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










Publication year range
1.
Anal Biochem ; 622: 114116, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33716126

ABSTRACT

Arabinose 5-phosphate isomerase (API) catalyzes the reversible isomerization of Ribulose 5-phosphate (Ru5P) to Arabinose 5-Phosphate (Ar5P) for the production of 3-deoxy-2-octulosonic acid 8-phosphate (KDO), a component of bacterial lipopolysaccharide (LPS) of gram-negative bacteria. API is an attractive target for therapeutic development against gram-negative bacterial pathogens. The current assay method of API activity utilizes a general reaction for keto sugar determination in a secondary, 3-h color development reaction with 25 N sulfuric acid which poses hazard to both personnel and instrumentation. We therefore aimed to develop a more user friendly assay of the enzyme. Since Ru5P absorbs in the UV region and contains at least 2 chiral centers, it can be expected to display circular dichroism (CD). A wavelength scan revealed indeed Ru5P displays a pronounced negative ellipticity of 30,560 mDeg M-1cm-1 at 279 nm in Tris buffer pH 9.1 but Ar5P does not have any CD. API enzymatic reactions were monitored directly and continuously in real time by following the disappearance of CD from the Ru5P substrate, or by the appearance of CD from Ar5P substrate. The CD signal at this wavelength was not affected by absorption of the enzyme protein or of small molecules, or turbidity of the solution. Common additives in protein and enzyme reaction mixtures such as detergents, metals, and 5% dimethylsulfoxide did not interfere with the CD signal. Assay reactions of 1-3 min consistently yielded reproducible results. Introduction of accessories in a spectropolarimeter will easily adapt this assay to high throughput format for screening thousands of small molecules as inhibitor candidates of API.


Subject(s)
Aldose-Ketose Isomerases/analysis , Circular Dichroism/methods , Enzyme Assays/methods , Bacterial Proteins/metabolism , Catalysis , Francisella tularensis/metabolism , Lipopolysaccharides/metabolism , Pentosephosphates/metabolism , Ribulosephosphates/analysis , Ribulosephosphates/metabolism , Substrate Specificity , Sugar Acids/metabolism , Sugar Phosphates/metabolism
2.
J Microbiol Methods ; 169: 105813, 2020 02.
Article in English | MEDLINE | ID: mdl-31862458

ABSTRACT

The intracellular Gram-negative bacterium, Coxiella burnetii, is a worldwide zoonotic pathogen and the causative agent of Q fever. The standard of care for C. burnetii infections involves extended periods of antibiotic treatment and the development of doxycycline-resistant strains stress the need for new treatment strategies. A previously developed axenic medium has facilitated in vitro growth of the organism. In this study, we have developed a simple culture method that is inexpensive, reliable and utilizes a modular hypoxic chamber system for either small or large scale production of bacteria without the need of a tri-gas incubator. This method provides consistent growth and yields sufficient viable bacteria within four days of culture and can be used for high-throughput screening. The viable bacteria were quantified by counting colony forming units and total bacteria were enumerated using a genomic equivalent method. The characterized bacterial inoculum was then used to optimize cell-based high-throughput immunofluorescence assays with a goal to quantify intracellular bacteria and then screen and identify compounds that inhibit early stages of C. burnetii infection in macrophages.


Subject(s)
Axenic Culture/methods , Coxiella burnetii/growth & development , High-Throughput Screening Assays/methods , Animals , Bacterial Load/methods , Cell Line , Fluorescent Antibody Technique/methods , Mice , Q Fever/microbiology , RAW 264.7 Cells
3.
Toxicon ; 98: 12-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25707753

