Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Antimicrob Chemother ; 71(10): 2874-82, 2016 10.
Article in English | MEDLINE | ID: mdl-27330072

ABSTRACT

OBJECTIVES: Inhibitors of uridine diphosphate-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC, which catalyses the first, irreversible step in lipid A biosynthesis) are a promising new class of antibiotics against Gram-negative bacteria. The objectives of the present study were to: (i) compare the antibiotic activities of three LpxC inhibitors (LPC-058, LPC-011 and LPC-087) and the reference inhibitor CHIR-090 against Gram-negative bacilli (including MDR and XDR isolates); and (ii) investigate the effect of combining these inhibitors with conventional antibiotics. METHODS: MICs were determined for 369 clinical isolates (234 Enterobacteriaceae and 135 non-fermentative Gram-negative bacilli). Time-kill assays with LPC-058 were performed on four MDR/XDR strains, including Escherichia coli producing CTX-M-15 ESBL and Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii producing KPC-2, VIM-1 and OXA-23 carbapenemases, respectively. RESULTS: LPC-058 was the most potent antibiotic and displayed the broadest spectrum of antimicrobial activity, with MIC90 values for Enterobacteriaceae, P. aeruginosa, Burkholderia cepacia and A. baumannii of 0.12, 0.5, 1 and 1 mg/L, respectively. LPC-058 was bactericidal at 1× or 2× MIC against CTX-M-15, KPC-2 and VIM-1 carbapenemase-producing strains and bacteriostatic at ≤4× MIC against OXA-23 carbapenemase-producing A. baumannii. Combinations of LPC-058 with ß-lactams, amikacin and ciprofloxacin were synergistic against these strains, albeit in a species-dependent manner. LPC-058's high efficacy was attributed to the presence of the difluoromethyl-allo-threonyl head group and a linear biphenyl-diacetylene tail group. CONCLUSIONS: These in vitro data highlight the therapeutic potential of the new LpxC inhibitor LPC-058 against MDR/XDR strains and set the stage for subsequent in vivo studies.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Enterobacteriaceae/drug effects , Enzyme Inhibitors/pharmacology , Gram-Negative Bacteria/drug effects , Hydroxamic Acids/pharmacology , Threonine/analogs & derivatives , Acinetobacter baumannii/drug effects , Bacterial Proteins/biosynthesis , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae/enzymology , Enterobacteriaceae Infections/microbiology , Escherichia coli/drug effects , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/pathogenicity , Humans , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Threonine/pharmacology , beta-Lactamases/biosynthesis
2.
J Org Chem ; 81(10): 4393-8, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27128325

ABSTRACT

The difluoromethyl-allo-threonyl hydroxamate-based compound LPC-058 is a potent inhibitor of UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) in Gram-negative bacteria. A scalable synthesis of this compound is described. The key step in the synthetic sequence is a transition metal/base-catalyzed aldol reaction of methyl isocyanoacetate and difluoroacetone, giving rise to 4-(methoxycarbonyl)-5,5-disubstituted 2-oxazoline. A simple NMR-based determination of enantiomeric purity is also described.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Benzamides/chemical synthesis , Benzamides/pharmacology , Catalysis , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Indicators and Reagents , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation
3.
Langmuir ; 31(11): 3431-40, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25738531

ABSTRACT

The need for effective enzymatic depolymerization of cellulose has stimulated an interest in interactions between protein and cellulose. Techniques utilized for quantitative measurements of protein-cellulose noncovalent association include microgravimetry, calorimetry, and atomic force microscopy (AFM), none of which differentiate between specific protein-cellulose binding and nonspecific adhesion. Here, we describe an AFM approach that differentiates nonspecific from specific interactions between cellulose-binding modules (CBMs) and cellulose. We demonstrate that the "mismatched" interaction between murine galectin-3, a lectin with no known affinity for cellulose, and cellulose shows molecular recognition force microscopy profiles similar to those observed during the interaction of a "matched" clostridial CBM3a with the same substrate. We also examine differences in binding probabilities and rupture profiles during CBM-cellulose binding experiments in the presence and absence of a blocking agent-a substrate specific for CBM that presumably blocks binding sites. By comparison of the behavior of the two proteins, we separate specific (i.e., blockable) and nonspecific adhesion events and show that both classes of interaction exhibit nearly identical rupture forces (45 pN at ∼0.4 nN/s). Our work provides an important caveat for the interpretation of protein-carbohydrate binding by force spectroscopy; delineation of the importance of such interactions to other classes of binding warrants further study.


