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










Publication year range
1.
FEBS J ; 279(14): 2555-67, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22607388

ABSTRACT

A targeted drug carrier (TDC) is described for transferring functional proteins or peptides into motor nerve terminals, a pivotal locus for therapeutics to treat neuromuscular disorders. It exploits the pronounced selectivity of botulinum neurotoxin type B (BoNT/B) for interacting with acceptors on these cholinergic nerve endings and becoming internalized. The gene encoding an innocuous BoNT/B protease-inactive mutant (BoTIM) was fused to that for core streptavidin, expressed in Escherichia coli and the purified protein was conjugated to surface-biotinylated liposomes. Such decorated liposomes, loaded with fluorescein as traceable cargo, acquired pronounced specificity for motor nerve terminals in isolated mouse hemidiaphragms and facilitated the intraneuronal transfer of the fluor, as revealed by confocal microscopy. Delivery of the protease light chain of botulinum neurotoxin type A (BoNT/A) via this TDC accelerated the onset of neuromuscular paralysis, indicative of improved translocation of this enzyme into the presynaptic cytosol with subsequent proteolytic inactivation of synaptosomal-associated protein of molecular mass 25 kDa (SNAP-25), an exocytotic soluble N-ethyl-maleimide-sensitive factor attachment protein receptor (SNARE) essential for neurotransmitter release. BoTIM-coupled liposomes, loaded with peptide inhibitors of proteases, yielded considerable attenuation of the neuroparalytic effects of BoNT/A or BoNT/F as a result of their cytosolic transfer, the first in situ demonstration of the ability of designer antiproteases to suppress the symptoms of botulism ex vivo. Delivery of the BoNT/A inhibitor by liposomes targeted with the full-length BoTIM proved more effective than that mediated by its C-terminal neuroacceptor-binding domain. This demonstrated versatility of TDC for nonviral cargo transfer into cholinergic nerve endings has unveiled its potential for direct delivery of functional targets into motor nerve endings.


Subject(s)
Botulinum Toxins/metabolism , Drug Delivery Systems/methods , Presynaptic Terminals/metabolism , Protease Inhibitors/administration & dosage , SNARE Proteins/metabolism , Animals , Botulinum Toxins/chemistry , Botulinum Toxins/genetics , Botulinum Toxins, Type A , Diaphragm/metabolism , Fluorescein/chemistry , Humans , Liposomes , Mice , Microscopy, Confocal , Motor Neurons/metabolism , Mutation , Nerve Endings/metabolism , Paralysis/metabolism , Paralysis/therapy , Protease Inhibitors/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Streptavidin/genetics , Streptavidin/metabolism , Synaptosomal-Associated Protein 25/metabolism
2.
PLoS One ; 5(4): e10129, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405003

ABSTRACT

Botulinum neurotoxin serotype A (BoNTA) causes a life-threatening neuroparalytic disease known as botulism that could afflict large, unprotected populations if the toxin were employed in an act of bioterrorism. Current post-exposure therapy is limited to symptomatic treatment or passive immunization that is effective for treating infant botulism at a cost of US $45,300 per treatment regimen. Antibodies can neutralize the extracellular but not the intracellular BoNTA. Moreover, antibody production, storage, and administration in a mass casualty scenario pose logistical challenges. Alternatively, small-molecule inhibitors of BoNTA endopeptidase (BoNTAe) are sought to antagonize the extracellular or intracellular toxin. While several such molecules reportedly demonstrated efficacy in protecting cells against BoNTA, there is scant information to show that small molecules can significantly protect mammals against BoNTA. Herein we report the development of effective small-molecules BoNTAe inhibitors with promising in vivo pharmacokinetics. One such molecule has an in vivo half-life of 6.5 hours and is devoid of obvious sign of toxicity. Pre-treatment with this molecule at 2 mg/kg protected 100% and 70% of treated mice against BoNTA at 5 times of its median-lethal dose during the periods of 2 and 4 half-lives of the inhibitor, respectively. In contrast, 40% and 0% of untreated mice survived during the respective periods. Similar levels of protection were also observed with two other small molecules. These results demonstrate that small molecules can significantly protect mice against BoNTA and support the pursuit of small-molecule antagonists as a cost-effective alternative or as an adjunct to passive immunity for treating botulism.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Protease Inhibitors/pharmacokinetics , Animals , Bioterrorism , Costs and Cost Analysis , Mice , Neurotoxins , Protease Inhibitors/economics , Protease Inhibitors/therapeutic use , Protective Agents , Survival Rate
3.
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
4.
PLoS One ; 4(11): e7730, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19901994