ABSTRACT

Two small molecular weight inhibitors, compounds CB7969312 and CB7967495, that displayed inhibition of botulinum neurotoxin serotype A in a previous study, were evaluated for inhibition of botulinum neurotoxin serotypes B, C, E, and F. The small molecular weight inhibitors were assessed by molecular modeling, UPLC-based peptide cleavage assay; and an ex vivo assay, the mouse phrenic nerve - hemidiaphragm assay (MPNHDA). While both compounds were inhibitors of botulinum neurotoxin (BoNT) serotypes B, C, and F in the MPNHDA, compound CB7969312 was effective at lower molar concentrations than compound CB7967495. However, compound CB7967495 was significantly more effective at preventing BoNTE intoxication than compound CB7969312. In the UPLC-based peptide cleavage assay, CB7969312 was also more effective against LcC. Both compounds inhibited BoNTE, but not BoNTF, LcE, or LcF in the UPLC-based peptide cleavage assay. Molecular modeling studies predicted that both compounds would be effective inhibitors of BoNTs B, C, E, and F. But CB7967495 was predicted to be a more effective inhibitor of the four serotypes (B, C, E, and F) than CB7969312. This is the first report of a small molecular weight compound that inhibits serotypes B, C, E, and F in the ex vivo assay.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Botulinum Toxins/antagonists & inhibitors , Clostridium botulinum/chemistry , Aminopyridines/chemistry , Animals , Botulinum Toxins/chemistry , Botulinum Toxins, Type A/chemistry , Cells, Cultured , Hydroxyquinolines/chemistry , Mice , Molecular Structure , Molecular Weight , Phrenic Nerve/drug effects , Protein Conformation
4.
PLoS One ; 9(4): e95188, 2014.
Article in English | MEDLINE | ID: mdl-24769566

ABSTRACT

Various substrates, catalysts, and assay methods are currently used to screen inhibitors for their effect on the proteolytic activity of botulinum neurotoxin. As a result, significant variation exists in the reported results. Recently, we found that one source of variation was the use of various catalysts, and have therefore evaluated its three forms. In this paper, we characterize three substrates under near uniform reaction conditions using the most active catalytic form of the toxin. Bovine serum albumin at varying optimum concentrations stimulated enzymatic activity with all three substrates. Sodium chloride had a stimulating effect on the full length synaptosomal-associated protein of 25 kDa (SNAP25) and its 66-mer substrates but had an inhibitory effect on the 17-mer substrate. We found that under optimum conditions, full length SNAP25 was a better substrate than its shorter 66-mer or 17-mer forms both in terms of kcat, Km, and catalytic efficiency kcat/Km. Assay times greater than 15 min introduced large variations and significantly reduced the catalytic efficiency. In addition to characterizing the three substrates, our results identify potential sources of variations in previous published results, and underscore the importance of using well-defined reaction components and assay conditions.


Subject(s)
Botulinum Toxins, Type A/chemistry , Synaptosomal-Associated Protein 25/chemistry , Botulinum Toxins, Type A/antagonists & inhibitors , Chlorides/chemistry , Dithiothreitol/chemistry , Humans , Kinetics , Peptide Fragments/chemistry , Proteolysis , Serum Albumin, Bovine/chemistry , Sodium Chloride/chemistry , Substrate Specificity , Zinc Compounds/chemistry
5.
J Biol Chem ; 288(33): 24223-33, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23779108

ABSTRACT

Botulinum neurotoxins are the most toxic of all compounds. The toxicity is related to a poor zinc endopeptidase activity located in a 50-kDa domain known as light chain (Lc) of the toxin. The C-terminal tail of Lc is not visible in any of the currently available x-ray structures, and it has no known function but undergoes autocatalytic truncations during purification and storage. By synthesizing C-terminal peptides of various lengths, in this study, we have shown that these peptides competitively inhibit the normal catalytic activity of Lc of serotype A (LcA) and have defined the length of the mature LcA to consist of the first 444 residues. Two catalytically inactive mutants also inhibited LcA activity. Our results suggested that the C terminus of LcA might interact at or near its own active site. By using synthetic C-terminal peptides from LcB, LcC1, LcD, LcE, and LcF and their respective substrate peptides, we have shown that the inhibition of activity is specific only for LcA. Although a potent inhibitor with a Ki of 4.5 µm, the largest of our LcA C-terminal peptides stimulated LcA activity when added at near-stoichiometric concentration to three versions of LcA differing in their C-terminal lengths. The result suggested a product removal role of the LcA C terminus. This suggestion is supported by a weak but specific interaction determined by isothermal titration calorimetry between an LcA C-terminal peptide and N-terminal product from a peptide substrate of LcA. Our results also underscore the importance of using a mature LcA as an inhibitor screening target.