Subject(s)
Cellulose/chemistry , Microscopy, Atomic Force/methods , Binding Sites , Calorimetry , Protein Binding , Weightlessness
4.
Proc Natl Acad Sci U S A ; 108(25): 10284-9, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21628561

ABSTRACT

Lipopolysaccharides (LPS) and lipooligosaccharides (LOS) are the main lipid components of bacterial outer membranes and are essential for cell viability in most Gram-negative bacteria. Here we show that small molecule inhibitors of LpxC [UDP-3-O-(R-3-hydroxymyristoyl)-GlcNAc deacetylase], the enzyme that catalyzes the first committed step in the biosynthesis of lipid A, block the synthesis of LOS in the obligate intracellular bacterial pathogen Chlamydia trachomatis. In the absence of LOS, Chlamydia remains viable and establishes a pathogenic vacuole ("inclusion") that supports robust bacterial replication. However, bacteria grown under these conditions were no longer infectious. In the presence of LpxC inhibitors, replicative reticulate bodies accumulated in enlarged inclusions but failed to express selected late-stage proteins and transition to elementary bodies, a Chlamydia developmental form that is required for invasion of mammalian cells. These findings suggest the presence of an outer membrane quality control system that regulates Chlamydia developmental transition to infectious elementary bodies and highlights the potential application of LpxC inhibitors as unique class of antichlamydial agents.


Subject(s)
Chlamydia trachomatis/pathogenicity , Inclusion Bodies/metabolism , Lipopolysaccharides/biosynthesis , Amidohydrolases/antagonists & inhibitors , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chlamydia Infections , Chlamydia trachomatis/cytology , Chlamydia trachomatis/physiology , HeLa Cells , Humans , Lipid A/biosynthesis , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Molecular Structure
5.
J Am Chem Soc ; 135(16): 6084-91, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23510488

ABSTRACT

Methonium (N(+)Me3) is an organic cation widely distributed in biological systems. As an organic cation, the binding of methonium to protein receptors requires the removal of a positive charge from water. The appearance of methonium in biological transmitters and receptors seems at odds with the large unfavorable desolvation free energy reported for tetramethylammonium (TMA(+)), a frequently utilized surrogate of methonium. Here, we report an experimental system that facilitates incremental internalization of methonium within the molecular cavity of cucurbit[7]uril (CB[7]). Using a combination of experimental and computational studies, we show that the transfer of methonium from bulk water (partially solvated methonium state) to the CB[7] cavity (mostly desolvated methonium state) is accompanied by a remarkably small desolvation enthalpy of just 0.5 ± 0.3 kcal·mol(-1), a value significantly less endothermic than those values suggested from gas-phase model studies. Our results are in accord with neutron scattering measurements that suggest methonium produces only a minimal perturbation in the bulk water structure, which highlights the limitations of gas-phase models. More surprisingly, the incremental withdrawal of the methonium surface from water produces a nonmonotonic response in desolvation enthalpy. A partially desolvated state exists, in which a portion of the methonium group remains exposed to solvent. This structure incurs an increased enthalpic penalty of ~3 kcal·mol(-1) compared to other solvation states. We attribute this observation to the pre-encapsulation dewetting of the methonium surface. Together, our results offer a rationale for the wide distribution of methonium in a biological context and suggest limitations to computational estimates of binding affinities based on simple parametrization of solvent-accessible surface area.


