of developing the disease they were meant to prevent. The introduction of attenuated and killed pathogens as vaccines dramatically reduced these risks; however, attenuated live vaccines still carry a risk of reversion to a pathogenic strain capable of causing disease. This risk is completely eliminated with recombinant protein or subunit vaccines, which are atoxic and non-infectious. However, these vaccines require adjuvants and often significant optimization to induce robust T-cell responses and long-lasting immune memory. Some pathogens produce protein toxins that cause or contribute to disease. To protect against the effects of such toxins, chemically inactivated toxoid vaccines have been found to be effective. Toxoid vaccines are successfully used today at a global scale to protect against tetanus and diphtheria. Recent developments for toxoid vaccines are investigating the possibilities of utilizing recombinant protein toxins mutated to eliminate biologic activity instead of chemically inactivated toxins. Finally, one of the most contemporary approaches toward vaccine design utilizes messenger RNA (mRNA) as a vaccine candidate. This approach was used globally to protect against coronavirus disease during the COVID-19 pandemic that began in 2019, due to its advantages of quick production and scale-up, and effectiveness in eliciting a neutralizing antibody response. Nonetheless, mRNA vaccines require specialized storage and transport conditions, posing challenges for low- and middle-income countries. Among multiple available technologies for vaccine design and formulation, which technology is most appropriate? This review focuses on the considerable developments that have been made in utilizing diverse vaccine technologies with a focus on vaccines targeting bacterial toxins. We describe how advancements in vaccine technology, combined with a deeper understanding of pathogen-host interactions, offer exciting and promising avenues for the development of new and improved vaccines.


Subject(s)
COVID-19 , Toxins, Biological , Humans , Pandemics , COVID-19/prevention & control , Vaccines, Attenuated , Vaccines, Synthetic , Bacterial Vaccines , Tetanus Toxoid
16.
Microorganisms ; 11(8)2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37630614

ABSTRACT

The Gram stain classifies most bacteria into one of two groups, Gram-negative or Gram-positive, based on the composition of their cell walls [...].

17.
Biochem Biophys Res Commun ; 427(2): 426-30, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23000406

ABSTRACT

Cancerous cell lines have traditionally shown low sensitivity to laboratory or pharmaceutical preparations of botulinum neurotoxin. The work presented here demonstrates that the mouse neuroblastoma/rat glioma hybrid cell line NG108-15 is capable of more sensitively detecting BoNT/A1 than any cell line previously described. This cell line has previously been described to have motor neuron like characteristics, therefore making it a good model to study BoNTs. Differentiation of NG108-15 cells in serum-free medium containing retinoic acid and purmorphamine dramatically increased sensitivity of the neurons to BoNT/A (EC(50) = ~16 LD(50) U). Additional pre-treatment with triasialoganglioside GT1B prior to toxin exposure reduced the EC(50) further to ~11 LD(50) U. Co-culture of the neurons with C2C12 myotubes also significantly increased BoNT/A sensitivity of NG108-15 cells (EC(50) = 26 U) in the absence of differentiation factors.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Cholinergic Neurons/drug effects , Clostridium botulinum , Motor Neurons/drug effects , Neurotoxins/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Coculture Techniques , Gangliosides/pharmacology , Mice , Models, Neurological , Morpholines/pharmacology , Purines/pharmacology , Rats , Tretinoin/pharmacology
18.
Proc Natl Acad Sci U S A ; 106(5): 1330-5, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19164566

ABSTRACT

Clostridium botulinum neurotoxin (BoNT) is the causative agent of botulism, a neuroparalytic disease. We describe here a semisynthetic strategy to identify inhibitors based on toosendanin, a traditional Chinese medicine reported to protect from BoNT intoxication. Using a single molecule assay of BoNT serotypes A and E light chain (LC) translocation through the heavy chain (HC) channel in neurons, we discovered that toosendanin and its tetrahydrofuran analog selectively arrest the LC translocation step of intoxication with subnanomolar potency, and increase the unoccluded HC channel propensity to open with micromolar efficacy. The inhibitory profile on LC translocation is accurately recapitulated in 2 different BoNT intoxication assays, namely the mouse protection and the primary rat spinal cord cell assays. Toosendanin has an unprecedented dual mode of action on the protein-conducting channel acting as a cargo-dependent inhibitor of translocation and as cargo-free channel activator. These results imply that the bimodal modulation by toosendanin depends on the dynamic interactions between channel and cargo, highlighting their tight interplay during the progression of LC transit across endosomes.


Subject(s)
Botulinum Toxins/antagonists & inhibitors , Animals , Botulinum Toxins/metabolism , Cells, Cultured , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Female , Mice , Patch-Clamp Techniques , Protein Transport , Rats , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/physiology
19.
Toxins (Basel) ; 14(12)2022 11 22.
Article in English | MEDLINE | ID: mdl-36548711

ABSTRACT

Botulinum neurotoxin serotype A (BoNT/A) is the most potent protein toxin for humans and is utilized as a therapy for numerous neurologic diseases. BoNT/A comprises a catalytic Light Chain (LC/A) and a Heavy Chain (HC/A) and includes eight subtypes (BoNT/A1-/A8). Previously we showed BoNT/A potency positively correlated with stable localization on the intracellular plasma membrane and identified a low homology domain (amino acids 268-357) responsible for LC/A1 stable co-localization with SNAP-25 on the plasma membrane, while LC/A3 was present in the cytosol of Neuro2A cells. In the present study, steady-state- and live-imaging of a cytosolic LC/A3 derivative (LC/A3V) engineered to contain individual structural elements of the A1 LDH showed that a 59 amino acid region (275-334) termed the MLD was sufficient to direct LC/A3V from the cytosol to the plasma membrane co-localized with SNAP-25. Informatics and experimental validation of the MLD-predicted R1 region (an α-helix, residues 275-300) and R2 region (a loop, α-helix, loop, residues 302-334) both contribute independent steps to the stable co-localization of LC/A1 with SNAP-25 on the plasma membrane of Neuro-2A cells. Understanding how these structural elements contribute to the overall association of LC/A1 on the plasma membrane may identify the molecular basis for the LC contribution of BoNT/A1 to high potency.


Subject(s)
Botulinum Toxins, Type A , Humans , Botulinum Toxins, Type A/metabolism , Cell Membrane/metabolism , Intracellular Membranes , Protein Domains , Catalysis , Neurons/metabolism
20.
Microorganisms ; 10(5)2022 Apr 24.
Article in English | MEDLINE | ID: mdl-35630331

ABSTRACT

Traumatic peripheral nerve injuries tend to be more common in younger, working age populations and can lead to long-lasting disability. Peripheral nerves have an impressive capacity to regenerate; however, successful recovery after injury depends on a number of factors including the mechanism and severity of the trauma, the distance from injury to the reinnervation target, connective tissue sheath integrity, and delay between injury and treatment. Even though modern surgical procedures have greatly improved the success rate, many peripheral nerve injuries still culminate in persistent neuropathic pain and incomplete functional recovery. Recent studies in animals suggest that botulinum neurotoxin A (BoNT/A) can accelerate nerve regeneration and improve functional recovery after injury to peripheral nerves. Possible mechanisms of BoNT/A action include activation or proliferation of support cells (Schwann cells, mast cells, and macrophages), increased angiogenesis, and improvement of blood flow to regenerating nerves.

SELECTION OF CITATIONS
SEARCH DETAIL