Subject(s)
Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/metabolism , Catalytic Domain , Clostridium botulinum/metabolism , Neurotoxins/chemistry , Neurotoxins/metabolism , Amino Acid Sequence , Biocatalysis/drug effects , Botulinum Toxins, Type A/antagonists & inhibitors , Calorimetry , Clostridium botulinum/classification , Enzyme Stability/drug effects , Kinetics , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Binding/drug effects , Protein Structure, Secondary , Protein Unfolding/drug effects , Serotyping , Structure-Activity Relationship , Substrate Specificity/drug effects , Synaptosomal-Associated Protein 25/metabolism , Temperature
6.
J Toxicol ; 2012: 678963, 2012.
Article in English | MEDLINE | ID: mdl-23125855
7.
Int J Biochem Mol Biol ; 3(3): 313-21, 2012.
Article in English | MEDLINE | ID: mdl-23097747

ABSTRACT

The catalytic domain, known as light chain (Lc), of the most poisonous botulinum neurotoxins (BoNTs), possesses endoprotease activity that triggers the ultimate poisonous effect to animals and humans. X-ray crystallographic structure of Lc of several BoNT serotypes has identified at least four small ligands at or near the respective active sites. They are sulfate ions in LcA, LcB, and LcE; an acetate ion in LcA; a calcium ion in LcB; and a potassium ion in LcD. Roles of these ligands on the structure and function of the proteins are not known. We have investigated the roles of sulfate, acetate, and calcium on the catalytic activities of LcA, LcB, and LcE using 17-35-residue synthetic peptide substrates. All three ligands inhibited all Lc activities. For LcA and LcB, the order of inhibition effectiveness was calcium>sulfate>acetate. The inhibition effectiveness expressed as IC(50), did not correlate with the occurrence or proximity of the ions to the active site. Moreover, addition of acetate or sulfate to LcA did not affect the near-UV circular dichroism spectra, tryptophan, and tyrosine fluorescence spectra, and mid points of thermal denaturation of LcA. Our results suggest that acetate, sulfate, and calcium nonspecifically interact with BoNT Lc, and their occurrence in the crystal structures could have been due to opportunistic binding to complementary pockets.

8.
Front Pharmacol ; 3: 102, 2012.
Article in English | MEDLINE | ID: mdl-22675300

ABSTRACT

Botulinum neurotoxins are most potent of all toxins. Their N-terminal light chain domain (Lc) translocates into peripheral cholinergic neurons to exert its endoproteolytic action leading to muscle paralysis. Therapeutic development against these toxins is a major challenge due to their in vitro and in vivo structural differences. Although three-dimensional structures and reaction mechanisms are very similar, the seven serotypes designated A through G vastly vary in their intracellular catalytic stability. To investigate if protein phosphorylation could account for this difference, we employed Src-catalyzed tyrosine phosphorylation of the Lc of six serotypes namely LcA, LcB, LcC1, LcD, LcE, and LcG. Very little phosphorylation was observed with LcD and LcE but LcA, LcB, and LcG were maximally phosphorylated by Src. Phosphorylation of LcA, LcB, and LcG did not affect their secondary and tertiary structures and thermostability significantly. Phosphorylation of Y250 and Y251 made LcA resistant to autocatalysis and drastically reduced its k(cat)/K(m) for catalysis. A tyrosine residue present near the essential cysteine at the C-terminal tail of LcA, LcB, and LcG was readily phosphorylated in vitro. Inclusion of a competitive inhibitor protected Y426 of LcA from phosphorylation, shedding light on the role of the C-terminus in the enzyme's substrate or product binding.