Subject(s)
Bis-Trimethylammonium Compounds/chemistry , Bridged-Ring Compounds/chemistry , Imidazoles/chemistry , Calorimetry , Choline/chemistry , Epitopes , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Neutrons , Protein Binding , Quaternary Ammonium Compounds/chemistry , Scattering, Radiation , Solvents , Surface Properties , Thermodynamics , Water/chemistry
6.
Sci Transl Med ; 15(708): eadf5668, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37556556

ABSTRACT

The UDP-3-O-(R-3-hydroxyacyl)-N-acetylglucosamine deacetylase LpxC is an essential enzyme in the biosynthesis of lipid A, the outer membrane anchor of lipopolysaccharide and lipooligosaccharide in Gram-negative bacteria. The development of LpxC-targeting antibiotics toward clinical therapeutics has been hindered by the limited antibiotic profile of reported non-hydroxamate inhibitors and unexpected cardiovascular toxicity observed in certain hydroxamate and non-hydroxamate-based inhibitors. Here, we report the preclinical characterization of a slow, tight-binding LpxC inhibitor, LPC-233, with low picomolar affinity. The compound is a rapid bactericidal antibiotic, unaffected by established resistance mechanisms to commercial antibiotics, and displays outstanding activity against a wide range of Gram-negative clinical isolates in vitro. It is orally bioavailable and efficiently eliminates infections caused by susceptible and multidrug-resistant Gram-negative bacterial pathogens in murine soft tissue, sepsis, and urinary tract infection models. It displays exceptional in vitro and in vivo safety profiles, with no detectable adverse cardiovascular toxicity in dogs at 100 milligrams per kilogram. These results establish the feasibility of developing oral LpxC-targeting antibiotics for clinical applications.


Subject(s)
Gram-Negative Bacteria , Lipid A , Animals , Mice , Dogs , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/chemistry
7.
Biochemistry ; 51(8): 1658-68, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22316217

ABSTRACT

The substrate specificity of enzymes is frequently narrow and constrained by multiple interactions, limiting the use of natural enzymes in biocatalytic applications. Aldolases have important synthetic applications, but the usefulness of these enzymes is hampered by their narrow reactivity profile with unnatural substrates. To explore the determinants of substrate selectivity and alter the specificity of Escherichia coli 2-keto-3-deoxy-6-phosphogluconate (KDPG) aldolase, we employed structure-based mutagenesis coupled with library screening of mutant enzymes localized to the bacterial periplasm. We identified two active site mutations (T161S and S184L) that work additively to enhance the substrate specificity of this aldolase to include catalysis of retro-aldol cleavage of (4S)-2-keto-4-hydroxy-4-(2'-pyridyl)butyrate (S-KHPB). These mutations improve the value of k(cat)/K(M)(S-KHPB) by >450-fold, resulting in a catalytic efficiency that is comparable to that of the wild-type enzyme with the natural substrate while retaining high stereoselectivity. Moreover, the value of k(cat)(S-KHPB) for this mutant enzyme, a parameter critical for biocatalytic applications, is 3-fold higher than the maximal value achieved by the natural aldolase with any substrate. This mutant also possesses high catalytic efficiency for the retro-aldol cleavage of the natural substrate, KDPG, and a >50-fold improved activity for cleavage of 2-keto-4-hydroxy-octonoate, a nonfunctionalized hydrophobic analogue. These data suggest a substrate binding mode that illuminates the origin of facial selectivity in aldol addition reactions catalyzed by KDPG and 2-keto-3-deoxy-6-phosphogalactonate aldolases. Furthermore, targeting mutations to the active site provides a marked improvement in substrate selectivity, demonstrating that structure-guided active site mutagenesis combined with selection techniques can efficiently identify proteins with characteristics that compare favorably to those of naturally occurring enzymes.


