Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 3: 3397, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24292269

ABSTRACT

We reported previously (±)-2-(5-methylthiophen-2-yl)-3-phenyl-2,3-dihydroquinazolin-4(1H)-one [(±)-Retro-2(cycl)] as the chemical structure of Retro-2 that showed mouse protection against ricin, a notorious ribosome inactivating protein (RIP). Herein we report our chemical resolution of (±)-Retro-2(cycl), analog synthesis, and cell-based evaluation showing that the two optically pure enantiomers and their achiral analog have nearly the same degree of cell protection against ricin as (±)-Retro-2(cycl). We also report our computational studies explaining the lack of stereo preference and revealing a common pharmacophore of structurally distinct inhibitors of intracellular retrograde trafficking of RIPs. This pharmacophore comprises a central aromatic ring o-substituted by an aromatic ring and a moiety bearing an O or S atom attached to sp² C atom(s). These results offer new insights into lead identification and optimization for RIP antidote development to minimize the global health threat caused by ribosome-inactivating proteins.


Subject(s)
Protein Transport/drug effects , Ribosome Inactivating Proteins/metabolism , Small Molecule Libraries/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Ricin/metabolism , Stereoisomerism , Vero Cells
2.
Sci Rep ; 3: 1068, 2013.
Article in English | MEDLINE | ID: mdl-23323211

ABSTRACT

We reported previously that insect acetylcholinesterases (AChEs) could be selectively and irreversibly inhibited by methanethiosulfonates presumably through conjugation to an insect-specific cysteine in these enzymes. However, no direct proof for the conjugation has been published to date, and doubts remain about whether such cysteine-targeting inhibitors have desirable kinetic properties for insecticide use. Here we report mass spectrometric proof of the conjugation and new chemicals that irreversibly inhibited African malaria mosquito AChE with bimolecular inhibition rate constants (k(inact)/K(I)) of 3,604-458,597 M(-1)sec(-1) but spared human AChE. In comparison, the insecticide paraoxon irreversibly inhibited mosquito and human AChEs with k(inact)/K(I) values of 1,915 and 1,507 M(-1)sec(-1), respectively, under the same assay conditions. These results further support our hypothesis that the insect-specific AChE cysteine is a unique and unexplored target to develop new insecticides with reduced insecticide resistance and low toxicity to mammals, fish, and birds for the control of mosquito-borne diseases.


Subject(s)
Acetylcholinesterase/metabolism , Culicidae/enzymology , Protozoan Proteins/metabolism , Acetylcholinesterase/chemistry , Animals , Antimalarials/chemistry , Antimalarials/metabolism , Antimalarials/toxicity , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Culicidae/drug effects , Humans , Insecticides/chemistry , Insecticides/metabolism , Insecticides/toxicity , Kinetics , Malaria/prevention & control , Mass Spectrometry , Paraoxon/chemistry , Paraoxon/metabolism , Paraoxon/toxicity , Protein Binding , Protozoan Proteins/chemistry
3.
Sci Rep ; 2: 631, 2012.
Article in English | MEDLINE | ID: mdl-22953052

ABSTRACT

Shiga-like toxins and ricin are ribosome-inactivating proteins (RIPs) that are lethal to mammals and pose a global health threat. No clinical vaccines or therapeutics currently exist to protect against these RIPs. Two small molecules (Retro-1 and Retro-2) were discovered with high-throughput screening and reported for their protection of cells against RIPs. Of great significance, Retro-2, reported as (E)-2-(((5-methylthiophen-2-yl)methylene)amino)-N-phenylbenzamide, fully protected mice from lethal nasal challenge with ricin. Herein, we report studies showing that the chemical structure of Retro-2 is (±)-2-(5-methylthiophen-2-yl)-3-phenyl-2,3-dihydroquinazolin-4(1H)-one rather than (E)-2-(((5-methylthiophen-2-yl)methylene)amino)-N-phenylbenzamide. The latter is an achiral molecule that converts spontaneously to the former, which is a racemate and showed cell protection against RIPs. This calls for attention to (±)-2-(5-methylthiophen-2-yl)-3-phenyl-2,3-dihydroquinazolin-4(1H)-one as a promising RIP inhibitor and for chemical characterization of drug leads obtained from high-throughput screens.