9.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 5): 511-20, 2012 May.
Article in English | MEDLINE | ID: mdl-22525749

ABSTRACT

Clostridium botulinum neurotoxins are classified as Category A bioterrorism agents by the Centers for Disease Control and Prevention (CDC). The seven serotypes (A-G) of the botulinum neurotoxin, the causative agent of the disease botulism, block neurotransmitter release by specifically cleaving one of the three SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins and induce flaccid paralysis. Using a structure-based drug-design approach, a number of peptide inhibitors were designed and their inhibitory activity against botulinum serotype A (BoNT/A) protease was determined. The most potent peptide, RRGF, inhibited BoNT/A protease with an IC(50) of 0.9 µM and a K(i) of 358 nM. High-resolution crystal structures of various peptide inhibitors in complex with the BoNT/A protease domain were also determined. Based on the inhibitory activities and the atomic interactions deduced from the cocrystal structures, the structure-activity relationship was analyzed and a pharmacophore model was developed. Unlike the currently available models, this pharmacophore model is based on a number of enzyme-inhibitor peptide cocrystal structures and improved the existing models significantly, incorporating new features.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Clostridium botulinum/enzymology , Neurotoxins/antagonists & inhibitors , Peptides/chemistry , Peptides/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Botulism/drug therapy , Clostridium botulinum/chemistry , Clostridium botulinum/drug effects , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Structure-Activity Relationship
10.
J Biol Chem ; 286(3): 1802-11, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-20961849

ABSTRACT

Botulinum neurotoxins (BoNT) are the most potent of all toxins that cause flaccid muscle paralysis leading to death. They are also potential biothreat agents. A systematic investigation of various short peptide inhibitors of the BoNT protease domain with a 17-residue peptide substrate led to arginine-arginine-glycine-cysteine having a basic tetrapeptide structure as the most potent inhibitor. When assayed in the presence of dithiothreitol (DTT), the inhibitory effect was drastically reduced. Replacing the terminal cysteine with one hydrophobic residue eliminated the DTT effect but with two hydrophobic residues made the pentapeptide a poor inhibitor. Replacing the first arginine with cysteine or adding an additional cysteine at the N terminus did not improve inhibition. When assessed using mouse brain lysates, the tetrapeptides also inhibited BoNT/A cleavage of the endogenous SNAP-25. The peptides penetrated the neuronal cell lines, N2A and BE(2)-M17, without adversely affecting metabolic functions as measured by ATP production and P-38 phosphorylation. Biological activity of the peptides persisted within cultured chick motor neurons and rat and mouse cerebellar neurons for more than 40 h and inhibited BoNT/A protease action inside the neurons in a dose- and time-dependent fashion. Our results define a tetrapeptide as the smallest peptide inhibitor in the backdrop of a large substrate protein of 200+ amino acids having multiple interaction regions with its cognate enzyme. The inhibitors should also be valuable candidates for drug development.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Oligopeptides/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Animals , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/metabolism , Cell Line , Cerebellum/metabolism , Chickens , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Inbred BALB C , Mutation, Missense , Neurons/metabolism , Oligopeptides/chemistry , Rats , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism
11.
PLoS One ; 5(9): e12872, 2010 Sep 22.
Article in English | MEDLINE | ID: mdl-20877571

ABSTRACT

Botulinum neurotoxins (BoNT) are the most potent of all toxins. The 50 kDa N-terminal endopeptidase catalytic light chain (LC) of BoNT is located next to its central, putative translocation domain. After binding to the peripheral neurons, the central domain of BoNT helps the LC translocate into cytosol where its proteolytic action on SNARE (soluble NSF attachment protein receptor) proteins blocks exocytosis of acetyl choline leading to muscle paralysis and eventual death. The translocation domain also contains 105 Š-long stretch of ∼100 residues, known as "belt," that crosses over and wraps around the LC to shield the active site from solvent. It is not known if the LC gets dissociated from the rest of the molecule in the cytosol before catalysis. To investigate the structural identity of the protease, we prepared four variants of type A BoNT (BoNT/A) LC, and compared their catalytic parameters with those of BoNT/A whole toxin. The four variants were LC + translocation domain, a trypsin-nicked LC + translocation domain, LC + belt, and a free LC. Our results showed that K(m) for a 17-residue SNAP-25 (synaptosomal associated protein of 25 kDa) peptide for these constructs was not very different, but the turnover number (k(cat)) for the free LC was 6-100-fold higher than those of its four variants. Moreover, none of the four variants of the LC was prone to autocatalysis. Our results clearly demonstrated that in vitro, the LC minus the rest of the molecule is the most catalytically active form. The results may have implication as to the identity of the active, toxic moiety of BoNT/A in vivo.