Subject(s)
Aldehyde-Lyases/chemistry , Escherichia coli/enzymology , Aldehyde-Lyases/metabolism , Binding Sites , Catalysis , Catalytic Domain , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Substrate Specificity
8.
Antimicrob Agents Chemother ; 56(1): 36-43, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21968370

ABSTRACT

Antibiotic therapy is the most commonly used strategy to control pathogenic infections; however, it has contributed to the generation of antibiotic-resistant bacteria. To circumvent this emerging problem, we are searching for compounds that target bacterial virulence factors rather than their viability. Pseudomonas aeruginosa, an opportunistic human pathogen, possesses a type III secretion system (T3SS) as one of the major virulence factors by which it secretes and translocates T3 effector proteins into human host cells. The fact that this human pathogen also is able to infect several plant species led us to screen a library of phenolic compounds involved in plant defense signaling and their derivatives for novel T3 inhibitors. Promoter activity screening of exoS, which encodes a T3-secreted toxin, identified two T3 inhibitors and two T3 inducers of P. aeruginosa PAO1. These compounds alter exoS transcription by affecting the expression levels of the regulatory small RNAs RsmY and RsmZ. These two small RNAs are known to control the activity of carbon storage regulator RsmA, which is responsible for the regulation of the key T3SS regulator ExsA. As RsmY and RsmZ are the only targets directly regulated by GacA, our results suggest that these phenolic compounds affect the expression of exoS through the GacSA-RsmYZ-RsmA-ExsA regulatory pathway.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Bacterial Secretion Systems/drug effects , Gene Expression Regulation, Bacterial/drug effects , Phenols/pharmacology , Pseudomonas aeruginosa/metabolism , Transcription Factors/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Genes, Regulator , Genes, Reporter , High-Throughput Screening Assays , Humans , Phenols/chemistry , Plant Extracts/chemistry , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription, Genetic/drug effects , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Bioorg Med Chem Lett ; 22(20): 6521-4, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22985855

ABSTRACT

Fragment based drug discovery remains a successful tool for pharmaceutical lead discovery. Although based upon the principle of thermodynamic additivity, the underlying thermodynamic basis is poorly understood. A thermodynamic additivity analysis was performed using stromelysin-1 and a series of biphenyl hydroxamate ligands identified through fragment additivity. Our studies suggest that, in this instance, additivity arises from enthalpic effects, while interaction entropies are unfavorable; this thermodynamic behavior is masked by proton transfer. Evaluation of the changes in constant pressure heat capacities during binding suggest that solvent exclusion from the binding site does not account for the dramatic affinity enhancements observed.


Subject(s)
Biphenyl Compounds/chemistry , Drug Design , Hydroxamic Acids/chemistry , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Thermodynamics , Binding Sites , Biphenyl Compounds/pharmacology , Humans , Hydroxamic Acids/pharmacology , Ligands , Matrix Metalloproteinase Inhibitors/pharmacology
10.
Proc Natl Acad Sci U S A ; 106(36): 15231-6, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19706892

ABSTRACT

The challenge in the synthesis of protein-polymer conjugates for biological applications is to synthesize a stoichiometric (typically 1:1) conjugate of the protein with a monodisperse polymer, with good retention of protein activity, significantly improved pharmacokinetics and increased bioavailability, and hence improved in vivo efficacy. Here we demonstrate, using myoglobin as an example, a general route to grow a PEG-like polymer, poly(oligo(ethylene glycol) methyl ether methacrylate) [poly(OEGMA)], with low polydispersity and high yield, solely from the N-terminus of the protein by in situ atom transfer radical polymerization (ATRP) under aqueous conditions, to yield a site-specific (N-terminal) and stoichiometric conjugate (1:1). Notably, the myoglobin-poly(OEGMA) conjugate [hydrodynamic radius (R(h)): 13 nm] showed a 41-fold increase in its blood exposure compared to the protein (R(h): 1.7 nm) after IV administration to mice, thereby demonstrating that comb polymers that present short oligo(ethylene glycol) side chains are a class of PEG-like polymers that can significantly improve the pharmacological properties of proteins. We believe that this approach to the synthesis of N-terminal protein conjugates of poly(OEGMA) may be applicable to a large subset of protein and peptide drugs, and thereby provide a general methodology for improvement of their pharmacological profiles.