Subject(s)
Benzamides/chemistry , Ricin/pharmacology , Shiga Toxin 2/pharmacology , Thiophenes/chemistry , Animals , Benzamides/chemical synthesis , Benzamides/pharmacology , Chlorocebus aethiops , Cytoprotection , Drug Stability , Magnetic Resonance Spectroscopy , Methanol/chemistry , Molecular Structure , Ribosome Inactivating Proteins , Solvents/chemistry , Stereoisomerism , Thiophenes/chemical synthesis , Thiophenes/pharmacology , Vero Cells
4.
PLoS One ; 6(3): e17883, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21455295

ABSTRACT

Ribosome-inactivating proteins (RIPs) are toxic because they bind to 28S rRNA and depurinate a specific adenine residue from the α-sarcin/ricin loop (SRL), thereby inhibiting protein synthesis. Shiga-like toxins (Stx1 and Stx2), produced by Escherichia coli, are RIPs that cause outbreaks of foodborne diseases with significant morbidity and mortality. Ricin, produced by the castor bean plant, is another RIP lethal to mammals. Currently, no US Food and Drug Administration-approved vaccines nor therapeutics exist to protect against ricin, Shiga-like toxins, or other RIPs. Development of effective small-molecule RIP inhibitors as therapeutics is challenging because strong electrostatic interactions at the RIP•SRL interface make drug-like molecules ineffective in competing with the rRNA for binding to RIPs. Herein, we report small molecules that show up to 20% cell protection against ricin or Stx2 at a drug concentration of 300 nM. These molecules were discovered using the doorstop approach, a new approach to protein•polynucleotide inhibitors that identifies small molecules as doorstops to prevent an active-site residue of an RIP (e.g., Tyr80 of ricin or Tyr77 of Stx2) from adopting an active conformation thereby blocking the function of the protein rather than contenders in the competition for binding to the RIP. This work offers promising leads for developing RIP therapeutics. The results suggest that the doorstop approach might also be applicable in the development of other protein•polynucleotide inhibitors as antiviral agents such as inhibitors of the Z-DNA binding proteins in poxviruses. This work also calls for careful chemical and biological characterization of drug leads obtained from chemical screens to avoid the identification of irrelevant chemical structures and to avoid the interference caused by direct interactions between the chemicals being screened and the luciferase reporter used in screening assays.


Subject(s)
Polynucleotides/pharmacology , Ribosome Inactivating Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Mice , Molecular Structure , Polynucleotides/chemistry , Ribosome Inactivating Proteins/chemistry , Ricin/chemistry , Shiga Toxin 2/chemistry
5.
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
6.
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
7.
FEBS Lett ; 583(13): 2208-12, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19527717

ABSTRACT

Indiscriminately suppressing total c-Jun N-terminal kinase (JNK) activity is not an appropriate strategy because each JNK appears to have a distinct function in cancer, asthma, diabetes, or Parkinson's disease. Herein, we report that 7-(6-N-phenylaminohexyl)amino-2H-anthra[1,9-cd]pyrazol-6-one (AV-7) inhibited JNK1 activity, but not JNK2 or JNK3. We found that ultraviolet B (UVB) induced c-Jun phosphorylation and sub-G1 accumulation in JNK2(-/-) murine embryonic fibroblasts, which contain an abundance of JNK1, but not JNK2. These results demonstrate that AV-7 is an isoform selective small-molecule inhibitor of JNK1 activity, which might be developed as a therapeutic against diabetes.


Subject(s)
Aniline Compounds/chemistry , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Humans , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Phosphorylation , Ultraviolet Rays
8.
Tetrahedron Lett ; 49(7): 1103-1106, 2008 Feb 11.
Article in English | MEDLINE | ID: mdl-18443652

ABSTRACT

Several resins have reportedly been used to synthesize hydroxamic acids except for the hydroxythiophenol (Marshall) resin. Herein we report the use of the Marshall resin to synthesize hydroxamic acids from carboxylic acids and its application to convert a library of fourteen discrete aliphatic and aromatic carboxylic acids including N-protected amino acids to their corresponding hydroxamic acids in good yields.

9.
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
10.
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
11.
Bioorg Med Chem Lett ; 16(4): 830-3, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16325400

ABSTRACT

Virtual screening, a fast, computational approach to identify drug leads [Perola, E.; Xu, K.; Kollmeyer, T. M.; Kaufmann, S. H.; Prendergast, F. G. J. Med. Chem.2000, 43, 401; Miller, M. A. Nat. Rev. Drug Disc.2002, 1 220], is limited by a known challenge in crystallographically determining flexible regions of proteins. This approach has not been able to identify active inhibitors of the severe acute respiratory syndrome-associated coronavirus (SARS-CoV) using solely the crystal structures of a SARS-CoV cysteine proteinase with a flexible loop in the active site [Yang, H. T.; Yang, M. J.; Ding, Y.; Liu, Y. W.; Lou, Z. Y. Proc. Natl. Acad. Sci. U.S.A.2003, 100, 13190; Jenwitheesuk, E.; Samudrala, R. Bioorg. Med. Chem. Lett.2003, 13, 3989; Rajnarayanan, R. V.; Dakshanamurthy, S.; Pattabiraman, N. Biochem. Biophys. Res. Commun.2004, 321, 370; Du, Q.; Wang, S.; Wei, D.; Sirois, S.; Chou, K. Anal. Biochem.2005, 337, 262; Du, Q.; Wang, S.; Zhu, Y.; Wei, D.; Guo, H. Peptides2004, 25, 1857; Lee, V.; Wittayanarakul, K.; Remsungenen, T.; Parasuk, V.; Sompornpisut, P. Science (Asia)2003, 29, 181; Toney, J.; Navas-Martin, S.; Weiss, S.; Koeller, A. J. Med. Chem.2004, 47, 1079; Zhang, X. W.; Yap, Y. L. Bioorg. Med. Chem.2004, 12, 2517]. This article demonstrates a genome-to-drug-lead approach that uses terascale computing to model flexible regions of proteins, thus permitting the utilization of genetic information to identify drug leads expeditiously. A small-molecule inhibitor of SARS-CoV, exhibiting an effective concentration (EC50) of 23 microM in cell-based assays, was identified through virtual screening against a computer-predicted model of the cysteine proteinase. Screening against two crystal structures of the same proteinase failed to identify the 23-microM inhibitor. This study suggests that terascale computing can complement crystallography, broaden the scope of virtual screening, and accelerate the development of therapeutics to treat emerging infectious diseases such as SARS and Bird Flu.


Subject(s)
Aminobenzoates/pharmacology , Cysteine Endopeptidases/drug effects , Drug Design , Enzyme Inhibitors/pharmacology , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/genetics , Aminobenzoates/chemistry , Computer Simulation , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Severe acute respiratory syndrome-related coronavirus/enzymology , Structure-Activity Relationship
12.
J Comb Chem ; 6(3): 407-13, 2004.
Article in English | MEDLINE | ID: mdl-15132601

ABSTRACT

The use of chlorotrityl resins for the immobilization of amines is sometimes deterred by the lengthy process of loading the reactants on the resins and product decomposition caused by the reactive chlorotrityl group in the presence of 1% TFA as a cleavage agent. Here, we report improved methods developed for selective and efficient loading of aminobenzoic acid derivatives on chlorotrityl resins and for cleavage of aniline-containing products from the resins without decomposition. These methods led to the synthesis of a library of 144 discrete chemicals as potential farnesyltransferase inhibitors (FTIs) using IRORI's radio-frequency-encoded sorting technique and to the study of the applicability of the bivalence approach to the development of FTIs.


Subject(s)
Farnesyltranstransferase , Solid-Phase Synthesis Techniques , Amino Acids , Combinatorial Chemistry Techniques , Enzyme Inhibitors , Molecular Structure , Resins, Plant
13.
Arch Pharm (Weinheim) ; 336(4-5): 230-5, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12916057

ABSTRACT

The synthesis and biological test of 5-(4-alkylsulfanyl-[1, 2, 5]thiadiazol-3-yl)-3-me-thyl-1, 2, 3, 4-tetrahydropyrimidine oxalate salts 7 as muscarinic receptor agonists are described. The key intermediate 4 was obtained by a modified Strecker reaction and cyclization, and the 3-methyl-1, 2, 3, 4-tetrahydropyrimidines were obtained by subsequent substitution, quarternization, and reduction. The final products 7 were obtained as oxalic acid salts. The prepared compounds were examined in vitro for their binding affinities to the cloned human muscarinic receptor by the [(3)H]-NMS binding assay.


Subject(s)
Muscarinic Agonists/chemical synthesis , Oxalates/chemical synthesis , Pyrimidines/chemical synthesis , Receptors, Muscarinic/metabolism , Thiazoles/chemical synthesis , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Humans , Molecular Structure , Muscarinic Agonists/chemistry , Muscarinic Agonists/pharmacology , Oxalates/chemistry , Oxalates/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptor, Muscarinic M1 , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...