Subject(s)
Botulinum Toxins, Type A/chemistry , Clostridium botulinum/chemistry , Neurotoxins/chemistry , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/metabolism , Catalysis , Clostridium botulinum/genetics , Clostridium botulinum/metabolism , Kinetics , Models, Molecular , Neurotoxins/genetics , Neurotoxins/metabolism , Protein Structure, Tertiary
12.
Anal Biochem ; 396(2): 188-93, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19782037

ABSTRACT

The ultimate molecular action of botulinum neurotoxin (BoNT) is a Zn-dependent endoproteolytic activity on one of the three SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins. There are seven serotypes (A-G) of BoNT having distinct cleavage sites on the SNARE substrates. The proteolytic activity is located on the N-terminal light chain (Lc) domain and is used extensively as the primary target toward therapeutic development against botulism. Here we describe an improved method using ultra-performance liquid chromatography (UPLC) whereby quantitative data were obtained in 1/10th the time using 1/20th the sample and solvent volumes compared with a widely used high-performance liquid chromatography (HPLC) method. We also synthesized a VAMP (vesicle-associated membrane protein)-based peptide containing an intact V1 motif that was efficiently used as a substrate by BoNT/D Lc. Although serotype C1 cleaves the serotype A substrate at a bond separated by only one residue, we were able to distinguish the two reactions by UPLC. The new method can accurately quantify as low as 7 pmol of the peptide substrates for BoNT serotypes A, B, C1, and D. We also report here that the catalytic efficiency of serotype A can be stimulated 35-fold by the addition of Triton X-100 to the reaction mixture. Combining the use of Triton X-100 with the newly introduced UPLC method, we were able to accurately detect very low levels of proteolytic activity in a very short time. Sensitivity of the assay and accuracy and rapidity of product analysis should greatly augment efforts in therapeutic development.


Subject(s)
Botulinum Toxins/metabolism , Chromatography, High Pressure Liquid/methods , Peptides/analysis , Amino Acid Sequence , Botulinum Toxins/classification , Limit of Detection , Molecular Sequence Data , Octoxynol/chemistry , Octoxynol/metabolism , Peptides/metabolism , Recombinant Proteins/classification , Recombinant Proteins/metabolism , Serotyping , Substrate Specificity
13.
J Pharm Sci ; 98(9): 3302-11, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19226630

ABSTRACT

Botulinum neurotoxins (BoNTs) and their fragments are targets of therapeutic developments and are increasingly used as therapeutic, prophylactic, and research reagents. However, published data on their properties vary widely. In order to gain a better understanding of these variations, we initiated a systematic investigation of the stability parameters of catalytic light chains (Lc) as well as of cell surface binding domains (Hc) of the neurotoxin. When followed by CD spectroscopy, we noticed that the recombinant light chains of serotypes A (LcA), B, D, E, and G rapidly lost their secondary structures by mild stirring. Denaturation of LcA increased with stirring speed and temperature resulting in a catalytically inactive precipitate. Reducing agents or an anaerobic environment were ineffective in the denaturation. Under identical conditions, bovine serum albumin, ovalbumin, carboxypeptidase B, and of thermolysin, a structural and functional analogue of LcA, remained unchanged. Hc domains of serotype A, B, C, E, and F were also denatured by mild stirring. Adding the nonionic detergent Tween-20 to LcA completely prevented the denaturation. We speculate that the BoNT domains undergo surface denaturation due to rapid exposure of hydrophobic residues by mechanical agitation. This study has important implications for handling BoNT proteins used in therapeutic development.