Subject(s)
Acrylates/chemistry , Biopolymers/biosynthesis , Biopolymers/pharmacokinetics , Drug Discovery/methods , Myoglobin/chemistry , Polyethylene Glycols/chemistry , Biological Availability , Molecular Structure
11.
Biochemistry ; 50(4): 546-57, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21142040

ABSTRACT

Flavin-dependent histone demethylases catalyze the posttranslational oxidative demethylation of mono- and dimethylated lysine residues, producing formaldehyde and hydrogen peroxide in addition to the corresponding demethylated protein. In vivo, histone demethylase LSD1 (KDM1; BCH110) is a component of the multiprotein complex that includes histone deacetylases (HDAC 1 and 2) and the scaffolding protein CoREST. Although little is known about the affinities of or the structural basis for the interaction between CoREST and HDACs, the structure of CoREST(286-482) bound to an α-helical coiled-coil tower domain within LSD1 has recently been reported. Given the significance of CoREST in directing demethylation to specific nucleosomal substrates, insight into the molecular basis of the interaction between CoREST and LSD1 may suggest a new means of inhibiting LSD1 activity by misdirecting the enzyme away from nucleosomal substrates. Toward this end, isothermal titration calorimetry studies were conducted to determine the affinity and thermodynamic parameters characterizing the binding interaction between LSD1 and CoREST(286-482). The proteins tightly interact in a 1:1 stoichiometry with a dissociation constant (K(d)) of 15.9 ± 2.07 nM, and their binding interaction is characterized by a favorable enthalpic contribution near room temperature with a smaller entropic penalty at pH 7.4. Additionally, one proton is transferred from the buffer to the heterodimeric complex at pH 7.4. From the temperature dependence of the enthalpy change of interaction, a constant-pressure heat capacity change (ΔC(p)) of the interaction was determined to be -0.80 ± 0.01 kcal mol(-1) K(-1). Notably, structure-driven truncation of CoREST revealed that the central binding determinant lies within the segment of residues 293-380, also known as the CoREST "linker" region, which is a central isolated helix that interacts with the LSD1 coiled-coil tower domain to create a triple-helical bundle. Thermodynamic parameters obtained from the binding between LSD1 and the linker region of CoREST are similar to those obtained from the interaction between LSD1 and CoREST(286-482). These results provide a framework for understanding the molecular basis of protein-protein interactions that govern nucleosomal demethylation.


Subject(s)
Histone Demethylases/chemistry , Histone Demethylases/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Thermodynamics , Calorimetry , Co-Repressor Proteins , Entropy , Histone Demethylases/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Nerve Tissue Proteins/genetics , Nucleosomes/enzymology , Nucleosomes/genetics , Protein Binding/genetics , Protein Interaction Mapping/methods , Protein Structure, Tertiary/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Substrate Specificity/genetics , Surface Plasmon Resonance
12.
J Am Chem Soc ; 133(30): 11515-23, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21692482

ABSTRACT

In recent years, interfacial mobility has gained popularity as a model with which to rationalize both affinity in ligand binding and the often observed phenomenon of enthalpy-entropy compensation. While protein contraction and reduced mobility, as demonstrated by computational and NMR techniques respectively, have been correlated to entropies of binding for a variety of systems, to our knowledge, Raman difference spectroscopy has never been included in these analyses. Here, nonresonance Raman difference spectroscopy, isothermal titration calorimetry, and X-ray crystallography were utilized to correlate protein contraction, as demonstrated by an increase in protein interior packing and decreased residual protein movement, with trends of enthalpy-entropy compensation. These results are in accord with the interfacial mobility model and lend additional credence to this view of protein activity.