ABSTRACT

Botulinum neurotoxin serotype A (BoNTA) causes a life-threatening neuroparalytic disease known as botulism. Current treatment for post exposure of BoNTA uses antibodies that are effective in neutralizing the extracellular toxin to prevent further intoxication but generally cannot rescue already intoxicated neurons. Effective small-molecule inhibitors of BoNTA endopeptidase (BoNTAe) are desirable because such inhibitors potentially can neutralize the intracellular BoNTA and offer complementary treatment for botulism. Previously we reported a serotype-selective, small-molecule BoNTAe inhibitor with a K(i) (app) value of 3.8+/-0.8 microM. This inhibitor was developed by lead identification using virtual screening followed by computer-aided optimization of a lead with an IC(50) value of 100 microM. However, it was difficult to further improve the lead from micromolar to even high nanomolar potency due to the unusually large enzyme-substrate interface of BoNTAe. The enzyme-substrate interface area of 4,840 A(2) for BoNTAe is about four times larger than the typical protein-protein interface area of 750-1,500 A(2). Inhibitors must carry several functional groups to block the unusually large interface of BoNTAe, and syntheses of such inhibitors are therefore time-consuming and expensive. Herein we report the development of a serotype-selective, small-molecule, and competitive inhibitor of BoNTAe with a K(i) value of 760+/-170 nM using synthesis-based computer-aided molecular design (SBCAMD). This new approach accounts the practicality and efficiency of inhibitor synthesis in addition to binding affinity and selectivity. We also report a three-dimensional model of BoNTAe in complex with the new inhibitor and the dynamics of the complex predicted by multiple molecular dynamics simulations, and discuss further structural optimization to achieve better in vivo efficacy in neutralizing BoNTA than those of our early micromolar leads. This work provides new insight into structural modification of known small-molecule BoNTAe inhibitors. It also demonstrates that SBCAMD is capable of improving potency of an inhibitor lead by nearly one order of magnitude, even for BoNTAe as one of the most challenging protein targets. The results are insightful for developing effective small-molecule inhibitors of protein targets with large active sites.


Subject(s)
Botulinum Toxins, Type A/chemistry , Endopeptidases/chemistry , Botulism/therapy , Computational Biology/methods , Computer Simulation , Drug Design , Humans , Inhibitory Concentration 50 , Kinetics , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Dynamics Simulation , Molecular Structure , Software , Structure-Activity Relationship
5.
Nat Struct Mol Biol ; 16(7): 789-94, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19543288

ABSTRACT

Clostridium botulinum neurotoxins (BoNTs) cleave neuronal proteins responsible for neurotransmitter release, causing the neuroparalytic disease botulism. BoNT serotypes B, D, F and G cleave and inactivate vesicle-associated membrane protein (VAMP), each at a unique peptide bond. The specificity of BoNTs depends on the mode of substrate recognition. We have investigated the mechanism of substrate recognition of BoNT F by determining the crystal structures of its complex with two substrate-based inhibitors, VAMP 22-58/Gln58D-cysteine and 27-58/Gln58D-cysteine. The inhibitors bind to BoNT F in the canonical direction (as seen for BoNTs A and E substrates) but are positioned specifically via three major exosites away from the active site. The cysteine sulfur of the inhibitors interacts with the zinc and exists as sulfinic acid in the inhibitor VAMP 27-58/Gln58D-cysteine. Arg133 and Arg171, which form part of two separate exosites, are crucial for substrate binding and catalysis.