Subject(s)
Botulinum Toxins/chemistry , Detergents/chemistry , Protein Denaturation , Protein Stability , Protein Structure, Tertiary , Stress, Mechanical
14.
PLoS Pathog ; 4(9): e1000165, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18818739

ABSTRACT

The seven antigenically distinct serotypes of Clostridium botulinum neurotoxins, the causative agents of botulism, block the neurotransmitter release by specifically cleaving one of the three SNARE proteins and induce flaccid paralysis. The Centers for Disease Control and Prevention (CDC) has declared them as Category A biowarfare agents. The most potent among them, botulinum neurotoxin type A (BoNT/A), cleaves its substrate synaptosome-associated protein of 25 kDa (SNAP-25). An efficient drug for botulism can be developed only with the knowledge of interactions between the substrate and enzyme at the active site. Here, we report the crystal structures of the catalytic domain of BoNT/A with its uncleavable SNAP-25 peptide (197)QRATKM(202) and its variant (197)RRATKM(202) to 1.5 A and 1.6 A, respectively. This is the first time the structure of an uncleavable substrate bound to an active botulinum neurotoxin is reported and it has helped in unequivocally defining S1 to S5' sites. These substrate peptides make interactions with the enzyme predominantly by the residues from 160, 200, 250 and 370 loops. Most notably, the amino nitrogen and carbonyl oxygen of P1 residue (Gln197) chelate the zinc ion and replace the nucleophilic water. The P1'-Arg198, occupies the S1' site formed by Arg363, Thr220, Asp370, Thr215, Ile161, Phe163 and Phe194. The S2' subsite is formed by Arg363, Asn368 and Asp370, while S3' subsite is formed by Tyr251, Leu256, Val258, Tyr366, Phe369 and Asn388. P4'-Lys201 makes hydrogen bond with Gln162. P5'-Met202 binds in the hydrophobic pocket formed by the residues from the 250 and 200 loop. Knowledge of interactions between the enzyme and substrate peptide from these complex structures should form the basis for design of potent inhibitors for this neurotoxin.


Subject(s)
Botulinum Toxins, Type A/chemistry , Drug Design , Binding Sites , Botulinum Toxins, Type A/antagonists & inhibitors , Crystallography, X-Ray , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Substrate Specificity , Synaptosomal-Associated Protein 25/chemistry
15.
J Biol Chem ; 283(27): 18883-91, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18434312

ABSTRACT

The seven antigenically distinct serotypes of Clostridium botulinum neurotoxins cleave specific soluble N-ethylmaleimide-sensitive factor attachment protein receptor complex proteins and block the release of neurotransmitters that cause flaccid paralysis and are considered potential bioweapons. Botulinum neurotoxin type A is the most potent among the clostridial neurotoxins, and to date there is no post-exposure therapeutic intervention available. To develop inhibitors leading to drug design, it is imperative that critical interactions between the enzyme and the substrate near the active site are known. Although enzyme-substrate interactions at exosites away from the active site are mapped in detail for botulinum neurotoxin type A, information about the active site interactions is lacking. Here, we present the crystal structures of botulinum neurotoxin type A catalytic domain in complex with four inhibitory substrate analog tetrapeptides, viz. RRGC, RRGL, RRGI, and RRGM at resolutions of 1.6-1.8 A. These structures show for the first time the interactions between the substrate and enzyme at the active site and delineate residues important for substrate stabilization and catalytic activity. We show that OH of Tyr(366) and NH(2) of Arg(363) are hydrogen-bonded to carbonyl oxygens of P1 and P1' of the substrate analog and position it for catalytic activity. Most importantly, the nucleophilic water is replaced by the amino group of the N-terminal residue of the tetrapeptide. Furthermore, the S1' site is formed by Phe(194), Thr(215), Thr(220), Asp(370), and Arg(363). The K(i) of the best inhibitory tetrapeptide is 157 nm.