Subject(s)
Computer Simulation , Hydroxamic Acids/chemistry , Matrix Metalloproteinase 3/chemistry , Thermodynamics , Binding Sites , Computational Biology , Crystallography, X-Ray , Ligands , Matrix Metalloproteinase 3/isolation & purification , Models, Molecular , Molecular Structure , Spectrum Analysis, Raman , Stereoisomerism
13.
Langmuir ; 27(10): 6486-9, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21504221

ABSTRACT

Here we report a simple, robust approach to patterning functional SAMs on germanium. The protocol relies on catalytic soft-lithographic pattern transfer from an elastomeric stamp bearing pendant immobilized sulfonic acid moieties to an NHS-functionalized bilayer molecular system comprising a primary ordered alkyl monolayer and a reactive ester secondary overlayer. The catalytic polyurethane-acrylate stamp was used to form micrometer-scale features of chemically distinct SAMs on germanium. The methodology represents the first example of patterned SAMs on germanium, a semiconductor material.


Subject(s)
Germanium/chemistry , Succinimides/chemistry , Acrylates/chemistry , Catalysis , Polyurethanes/chemistry , Surface Properties
14.
Langmuir ; 27(10): 6478-85, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21520913

ABSTRACT

We report a simple, reliable high-throughput method for patterning passivated silicon with reactive organic monolayers and demonstrate selective functionalization of the patterned substrates with both small molecules and proteins. The approach completely protects silicon from chemical oxidation, provides precise control over the shape and size of the patterned features in the 100 nm domain, and gives rapid, ready access to chemically discriminated patterns that can be further functionalized with both organic and biological molecules.


Subject(s)
Nanotechnology/methods , Printing/methods , Silicon/chemistry , Acrylates/chemistry , Carboxylic Acids/chemistry , Catalysis , Hydrolysis , Oxidation-Reduction , Polyurethanes/chemistry , Reproducibility of Results , Succinimides/chemistry , Surface Properties
15.
Bioorg Med Chem ; 19(21): 6447-53, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21944547

ABSTRACT

The use of biological catalysts for industrial scale synthetic chemistry is highly attractive, given their cost effectiveness, high specificity that obviates the need for protecting group chemistry, and the environmentally benign nature of enzymatic procedures. Here we evolve the naturally occurring 2-keto-3-deoxy-6-phosphogluconate (KDPG) aldolases from Thermatoga maritima and Escherichia coli, into enzymes that recognize a nonfunctionalized electrophilic substrate, 2-keto-4-hydroxyoctonoate (KHO). Using an in vivo selection based on pyruvate auxotrophy, mutations were identified that lower the K(M) value up to 100-fold in E. coli KDPG aldolase, and that enhance the efficiency of retro-aldol cleavage of KHO by increasing the value of k(cat)/K(M) up to 25-fold in T. maritima KDPG aldolase. These data indicate that numerous mutations distal from the active site contribute to enhanced 'uniform binding' of the substrates, which is the first step in the evolution of novel catalytic activity.


Subject(s)
Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Caprylates/metabolism , Protein Engineering/methods , Aldehyde-Lyases/chemistry , Catalysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Directed Molecular Evolution/methods , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Peptide Library , Polymerase Chain Reaction
16.
Bioorg Med Chem ; 19(2): 852-60, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21194954

ABSTRACT

Compounds inhibiting LpxC in the lipid A biosynthetic pathway are promising leads for novel antibiotics against multidrug-resistant Gram-negative pathogens. We report the syntheses and structural and biochemical characterizations of LpxC inhibitors based on a diphenyl-diacetylene (1,4-diphenyl-1,3-butadiyne) threonyl-hydroxamate scaffold. These studies provide a molecular interpretation for the differential antibiotic activities of compounds with a substituted distal phenyl ring as well as the absolute stereochemical requirement at the C2, but not C3, position of the threonyl group.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Escherichia coli Proteins/antagonists & inhibitors , Amidohydrolases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Computer Simulation , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests , Molecular Sequence Data , Protein Structure, Tertiary , Structure-Activity Relationship
17.
Nano Lett ; 10(1): 43-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19950928

ABSTRACT

We report a new inkless catalytic muCP technique that achieves accurate, fast, and complete pattern reproduction on SAMs of Boc- and TBS-protected thiols immobilized on gold using a polyurethane-acrylate stamp functionalized with covalently bound sulfonic acids. Pattern transfer is complete at room temperature just after one minute of contact and renders sub-200 nm size structures of chemically differentiated SAMs.