Subject(s)
Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins/chemistry , Protein Isoforms/metabolism , R-SNARE Proteins/metabolism , Amino Acid Sequence , Botulinum Toxins/metabolism , Catalytic Domain , Crystallography, X-Ray , Cysteine/chemistry , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Protein Conformation , Protein Isoforms/genetics , R-SNARE Proteins/genetics , Substrate Specificity , Sulfur/chemistry
6.
ChemMedChem ; 3(12): 1905-12, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19006141

ABSTRACT

A search query consisting of two aromatic centers and two cationic centers was defined based on previously identified small molecule inhibitors of the botulinum neurotoxin serotype A light chain (BoNT/A LC) and used to mine the National Cancer Institute Open Repository. Ten small molecule hits were identified, and upon testing, three demonstrated inhibitory activity. Of these, one was structurally unique, possessing a rigid diazachrysene scaffold. The steric limitations of the diazachrysene imposed a separation between the overlaps of previously identified inhibitors, revealing an extended binding mode. As a result, the pharmacophore for BoNT/A LC inhibition has been modified to encompass three zones. To demonstrate the utility of this model, a novel three-zone inhibitor was mined and its activity was confirmed.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Chrysenes/chemistry , Models, Molecular , Botulinum Toxins, Type A/pharmacology , Chrysenes/pharmacology , Computer Simulation , Databases, Factual , Drug Design , Imaging, Three-Dimensional , Structure-Activity Relationship
7.
Structure ; 16(10): 1588-97, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18940613

ABSTRACT

Botulinum neurotoxin serotype A is the most lethal of all known toxins. Here, we report the crystal structure, along with SAR data, of the zinc metalloprotease domain of BoNT/A bound to a potent peptidomimetic inhibitor (K(i)=41 nM) that resembles the local sequence of the SNAP-25 substrate. Surprisingly, the inhibitor adopts a helical conformation around the cleavage site, in contrast to the extended conformation of the native substrate. The backbone of the inhibitor's P1 residue displaces the putative catalytic water molecule and concomitantly interacts with the "proton shuttle" E224. This mechanism of inhibition is aided by residue contacts in the conserved S1' pocket of the substrate binding cleft and by the induction of new hydrophobic pockets, which are not present in the apo form, especially for the P2' residue of the inhibitor. Our inhibitor is specific for BoNT/A as it does not inhibit other BoNT serotypes or thermolysin.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Synaptosomal-Associated Protein 25/chemistry , Amino Acid Sequence , Binding Sites , Biomimetics , Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/metabolism , Drug Evaluation, Preclinical , Models, Biological , Models, Molecular , Molecular Sequence Data , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Substrate Specificity , Synaptosomal-Associated Protein 25/metabolism
8.
Ann N Y Acad Sci ; 1132: 61-70, 2008.
Article in English | MEDLINE | ID: mdl-18567854

ABSTRACT

Although the neuromuscular nicotinic acetylcholine receptor (nAChR) is one of the most intensively studied ion channels in the nervous system, the differential roles of fetal and adult subtypes of the nAChR under normal and pathological conditions are still incompletely defined. Until recently, no pharmacological tools distinguished between fetal and adult subtypes. Waglerin toxins (from snake venom) and alphaA(S)-conotoxins (from cone-snail venom) have provided such tools. Because these peptides were characterized by different research groups using different methods, we have: 1) more extensively tested their subtype selectivity, and 2) begun to explore how these peptides may be used in concert to elucidate expression patterns and functions of fetal and adult nAChRs. In heterologous expression systems and native tissues, Waglerin-1 and an alphaA(S)-conotoxin analog, alphaA-OIVA[K15N], are high-affinity, highly selective inhibitors of the adult and fetal muscle nAChRs, respectively. We have used the peptides and their fluorescent derivatives to explore the expression and function of the fetal and adult nAChR subtypes. While fluorescent derivatives of these peptides indicated a gradual transition from fetal to adult muscle nAChRs in mice during the first 2 weeks postnatal, we unexpectedly observed a steeper transition in functional expression in the mouse diaphragm muscle using electrophysiology. As a toolkit of pharmacological agents with complementary specificity, alphaA-OIVA[K15N] and Waglerin-1 should have further utility in determining the roles of fetal and adult nAChR subtypes in development, in mature tissues, and under pathological conditions.


Subject(s)
Aging/physiology , Conotoxins/pharmacology , Crotalid Venoms/pharmacology , Receptors, Nicotinic/classification , Receptors, Nicotinic/metabolism , Animals , Electrophysiology , Kinetics , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Protein Binding , Protein Subunits/classification , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Nicotinic/genetics , Xenopus laevis
9.
Appl Environ Microbiol ; 74(3): 653-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18083881

ABSTRACT

The seven serotypes of botulinum neurotoxin (BoNTs) are zinc metalloproteases that cleave and inactivate proteins critical for neurotransmission. The synaptosomal protein of 25 kDa (SNAP-25) is cleaved by BoNTs A, C, and E, while vesicle-associated membrane protein (VAMP) is the substrate for BoNTs B, D, F, and G. BoNTs not only are medically useful drugs but also are potential bioterrorist and biowarfare threat agents. Because BoNT protease activity is required for toxicity, inhibitors of that activity might be effective for antibotulinum therapy. To expedite inhibitor discovery, we constructed a hybrid gene encoding (from the N terminus to the C terminus, with respect to the expressed product) green fluorescent protein, then a SNAP-25 fragment encompassing residues Met-127 to Gly-206, and then VAMP residues Met-1 to Lys-94. Cysteine was added as the C terminus. The expressed product, which contained the protease cleavage sites for all seven botulinum serotypes, was purified and coupled covalently through the C-terminal sulfhydryl group to maleimide-activated 96-well plates. The substrate was readily cleaved by BoNTs A, B, D, E, and F. Using this assay and an automated 96-well pipettor, we screened 528 natural product extracts for inhibitors of BoNT A, B, and E protease activities. Serotype-specific inhibition was found in 30 extracts, while 5 others inhibited two serotypes.


Subject(s)
Botulinum Toxins, Type A/metabolism , Botulinum Toxins/metabolism , Green Fluorescent Proteins , Recombinant Proteins , Antitoxins/chemistry , Antitoxins/metabolism , Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins, Type A/antagonists & inhibitors , Fluorescent Dyes , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Vesicle-Associated Membrane Protein 2/chemistry , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
10.
PLoS One ; 2(8): e761, 2007 Aug 22.
Article in English | MEDLINE | ID: mdl-17712409

ABSTRACT

Optimization of a serotype-selective, small-molecule inhibitor of botulinum neurotoxin serotype A (BoNTA) endopeptidase is a formidable challenge because the enzyme-substrate interface is unusually large and the endopeptidase itself is a large, zinc-binding protein with a complex fold that is difficult to simulate computationally. We conducted multiple molecular dynamics simulations of the endopeptidase in complex with a previously described inhibitor (K(i) (app) of 7+/-2.4 microM) using the cationic dummy atom approach. Based on our computational results, we hypothesized that introducing a hydroxyl group to the inhibitor could improve its potency. Synthesis and testing of the hydroxyl-containing analog as a BoNTA endopeptidase inhibitor showed a twofold improvement in inhibitory potency (K(i) (app) of 3.8+/-0.8 microM) with a relatively small increase in molecular weight (16 Da). The results offer an improved template for further optimization of BoNTA endopeptidase inhibitors and demonstrate the effectiveness of the cationic dummy atom approach in the design and optimization of zinc protease inhibitors.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Drug Design , Endopeptidases/metabolism , Protease Inhibitors/chemistry , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Catalytic Domain , Computer Simulation , Endopeptidases/chemistry , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism
11.
J Med Chem ; 50(9): 2127-36, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17417831

ABSTRACT

We previously identified structurally diverse small molecule (non-peptidic) inhibitors (SMNPIs) of the botulinum neurotoxin serotype A (BoNT/A) light chain (LC). Of these, several (including antimalarial drugs) contained a 4-amino-7-chloroquinoline (ACQ) substructure and a separate positive ionizable amine component. The same antimalarials have also been found to interfere with BoNT/A translocation into neurons, via pH elevation of the toxin-mediated endosome. Thus, this structural class of small molecules may serve as dual-function BoNT/A inhibitors. In this study, we used a refined pharmacophore for BoNT/A LC inhibition to identify four new, potent inhibitors of this structural class (IC50's ranged from 3.2 to 17 muM). Molecular docking indicated that the binding modes for the new SMNPIs are consistent with those of other inhibitors that we have identified, further supporting our structure-based pharmacophore. Finally, structural motifs of the new SMNPIs, as well as two structure-based derivatives, were examined for activity, providing valuable information about pharmacophore component contributions to inhibition.


Subject(s)
Aminoquinolines/chemical synthesis , Botulinum Toxins, Type A/antagonists & inhibitors , Metalloproteases/antagonists & inhibitors , Metalloproteases/chemistry , Models, Molecular , Aminoquinolines/chemistry , Binding Sites , Protein Binding , Structure-Activity Relationship
12.
J Biol Chem ; 282(7): 5004-5014, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17092934

ABSTRACT

An efficient research strategy integrating empirically guided, structure-based modeling and chemoinformatics was used to discover potent small molecule inhibitors of the botulinum neurotoxin serotype A light chain. First, a modeled binding mode for inhibitor 2-mercapto-3-phenylpropionyl-RATKML (K(i) = 330 nM) was generated, and required the use of a molecular dynamic conformer of the enzyme displaying the reorientation of surface loops bordering the substrate binding cleft. These flexible loops are conformationally variable in x-ray crystal structures, and the model predicted that they were pivotal for providing complementary binding surfaces and solvent shielding for the pseudo-peptide. The docked conformation of 2-mercapto-3-phenylpropionyl-RATKML was then used to refine our pharmacophore for botulinum serotype A light chain inhibition. Data base search queries derived from the pharmacophore were employed to mine small molecule (non-peptidic) inhibitors from the National Cancer Institute's Open Repository. Four of the inhibitors possess K(i) values ranging from 3.0 to 10.0 microM. Of these, NSC 240898 is a promising lead for therapeutic development, as it readily enters neurons, exhibits no neuronal toxicity, and elicits dose-dependent protection of synaptosomal-associated protein (of 25 kDa) in a primary culture of embryonic chicken neurons. Isothermal titration calorimetry showed that the interaction between NSC 240898 and the botulinum A light chain is largely entropy-driven, and occurs with a 1:1 stoichiometry and a dissociation constant of 4.6 microM.


Subject(s)
Botulinum Toxins, Type A/chemistry , Metalloproteases/chemistry , Models, Molecular , Neurons/chemistry , Protease Inhibitors/chemistry , Animals , Botulinum Toxins, Type A/metabolism , Botulism/drug therapy , Botulism/enzymology , Cells, Cultured , Chick Embryo , Metalloproteases/metabolism , Neurons/enzymology , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use
13.
Bioorg Med Chem ; 14(10): 3583-91, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16458011

ABSTRACT

Botulinum neurotoxin serotype A (BoNTA) is highly toxic, and its antidote is currently unavailable. The essential light-chain subunit of BoNTA is a zinc endopeptidase that can be used as a target for developing antidotes. However, the development of high-affinity, small-molecule inhibitors of the endopeptidase is as challenging as the development of small-molecule inhibitors of protein-protein complexation. This is because the polypeptide substrate wraps around the circumference of the endopeptidase upon binding, thereby constituting an unusually large substrate-enzyme interface of 4840 angstroms2. To overcome the large-interface problem, we propose using the zinc-coordination and bivalence approaches to design inhibitors of BoNTA. Here we report the development of alkylene-linked bis-imidazoles that inhibit the endopeptidase in a two-site binding mode. The bis-imidazole tethered with 13 methylene groups, the most potent of the alkylene-linked dimers, showed 61% inhibition of the zinc endopeptidase of BoNTA at a concentration of 100 microM. The results demonstrate the presence of a peripheral binding site for an imidazolium group at the rim of the BoNTA active-site cleft. This peripheral site enables the use of the bivalence approach to improve our previously reported small-molecule inhibitors that were developed according to the zinc-coordination approach.


Subject(s)
Botulinum Toxins, Type A/chemistry , Endopeptidases/chemistry , Enzyme Inhibitors/chemistry , Imidazoles/chemistry , Molecular Probes/chemistry , Zinc/chemistry , Binding Sites , Botulinum Toxins, Type A/antagonists & inhibitors , Computer Simulation , Endopeptidases/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Models, Biological , Models, Molecular , Molecular Probes/pharmacology , Molecular Structure
14.
Bioorg Med Chem ; 14(2): 395-408, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16203152

ABSTRACT

Botulinum neurotoxin serotype A (BoNTA) is one of the most toxic substances known. Currently, there is no antidote to BoNTA. Small molecules identified from high-throughput screening reportedly inhibit the endopeptidase--the zinc-bound, catalytic domain of BoNTA--at a drug concentration of 20 microM. However, optimization of these inhibitors is hampered by challenges including the computational evaluation of the ability of a zinc ligand to compete for coordination with nearby residues in the active site of BoNTA. No improved inhibitor of the endopeptidase has been reported. This article reports the development of a serotype-selective, small-molecule inhibitor of BoNTA with a K(i) of 12 microM. This inhibitor was designed to coordinate the zinc ion embedded in the active site of the enzyme for affinity and to interact with a species-specific residue in the active site for selectivity. It is the most potent small-molecule inhibitor of BoNTA reported to date. The results suggest that multiple molecular dynamics simulations using the cationic dummy atom approach are useful to structure-based design of zinc protease inhibitors.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Endopeptidases/chemistry , Protease Inhibitors/pharmacology , Botulinum Toxins, Type A/chemistry , Catalytic Domain , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Protease Inhibitors/chemistry , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Infrared
15.
J Biomol Screen ; 10(8): 788-94, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16234350

ABSTRACT

A microfluidic assay was developed for screening botulinum neurotoxin serotype A (BoNT-A) by using a fluorescent resonance energy transfer (FRET) assay. Molded silicone microdevices with integral valves, pumps, and reagent reservoirs were designed and fabricated. Electrical and pneumatic control hardware were constructed, and software was written to automate the assay protocol and data acquisition. Detection was accomplished by fluorescence microscopy. The system was validated with a peptide inhibitor, running 2 parallel assays, as a feasibility demonstration. The small footprint of each bioreactor cell (0.5 cm2) and scalable fluidic architecture enabled many parallel assays on a single chip. The chip is programmable to run a dilution series in each lane, generating concentration-response data for multiple inhibitors. The assay results showed good agreement with the corresponding experiments done at a macroscale level. Although the system has been developed for BoNT-A screening, a wide variety of assays can be performed on the microfluidic chip with little or no modification.


Subject(s)
Biological Assay/methods , Botulinum Toxins, Type A/analysis , Microfluidics/instrumentation , Biological Assay/instrumentation , Feasibility Studies , Microchemistry/instrumentation , Miniaturization
16.
Biochemistry ; 44(10): 4067-73, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15751983

ABSTRACT

Botulinum neurotoxins (BoNTs A-G) are zinc metalloendoproteases that exhibit extraordinary specificities for proteins involved in neurotransmitter release. In view of the extreme toxicities of these molecules, their applications in human medicine, and potential for misuse, it is of considerable importance to elucidate the mechanisms underlying substrate recognition and to develop inhibitors, with the ultimate goal of obtaining anti-botulinum drugs. We synthesized peptides based on vesicle-associated membrane protein (VAMP) to investigate the substrate requirements of BoNT F, which cleaves VAMP between residues Q58 and K59. The minimum substrate was a peptide containing VAMP residues 32-65, which includes only one of the two VAMP structural motifs thought to be required for botulinum substrate recognition. BoNT F exhibited a strict requirement for residues D57 (P(2)), K59 (P(1)'), and L60 (P(2)'), but peptides containing substitutions for R56 (P(3)), Q58 (P(1)), and S61 (P(3)') were cleaved. Therefore, the P(2), P(1)', and P(2)' residues of VAMP are of paramount importance for BoNT F substrate recognition near the scissile bond. K(i) values of uncleavable analogues were similar to K(m) values of the substrate, suggesting that substrate discrimination occurs at the cleavage step, not at the initial binding step. We then synthesized inhibitors of BoNT F that incorporated d-cysteine in place of glutamine 58, exhibited K(i) values of 1-2 nM, and required binding groups on the N-terminal but not the C-terminal side of the zinc ligand. The latter characteristic distinguishes BoNT F from other zinc metalloendoproteases, including BoNTs A and B.


Subject(s)
Antitoxins/metabolism , Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins/metabolism , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Protease Inhibitors/metabolism , Amino Acid Sequence , Amino Acid Substitution , Antitoxins/chemistry , Botulinum Toxins/chemistry , Catalysis , Cysteine/chemistry , Cysteine/metabolism , Humans , Hydrolysis , Kinetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemical synthesis , Membrane Proteins/metabolism , Metalloproteases/chemistry , Molecular Sequence Data , Nanotechnology/methods , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protease Inhibitors/chemical synthesis , Protein Binding , R-SNARE Proteins , Serotyping , Substrate Specificity , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
17.
Bioorg Med Chem ; 13(2): 333-41, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15598556

ABSTRACT

Botulinum neurotoxins (BoNTs) are the most potent of the known biological toxins, and consequently are listed as category A biowarfare agents. Currently, the only treatments against BoNTs include preventative antitoxins and long-term supportive care. Consequently, there is an urgent need for therapeutics to counter these enzymes--post exposure. In a previous study, we identified a number of small, nonpeptidic lead inhibitors of BoNT serotype A light chain (BoNT/A LC) metalloprotease activity, and we identified a common pharmacophore for these molecules. In this study, we have focused on how the dynamic movement of amino acid residues in and surrounding the substrate binding cleft of the BoNT/A LC might affect inhibitor binding modes. The X-ray crystal structures of two BoNT/A LCs (PDB refcodes=3BTA and 1E1H) were examined. Results from these analyses indicate that the core structural features of the examined BoNT/A LCs, including alpha-helices and beta-sheets, remained relatively unchanged during 1 ns dynamics trajectories. However, conformational flexibility was observed in surface loops bordering the substrate binding clefts in both examined structures. Our analyses indicate that these loops may possess the ability to decrease the solvent accessibility of the substrate binding cleft, while at the same time creating new residue contacts for the inhibitors. Loop movements and conformational/positional analyses of residues within the substrate binding cleft are discussed with respect to BoNT/A LC inhibitor binding and our common pharmacophore for inhibition. The results from these studies may aid in the future identification/development of more potent small molecule inhibitors that take advantage of new binding contacts in the BoNT/A LC.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/metabolism , Isoquinolines/chemistry , Metalloproteases/antagonists & inhibitors , Models, Molecular , Molecular Structure , Naphthalenes/chemistry , Protein Binding , Protein Conformation , Protein Subunits , Quinolines/chemistry , Silver/chemistry , Structure-Activity Relationship
18.
Biochem Biophys Res Commun ; 310(1): 84-93, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-14511652

ABSTRACT

Botulinum neurotoxins (BoNTs) are among the most lethal biological substances to have been weaponized and are listed as biodefense category A agents. Currently, no small molecule (non-peptidic) therapeutics exist to counter this threat; hence, identifying and developing compounds that inhibit BoNTs is a high priority. In the present study, a high-throughput assay was used to identify small molecules that inhibit the metalloprotease activity of BoNT serotype A light chain (BoNT/A LC). All inhibitors were further verified using a HPLC-based assay. Conformational analyses of these compounds, in conjunction with molecular docking studies, were used to predict structural features that contribute to inhibitor binding and potency. Based on these results, a common pharmacophore for BoNT/A LC inhibitors is proposed. This is the first study to report small molecules (non-peptidics) that inhibit BoNT/A LC metalloprotease activity in the low microM range.


Subject(s)
Botulinum Toxins/antagonists & inhibitors , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/pharmacology
19.
Appl Environ Microbiol ; 69(1): 297-303, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12514008

ABSTRACT

The seven botulinum neurotoxins (BoNTs) are zinc metalloproteases that cleave neuronal proteins involved in neurotransmitter release and are among the most toxic natural products known. High-throughput BoNT assays are needed for use in antibotulinum drug discovery and to characterize BoNT protease activities. Compared to other proteases, BoNTs exhibit unusually stringent substrate requirements with respect to amino acid sequences and polypeptide lengths. Nonetheless, we have devised a strategy for development of fluorigenic BoNT protease assays, based on earlier structure-function studies, that has proven successful for three of the seven serotypes: A, B, and F. In synthetic peptide substrates, the P(1) and P(3)' residues were substituted with 2,4-dinitrophenyl-lysine and S-(N-[4-methyl-7-dimethylamino-coumarin-3-yl]-carboxamidomethyl)-cysteine, respectively. By monitoring the BoNT-catalyzed increase in fluorescence over time, initial hydrolysis rates could be obtained in 1 to 2 min when BoNT concentrations were 60 ng/ml (about 1 nM) or higher. Each BoNT cleaved its fluorigenic substrate at the same location as in the neuronal target protein, and kinetic constants indicated that the substrates were selective and efficient. The fluorigenic assay for BoNT B was used to characterize a new competitive inhibitor of BoNT B protease activity with a K(i) value of 4 micro M. In addition to real-time activity measurements, toxin concentration determinations, and kinetic studies, the BoNT substrates described herein may be directly incorporated into automated high-throughput assay systems to screen large numbers of compounds for potential antibotulinum drugs.


Subject(s)
Botulinum Toxins/metabolism , Endopeptidases/metabolism , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Amino Acid Sequence , Botulinum Toxins, Type A/metabolism , Drug Evaluation, Preclinical , Fluorescent Dyes/chemistry , Molecular Sequence Data , Peptides/chemistry , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Substrate Specificity
20.
FEBS Lett ; 532(3): 423-6, 2002 Dec 18.
Article in English | MEDLINE | ID: mdl-12482605

ABSTRACT

The peptide N-acetyl-CRATKML-amide is an effective inhibitor of type A botulinum neurotoxin (BoNT A) protease activity [Schmidt et al., FEBS Lett. 435 (1998) 61-64]. To improve inhibitor binding, the peptide was modified by replacing cysteine with other sulfhydryl-containing compounds. Ten peptides were synthesized. One peptide adapted the structure of captopril to the binding requirements of BoNT A, but it was a weak inhibitor, suggesting that angiotensin-converting enzyme is not a good model for BoNT A inhibitor development. However, replacing cysteine with 2-mercapto-3-phenylpropionyl yielded a peptide with K(i) of 330 nM, the best inhibitor of BoNT A protease activity reported to date. Additional modifications of the inhibitor revealed structural elements important for binding and supported our earlier findings that, with the exception of P1' arginine, subsites on BoNT A are not highly specific for particular amino acid side chains.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Botulinum Toxins, Type A/chemistry , Protease Inhibitors/pharmacology , Binding, Competitive , Catalytic Domain , Chromatography, High Pressure Liquid , Kinetics , Models, Chemical , Peptide Biosynthesis , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...