Subject(s)
Biological Warfare Agents , Botulinum Toxins, Type A/antagonists & inhibitors , Botulinum Toxins, Type A/chemistry , Clostridium botulinum/enzymology , Enzyme Inhibitors/chemistry , Oligopeptides/chemistry , Binding Sites , Botulinum Toxins, Type A/toxicity , Crystallography, X-Ray , Enzyme Inhibitors/therapeutic use , Oligopeptides/therapeutic use , Paralysis/chemically induced , Paralysis/drug therapy , Paralysis/enzymology , Protein Structure, Tertiary/physiology , Structure-Activity Relationship
16.
Protein J ; 27(3): 151-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18213512

ABSTRACT

Type A botulinum neurotoxin is one of the most lethal of the seven serotypes and is increasingly used as a therapeutic agent in neuromuscular dysfunctions. Its toxic function is related to zinc-endopeptidase activity of the N-terminal light chain (LC) on synaptosome-associated protein-25 kDa (SNAP-25) of the SNARE complex. To understand the determinants of substrate specificity and assist the development of strategies for effective inhibitors, we used site-directed mutagenesis to investigate the effects of 13 polar residues of the LC on substrate binding and catalysis. Selection of the residues for mutation was based on a computational analysis of the three-dimensional structure of the LC modeled with a 17-residue substrate fragment of SNAP-25. Steady-state kinetic parameters for proteolysis of the substrate fragment were determined for a set of 16 single mutants. Of the mutated residues non-conserved among the serotypes, replacement of Arg-230 and Asp-369 by polar or apolar residues resulted in drastic lowering of the catalytic rate constant (k(ca)), but had less effect on substrate affinity (K(m)). Substitution of Arg-230 with Lys decreased the catalytic efficiency (k(cat)/K(m)) by 50-fold, whereas replacement by Leu yielded an inactive protein. Removal of the electrostatic charge at Asp-369 by mutation to Asn resulted in 140-fold decrease in k(cat)/K(m). Replacement of other variable residues surrounding the catalytic cleft (Glu-54, Glu-63, Asn-66, Asp-130, Asn-161, Glu-163, Glu-170, Glu-256), had only marginal effect on decreasing the catalytic efficiency, but unexpectedly the substitution of Lys-165 with Leu resulted in fourfold increase in k(cat)/K(m). For comparison purposes, two conserved residues Arg-362 and Tyr-365 were investigated with substitutions of Leu and Phe, respectively, and their catalytic efficiency decreased 140- and 10-fold, respectively, whereas substitution of the tyrosine ring with Asn abolished activity. The altered catalytic efficiencies of the mutants were not due to any significant changes in secondary or tertiary structures, or in zinc content and thermal stability. We suggest that, despite the large minimal substrate size for catalysis, only a few non-conserved residues surrounding the active site are important to render the LC competent for catalysis or provide conformational selection of the substrate.


Subject(s)
Botulinum Toxins, Type A/chemistry , Clostridium botulinum/chemistry , Neurotoxins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/isolation & purification , Botulinum Toxins, Type A/metabolism , Catalysis , Circular Dichroism , Clostridium botulinum/genetics , Clostridium botulinum/metabolism , Crystallography, X-Ray , Models, Molecular , Neurotoxins/genetics , Neurotoxins/isolation & purification , Neurotoxins/metabolism , Protein Structure, Secondary , Substrate Specificity
17.
Biochem Biophys Res Commun ; 330(1): 97-103, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15781237

ABSTRACT

Botulinum neurotoxins comprise seven distinct serotypes (A-G) produced by Clostridium botulinum. The crystal structure of the binding domain of the botulinum neurotoxin type B (BBHc) has been determined to 2A resolution. The overall structure of BBHc is well ordered and similar to that of the binding domain of the holotoxin. However, significant structural changes occur at what would be the interface of translocation and binding domains of the holotoxin. The loop 911-924 shows a maximum displacement of 14.8A at the farthest point. The N-terminal helix reorients and moves by 19.5A from its original position. BBHc is compared with the binding domain of the holotoxin of botulinum type A and B, and the tetanus C-fragment to characterize the heavy chain-carbohydrate interactions. The probable reasons for different binding affinity of botulinum and tetanus toxins are discussed.


Subject(s)
Botulinum Toxins/chemistry , Clostridium botulinum/chemistry , Botulinum Toxins, Type A , Carbohydrates/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry
18.
Basic Clin Pharmacol Toxicol ; 95(5): 215-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15546475

ABSTRACT

We investigated the inhibitory effect of stinging nettle leaf extract on the protease activity of botulinum neurotoxin type A and B light chains. The nettle leaf infusion was fractionated and HPLC-based enzymatic assays were performed to determine the capacity of each fraction to inhibit the protease activity of botulinum neurotoxin type A and B light chains. Assay results demonstrated that a water-soluble fraction obtained from the nettle leaf infusion inhibited type A, but did not inhibit type B light chain protease activity. The inhibition mode of water soluble fraction against protease activity of type A light chain was analyzed and found to be a non-competitive.


Subject(s)
Botulinum Toxins, Type A/metabolism , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Urtica dioica/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry
19.
Protein J ; 23(7): 445-51, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15635936

ABSTRACT

The light chain of botulinum neurotoxin serotype A undergoes autocatalytic fragmentation into two major peptides during purification and storage (Ahmed S. A. et al. 2001, J. Protein Chem. 20:221-231) by both intermolecular and intramolecular mechanisms (Ahmed S. A. et al. 2003, Biochemistry 42:12539 12549). In this study, we investigated the effects of buffers and salts on this autocatalytic reaction in the presence and absence of zinc chloride. In the presence of zinc chloride, the fragmentation reaction was enhanced in each of acetate, MES, HEPES and phosphate buffers with maximum occurring in acetate when compared to those in the absence of zinc chloride. Adding sodium chloride in phosphate buffer in the presence of zinc chloride increased the extent of proteolysis. Irrespective of the presence of zinc chloride, adding sodium chloride or potassium chloride in phosphate buffer elicited an additional proteolytic reaction. Higher concentrations of sodium phosphate buffer enhanced the autocatalytic reaction in the absence of zinc chloride. In contrast, in the presence of zinc chloride, higher concentrations of sodium phosphate decreased the autocatalytic reaction. Optimum pH of autocatalysis was not affected significantly by the absence or presence of zinc chloride. Like zinc chloride, other chlorides of divalent metals, such as magnesium, cobalt, iron and calcium also enhanced the autocatalytic reaction. Polyols such as ethylene glycol protected the light chain from fragmentation. Exposure of light chain to UV radiation led to enhanced fragmentation. In order to avoid fragmentation, the protein should be stored frozen in a low concentration buffer of neutral or higher pH devoid of any metal. Our results provide a choice of buffers and salts for isolation, purification and storage of intact botulinum neurotoxin serotype A light chain.


Subject(s)
Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/isolation & purification , Metals/chemistry , Catalysis , Endopeptidases/chemistry
20.
Biochemistry ; 42(43): 12539-49, 2003 Nov 04.
Article in English | MEDLINE | ID: mdl-14580200

ABSTRACT

The zinc-endopeptidase light chain of botulinum A neurotoxin undergoes autocatalytic fragmentation that is accelerated by the presence of the metal cofactor, zinc [Ahmed, S. A. et al. (2001) J. Protein Chem. 20, 221-231]. We show in this paper that >95% fragmented light chain obtained in the absence of added zinc retained 100% of its original catalytic activity against a SNAP-25-derived synthetic peptide substrate. In the presence of zinc chloride, when >95% of the light chain had undergone autocatalytic fragmentation, the preparation retained 35% of its original catalytic activity. On the other hand, in the presence of glycerol, the light chain did not display autocatalysis and retained 100% of the original activity. These results suggest that the activity loss by incubation with zinc was not a direct consequence of autocatalysis and that the environment of the active site was not affected significantly by the fragmentation. The optimum pH 4.2-4.6 for autocatalysis was different than that (pH 7.3) for intrinsic catalytic activity. Inhibition of autocatalysis at low pH by a competitive inhibitor of catalytic activity rules out the presence of a contaminating protease but suggests a rate-limiting step of low pH-induced conformational change suitable for autocatalysis. Our results of LC concentration dependence of the fragmentation reaction indicate that the autocatalysis occurs by both intramolecular and intermolecular mechanisms.


Subject(s)
Botulinum Toxins/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Botulinum Toxins/chemistry , Catalysis , Electrophoresis, Polyacrylamide Gel , Hydrogen-Ion Concentration , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...