Subject(s)
Acrylates/chemistry , Nanotechnology/methods , Polymers/chemistry , Sulfhydryl Compounds/chemistry , Catalysis , DNA/chemistry , Materials Testing , Metal Nanoparticles/chemistry , Microscopy, Atomic Force/methods , Microscopy, Electron, Scanning/methods , Oxygen/chemistry , Polyurethanes/chemistry , Sulfonic Acids/chemistry , Surface Properties , Temperature
18.
Biochemistry ; 49(19): 4244-54, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20392081

ABSTRACT

We developed a high-throughput yeast-based assay to screen for chemical inhibitors of Ca(2+)/calmodulin-dependent kinase pathways. After screening two small libraries, we identified the novel antagonist 125-C9, a substituted ethyleneamine. In vitro kinase assays confirmed that 125-C9 inhibited several calmodulin-dependent kinases (CaMKs) competitively with Ca(2+)/calmodulin (Ca(2+)/CaM). This suggested that 125-C9 acted as an antagonist for Ca(2+)/CaM rather than for CaMKs. We confirmed this hypothesis by showing that 125-C9 binds directly to Ca(2+)/CaM using isothermal titration calorimetry. We further characterized binding of 125-C9 to Ca(2+)/CaM and compared its properties with those of two well-studied CaM antagonists: trifluoperazine (TFP) and W-13. Isothermal titration calorimetry revealed that binding of 125-C9 to CaM is absolutely Ca(2+)-dependent, likely occurs with a stoichiometry of five 125-C9 molecules to one CaM molecule, and involves an exchange of two protons at pH 7.0. Binding of 125-C9 is driven overall by entropy and appears to be competitive with TFP and W-13, which is consistent with occupation of similar binding sites. To test the effects of 125-C9 in living cells, we evaluated mitogen-stimulated re-entry of quiescent cells into proliferation and found similar, although slightly better, levels of inhibition by 125-C9 than by TFP and W-13. Our results not only define a novel Ca(2+)/CaM inhibitor but also reveal that chemically unique CaM antagonists can bind CaM by distinct mechanisms but similarly inhibit cellular actions of CaM.


Subject(s)
Calmodulin/antagonists & inhibitors , Calmodulin/chemistry , Binding Sites , Hydrogen-Ion Concentration , Substrate Specificity , Sulfonamides/chemistry , Trifluoperazine/chemistry
19.
Langmuir ; 26(3): 1449-51, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-19950970

ABSTRACT

We report a novel inkless soft lithographic fabrication protocol that permits uniform parallel patterning of hydrogen-terminated silicon surfaces using catalytic elastomeric stamps. Pattern transfer is achieved catalytically via reaction between sulfonic acid moieties covalently bound to an elastomeric stamp and a Boc-functionalized SAM grafted to passivated silicon. The approach represents the first example of a soft lithographic printing technique that creates patterns of chemically distinctive SAMs on oxide-free silicon substrates.


Subject(s)
Hydrogen/chemistry , Silicon/chemistry , Catalysis , Hydrogen-Ion Concentration , Photoelectron Spectroscopy , Sulfonic Acids/chemistry , Surface Properties
20.
Bioorg Med Chem Lett ; 20(1): 280-2, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19942433

ABSTRACT

We describe a novel single-step method for the purification of stromelysin-1 catalytic domain (SCD) via immobilized metal affinity chromatography under denaturing conditions that inhibit proteolytic activity followed by on-column refolding and spontaneous autolysis of the fusion peptide to yield pure, active stromelysin-1 catalytic domain. The methodology provides a general approach for the rapid purification of large quantities of zinc proteinases.


Subject(s)
Matrix Metalloproteinase 3/isolation & purification , Autolysis , Catalytic Domain , Chromatography, Affinity , Histidine/chemistry , Imidazoles/chemistry , Oligopeptides/chemistry , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL