Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Biochemistry ; 58(9): 1188-1197, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30714720

ABSTRACT

Enolase is a glycolytic metalloenzyme involved in carbon metabolism. The advantage of targeting enolase lies in its essentiality in many biological processes such as cell wall formation and RNA turnover and as a plasminogen receptor. We initially used a DARTS assay to identify enolase as a target in Escherichia coli. The antibacterial activities of α-, ß-, and γ-substituted seven-member ring tropolones were first evaluated against four strains representing a range of Gram-negative bacteria. We observed that the chemical properties and position of the substituents on the tropolone ring play an important role in the biological activity of the investigated compounds. Both α- and ß-substituted phenyl derivatives of tropolone were the most active with minimum inhibitory concentrations in the range of 11-14 µg/mL. The potential inhibitory activity of the synthetic tropolones was further evaluated using an enolase inhibition assay, X-ray crystallography, and molecular docking simulations. The catalytic activity of enolase was effectively inhibited by both the naturally occurring ß-thujaplicin and the α- and ß-substituted phenyl derivatives of tropolones with IC50 values in range of 8-11 µM. Ligand binding parameters were assessed by isothermal titration calorimetry and differential scanning calorimetry techniques and agreed with the in vitro data. Our studies validate the antibacterial potential of tropolones with careful consideration of the position and character of chelating moieties for stronger interaction with metal ions and residues in the enolase active site.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Gram-Negative Bacteria/drug effects , Phosphopyruvate Hydratase/antagonists & inhibitors , Tropolone/pharmacology , Anti-Bacterial Agents/chemistry , Calorimetry , Catalytic Domain , Crystallography, X-Ray , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gram-Negative Bacteria/enzymology , Microbial Sensitivity Tests , Molecular Docking Simulation , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Conformation , Structure-Activity Relationship , Tropolone/chemistry
2.
Article in English | MEDLINE | ID: mdl-28314188

ABSTRACT

Antimicrobial resistance to current antibiotics is a significant public health problem and the need for new antibiotics is a compelling one. We have been developing a new series of antibiotics, propargyl-linked diaminopyrimidines, based on the structure of trimethoprim. To date we have discovered compounds that are effective inhibitors of dihydrofolate reductase (the target of trimethoprim), that are potent antibiotics in vitro against a range of Gram-positive pathogens including methicillin-resistant S. aureus, and that are non-toxic in mammalian cell culture. In this study we report the development of an LC-MS-based protocol for the quantification of our lead antibiotic 37D1-UCP1099 and the application of this assay to follow the concentration of the compound in mouse plasma after intraperitoneal administration. Extraction of 37D1-UCP1099 from mouse plasma was achieved through a liquid-liquid extraction with ethyl acetate. Separation was performed utilizing a reverse-phase C18 column with a ten minute isocratic elution using 47:53 (v/v) 10mM NH4HCO3:acetonitrile. The lower limit of quantitation for 37D1-UCP1099 was 50ngmL-1 and the assay showed a dynamic range of 50-4000ngmL-1 with good linearity (r2≥0.996 for all fits). Intra-day and inter-day precision and accuracy were within 11.3% (%RSD) and 6.6% (%RE) respectably. We have demonstrated that the compound is stable under the assay procedures. The compound was shown to have a mean residence time of 26.2±1.0min and a half-life of 18.2±0.7min after intraperitoneal delivery at 5mgkg-1. These studies now form the foundation of our work to develop additional analogs of 37D1-UCP1099 with improved pharmacokinetic properties.


Subject(s)
Anti-Bacterial Agents/blood , Chromatography, High Pressure Liquid/methods , Folic Acid Antagonists/blood , Liquid-Liquid Extraction/methods , Animals , Anti-Bacterial Agents/administration & dosage , Female , Folic Acid Antagonists/administration & dosage , Injections, Intraperitoneal , Limit of Detection , Mice , Spectrometry, Mass, Electrospray Ionization/methods
3.
Org Lett ; 19(1): 142-145, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27959567

ABSTRACT

To develop next generation antifolates for the treatment of trimethoprim-resistant bacteria, synthetic methods were needed to prepare a diverse array of 3-aryl-propynes with various substitutions at the propargyl position. A direct route was sought whereby nucleophilic addition of acetylene to aryl carboxaldehydes would be followed by reduction or substitution of the resulting propargyl alcohol. The direct reduction, methylation, and dimethylation of these readily available alcohols provide efficient access to this uncommon functional array. In addition, an unusual silane exchange reaction was observed in the reduction of the propargylic alcohols.


Subject(s)
Alcohols/chemical synthesis , Alkynes/chemistry , Anti-Bacterial Agents/chemistry , Folic Acid Antagonists/chemistry , Aldehydes/chemistry , Alkynes/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Drug Design , Drug Resistance, Bacterial , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Humans , Methylation , Molecular Structure , Oxidation-Reduction , Stereoisomerism , Structure-Activity Relationship , Trimethoprim/pharmacology
4.
Methods Mol Biol ; 1529: 291-306, 2017.
Article in English | MEDLINE | ID: mdl-27914058

ABSTRACT

Drug resistance in protein targets is an increasingly common phenomenon that reduces the efficacy of both existing and new antibiotics. However, knowledge of future resistance mutations during pre-clinical phases of drug development would enable the design of novel antibiotics that are robust against not only known resistant mutants, but also against those that have not yet been clinically observed. Computational structure-based protein design (CSPD) is a transformative field that enables the prediction of protein sequences with desired biochemical properties such as binding affinity and specificity to a target. The use of CSPD to predict previously unseen resistance mutations represents one of the frontiers of computational protein design. In a recent study (Reeve et al. Proc Natl Acad Sci U S A 112(3):749-754, 2015), we used our OSPREY (Open Source Protein REdesign for You) suite of CSPD algorithms to prospectively predict resistance mutations that arise in the active site of the dihydrofolate reductase enzyme from methicillin-resistant Staphylococcus aureus (SaDHFR) in response to selective pressure from an experimental competitive inhibitor. We demonstrated that our top predicted candidates are indeed viable resistant mutants. Since that study, we have significantly enhanced the capabilities of OSPREY with not only improved modeling of backbone flexibility, but also efficient multi-state design, fast sparse approximations, partitioned continuous rotamers for more accurate energy bounds, and a computationally efficient representation of molecular-mechanics and quantum-mechanical energy functions. Here, using SaDHFR as an example, we present a protocol for resistance prediction using the latest version of OSPREY. Specifically, we show how to use a combination of positive and negative design to predict active site escape mutations that maintain the enzyme's catalytic function but selectively ablate binding of an inhibitor.


Subject(s)
Computational Biology/methods , Drug Resistance/genetics , Mutation , Protein Engineering/methods , Proteins/chemistry , Proteins/genetics , Software , Algorithms , Amino Acid Sequence , Databases, Genetic , Models, Molecular , Pharmacogenetics/methods , Protein Conformation , Web Browser
5.
Cell Chem Biol ; 23(12): 1458-1467, 2016 Dec 22.
Article in English | MEDLINE | ID: mdl-27939900

ABSTRACT

Antibiotic resistance is a rapidly evolving health concern that requires a sustained effort to understand mechanisms of resistance and to develop new agents that overcome those mechanisms. The dihydrofolate reductase (DHFR) inhibitor, trimethoprim (TMP), remains one of the most important orally administered antibiotics. However, resistance through chromosomal mutations and mobile, plasmid-encoded insensitive DHFRs threatens the continued use of this agent. We are pursuing the development of new propargyl-linked antifolate (PLA) DHFR inhibitors designed to evade these mechanisms. While analyzing contemporary TMP-resistant clinical isolates of methicillin-resistant and sensitive Staphylococcus aureus, we discovered two mobile resistance elements, dfrG and dfrK. This is the first identification of these resistance mechanisms in the United States. These resistant organisms were isolated from a variety of infection sites, show clonal diversity, and each contain distinct resistance genotypes for common antibiotics. Several PLAs showed significant activity against these resistant strains by direct inhibition of the TMP resistance elements.

6.
Pharmacol Res ; 113(Pt A): 438-448, 2016 11.
Article in English | MEDLINE | ID: mdl-27663262

ABSTRACT

Tropolones, such as ß-thujaplicin, are small lead-like natural products that possess a variety of biological activities. While the ß-substituted natural products and their synthetic analogs are potent inhibitors of human cancer cell growth, less is known about their α-substituted counterparts. Recently, we synthesized a series of α-substituted tropolones including 2-hydroxy-7-(naphthalen-2-yl)cyclohepta-2,4,6-trien-1-one (α-naphthyl tropolone). Here, we evaluate the antiproliferative mechanisms of α-naphthyl tropolone and the related α-benzodioxinyl analog. The α-substituted tropolones inhibit growth of lymphocytic leukemia cells, but not healthy blood cells, with nanomolar potency. Treatment of leukemia cell lines with the tropolone dose-dependently induces apoptosis as judged by staining with annexin V and propidium iodide and Western blot analysis of cleaved caspase 3 and 7. Moreover, pre-treatment of cells with the caspase inhibitor Z-VAD-FMK inhibited the apoptotic effects of the tropolone in two lymphocytic lines. Caspase inhibition also blocked elevated histone acetylation caused by the tropolone, indicating that its effects on histone acetylation are potentiated by caspases. In contrast, α-naphthyl tropolone upregulated p53 expression and phosphorylation of Akt and mTOR in a manner that was not rescued by caspase inhibition. The effects of tropolone were blocked by co-incubation with high levels of free extracellular iron but not by pre-loading with iron. Additionally, dose and time dependent reduction in ex vivo viability of cells from leukemia patients was observed. Taken together, we demonstrate that α-substituted tropolones upregulate DNA damage repair pathways leading to caspase-dependent apoptosis in malignant lymphocytes.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Leukemia/drug therapy , Tropolone/pharmacology , Acetylation/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Repair/drug effects , Histones/metabolism , Humans , Leukemia/metabolism , Leukocytes, Mononuclear , Monoterpenes/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Tropolone/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
7.
ACS Infect Dis ; 2(2): 149-56, 2016 02 12.
Article in English | MEDLINE | ID: mdl-27624966

ABSTRACT

Multidrug-resistant Enterobacteriaceae, notably Escherichia coli and Klebsiella pneumoniae, have become major health concerns worldwide. Resistance to effective therapeutics is often carried by class I and II integrons that can confer insensitivity to carbapenems, extended spectrum ß-lactamases, the antifolate trimethoprim, fluoroquinolones, and aminoglycosides. Specifically of interest to the study here, a prevalent gene (dfrA1) coding for an insensitive dihydrofolate reductase (DHFR) confers 190- or 1000-fold resistance to trimethoprim for K. pneumoniae and E. coli, respectively. Attaining inhibition of both the wild-type and resistant forms of the enzyme is critical for new antifolates. For several years, we have been developing the propargyl-linked antifolates (PLAs) as effective inhibitors against trimethoprim-resistant DHFR enzymes. Here, we show that the PLAs are active against both the wild-type and DfrA1 DHFR proteins. We report two high-resolution crystal structures of DfrA1 bound to potent PLAs. The structure-activity relationships and crystal structures will be critical in driving the design of broadly active inhibitors against wild-type and resistant DHFR.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/chemistry , Trimethoprim Resistance/drug effects , Trimethoprim/pharmacology , Bacterial Proteins/chemistry , Crystallography, X-Ray , Escherichia coli/enzymology , Humans , Integrons , Klebsiella pneumoniae/enzymology , Structure-Activity Relationship , beta-Lactamases/metabolism
8.
PLoS One ; 11(8): e0161740, 2016.
Article in English | MEDLINE | ID: mdl-27580226

ABSTRACT

Mycobacterium tuberculosis continues to cause widespread, life-threatening disease. In the last decade, this threat has grown dramatically as multi- and extensively-drug resistant (MDR and XDR) bacteria have spread globally and the number of agents that effectively treat these infections is significantly reduced. We have been developing the propargyl-linked antifolates (PLAs) as potent inhibitors of the essential enzyme dihydrofolate reductase (DHFR) from bacteria and recently found that charged PLAs with partial zwitterionic character showed improved mycobacterial cell permeability. Building on a hypothesis that these PLAs may penetrate the outer membrane of M. tuberculosis and inhibit the essential cytoplasmic DHFR, we screened a group of PLAs for antitubercular activity. In this work, we identified several PLAs as potent inhibitors of the growth of M. tuberculosis with several of the compounds exhibiting minimum inhibition concentrations equal to or less than 1 µg/mL. Furthermore, two of the compounds were very potent inhibitors of MDR and XDR strains. A high resolution crystal structure of one PLA bound to DHFR from M. tuberculosis reveals the interactions of the ligands with the target enzyme.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins , Drug Resistance, Multiple, Bacterial/drug effects , Folic Acid Antagonists/chemistry , Mycobacterium tuberculosis/enzymology , Tetrahydrofolate Dehydrogenase/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Humans
9.
ACS Med Chem Lett ; 7(7): 692-6, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27437079

ABSTRACT

Although classical, negatively charged antifolates such as methotrexate possess high affinity for the dihydrofolate reductase (DHFR) enzyme, they are unable to penetrate the bacterial cell wall, rendering them poor antibacterial agents. Herein, we report a new class of charged propargyl-linked antifolates that capture some of the key contacts common to the classical antifolates while maintaining the ability to passively diffuse across the bacterial cell wall. Eight synthesized compounds exhibit extraordinary potency against Gram-positive S. aureus with limited toxicity against mammalian cells and good metabolic profile. High resolution crystal structures of two of the compounds reveal extensive interactions between the carboxylate and active site residues through a highly organized water network.

10.
Article in English | MEDLINE | ID: mdl-27352799

ABSTRACT

The folate cycle is one of the key metabolic pathways used by bacteria to synthesize vital building blocks required for proliferation. Therapeutic agents targeting enzymes in this cycle, such as trimethoprim and sulfamethoxazole, are among some of the most important and continually used antibacterials to treat both Gram-positive and Gram-negative pathogens. As with all antibacterial agents, the emergence of resistance threatens the continued clinical use of these life-saving drugs. In this article, we describe and analyze resistance mechanisms that have been clinically observed and review newer generations of preclinical compounds designed to overcome the molecular basis of the resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Folic Acid Antagonists/pharmacology , Drug Resistance, Bacterial/drug effects , Gram-Negative Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/drug therapy , Humans , Sulfamethoxazole/pharmacology , Trimethoprim Resistance
11.
J Med Chem ; 59(13): 6493-500, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27308944

ABSTRACT

Drug-resistant enzymes must balance catalytic function with inhibitor destabilization to provide a fitness advantage. This sensitive balance, often involving very subtle structural changes, must be achieved through a selection process involving a minimal number of eligible point mutations. As part of a program to design propargyl-linked antifolates (PLAs) against trimethoprim-resistant dihydrofolate reductase (DHFR) from Staphylococcus aureus, we have conducted a thorough study of several clinically observed chromosomal mutations in the enzyme at the cellular, biochemical, and structural levels. Through this work, we have identified a promising lead series that displays significantly greater activity against these mutant enzymes and strains than TMP. The best inhibitors have enzyme inhibition and MIC values near or below that of trimethoprim against wild-type S. aureus. Moreover, these studies employ a series of crystal structures of several mutant enzymes bound to the same inhibitor; analysis of the structures reveals a more detailed molecular understanding of drug resistance in this important enzyme.


Subject(s)
Anti-Bacterial Agents/pharmacology , Folic Acid Antagonists/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Trimethoprim/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Folic Acid/metabolism , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/chemistry , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Structure-Activity Relationship
13.
Future Microbiol ; 10(11): 1727-33, 2015.
Article in English | MEDLINE | ID: mdl-26516790

ABSTRACT

Understanding the structural basis of antibacterial resistance may enable rational design principles that avoid and subvert that resistance, thus leading to the discovery of more effective antibiotics. In this review, we explore the use of crystal structures to guide new discovery of antibiotics that are effective against resistant organisms. Structures of efflux pumps bound to substrates and inhibitors have aided the design of compounds with lower affinity for the pump or inhibitors that more effectively block the pump. Structures of ß-lactamase enzymes have revealed the mechanisms of action toward key carbapenems and structures of gyrase have aided the design of compounds that are less susceptible to point mutations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Proteins/chemistry , Drug Discovery/methods , Drug Resistance, Bacterial , Anti-Bacterial Agents/isolation & purification , Bacterial Proteins/genetics , Crystallography, X-Ray , Humans , Molecular Docking Simulation , Protein Conformation
14.
J Am Chem Soc ; 137(28): 8983-90, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26098608

ABSTRACT

While antifolates such as Bactrim (trimethoprim-sulfamethoxazole; TMP-SMX) continue to play an important role in treating community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA), resistance-conferring mutations, specifically F98Y of dihydrofolate reductase (DHFR), have arisen and compromise continued use. In an attempt to extend the lifetime of this important class, we have developed a class of propargyl-linked antifolates (PLAs) that exhibit potent inhibition of the enzyme and bacterial strains. Probing the role of the configuration at the single propargylic stereocenter in these inhibitors required us to develop a new approach to nonracemic 3-aryl-1-butyne building blocks by the pairwise use of asymmetric conjugate addition and aldehyde dehydration protocols. Using this new route, a series of nonracemic PLA inhibitors was prepared and shown to possess potent enzyme inhibition (IC50 values <50 nM), antibacterial effects (several with MIC values <1 µg/mL) and to form stable ternary complexes with both wild-type and resistant mutants. Unexpectedly, crystal structures of a pair of individual enantiomers in the wild-type DHFR revealed that the single change in configuration of the stereocenter drove the selection of an alternative NADPH cofactor, with the minor α-anomer appearing with R-27. Remarkably, this cofactor switching becomes much more prevalent when the F98Y mutation is present. The observation of cofactor site plasticity leads to a postulate for the structural basis of TMP resistance in DHFR and also suggests design strategies that can be used to target these resistant enzymes.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , Tetrahydrofolate Dehydrogenase/metabolism , Crystallography, X-Ray , Drug Design , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Models, Molecular , Point Mutation , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Stereoisomerism , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/genetics
15.
Biochemistry ; 54(17): 2719-26, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25871808

ABSTRACT

We report the first Raman spectroscopic study of propargyl-linked dihydrofolate reductase (DHFR) inhibitors being taken up by wild type Escherichia coli, Klebsiella pneumoniae, and Staphylococcus aureus cells. A novel protocol is developed where cells are exposed to the fermentation medium containing a known amount of an inhibitor. At a chosen time point, the cells are centrifuged and washed to remove the extracellular compound, then frozen and freeze-dried. Raman difference spectra of the freeze-dried cells (cells exposed to the drug minus cells alone) provide spectra of the compounds inside the cells, where peak intensities allow us to quantify the number of inhibitors within each cell. A time course for the propargyl-linked DHFR inhibitor UCP 1038 soaking into E. coli cells showed that penetration occurs very quickly and reaches a plateau after 10 min exposure to the inhibitor. After 10 min drug exposure, the populations of two inhibitors, UCP 1038 and UCP 1089, were ~1.5 × 10(6) molecules in each E. coli cell, ~4.7 × 10(5) molecules in each K. pneumonia cell, and ~2.7 × 10(6) in each S. aureus cell. This is the first in situ comparison of inhibitor population in Gram-negative and Gram-positive bacterial cells. The positions of the Raman peaks also reveal the protonation of diaminopyrimidine ring upon binding to DHFR inside cells. The spectroscopic signature of protonation was characterized by binding an inhibitor to a single crystal of DHFR.


Subject(s)
Escherichia coli/metabolism , Folic Acid Antagonists/pharmacokinetics , Klebsiella pneumoniae/metabolism , Microscopy/methods , Spectrum Analysis, Raman/methods , Staphylococcus aureus/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Crystallography, X-Ray , Tetrahydrofolate Dehydrogenase/chemistry
16.
Proc Natl Acad Sci U S A ; 112(3): 749-54, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25552560

ABSTRACT

Methods to accurately predict potential drug target mutations in response to early-stage leads could drive the design of more resilient first generation drug candidates. In this study, a structure-based protein design algorithm (K* in the OSPREY suite) was used to prospectively identify single-nucleotide polymorphisms that confer resistance to an experimental inhibitor effective against dihydrofolate reductase (DHFR) from Staphylococcus aureus. Four of the top-ranked mutations in DHFR were found to be catalytically competent and resistant to the inhibitor. Selection of resistant bacteria in vitro reveals that two of the predicted mutations arise in the background of a compensatory mutation. Using enzyme kinetics, microbiology, and crystal structures of the complexes, we determined the fitness of the mutant enzymes and strains, the structural basis of resistance, and the compensatory relationship of the mutations. To our knowledge, this work illustrates the first application of protein design algorithms to prospectively predict viable resistance mutations that arise in bacteria under antibiotic pressure.


Subject(s)
Algorithms , Folic Acid Antagonists/pharmacology , Proteins/chemistry , Drug Resistance/genetics , Polymorphism, Single Nucleotide , Staphylococcus aureus/enzymology , Tetrahydrofolate Dehydrogenase/drug effects
17.
Antimicrob Agents Chemother ; 58(12): 7484-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25288083

ABSTRACT

Resistance to the antibacterial antifolate trimethoprim (TMP) is increasing in members of the family Enterobacteriaceae, driving the design of next-generation antifolates effective against these Gram-negative pathogens. The propargyl-linked antifolates are potent inhibitors of dihydrofolate reductases (DHFR) from several TMP-sensitive and -resistant species, including Klebsiella pneumoniae. Recently, we have determined that these antifolates inhibit the growth of strains of K. pneumoniae, some with MIC values of 1 µg/ml. In order to further the design of potent and selective antifolates against members of the Enterobacteriaceae, we determined the first crystal structures of K. pneumoniae DHFR bound to two of the propargyl-linked antifolates. These structures highlight that interactions with Leu 28, Ile 50, Ile 94, and Leu 54 are necessary for potency; comparison with structures of human DHFR bound to the same inhibitors reveal differences in residues (N64E, P61G, F31L, and V115I) and loop conformations (residues 49 to 53) that may be exploited for selectivity.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Folic Acid Antagonists/chemistry , Klebsiella pneumoniae/chemistry , Tetrahydrofolate Dehydrogenase/chemistry , Trimethoprim/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Klebsiella pneumoniae/enzymology , Molecular Docking Simulation , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/genetics , Trimethoprim Resistance/genetics
18.
Bioorg Med Chem ; 22(7): 2188-93, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24613456

ABSTRACT

Thujaplicins are tropolone-derived natural products with antiproliferative properties. We recently reported that certain tropolones potently and selectively target histone deacetylases (HDAC) and inhibit the growth of hematological cell lines. Here, we investigated the mechanisms by which these compounds exert their antiproliferative activity in comparison with the pan-selective HDAC inhibitor, vorinostat, using Jurkat T-cell leukemia cells. The tropolones appear to work through a mechanism distinct from vorinostat. These studies suggest that tropolone derivatives may serve as selective epigenetic modulators of hematological cells with potential applications as anti-leukemic or anti-inflammatory agents.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, T-Cell/drug therapy , Tropolone/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Jurkat Cells , Leukemia, T-Cell/pathology , Molecular Structure , Structure-Activity Relationship , Tropolone/chemical synthesis , Tropolone/chemistry , Tumor Cells, Cultured
19.
Expert Opin Ther Pat ; 24(6): 687-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24655343

ABSTRACT

INTRODUCTION: The folate biosynthetic pathway, responsible for the de novo synthesis of thymidine and other key cellular components, is essential in all life forms and is especially critical in rapidly proliferating cells. As such, druggable targets along this pathway offer opportunities to impact many disease states such as cancer, infectious disease and autoimmune disease. In this article, recent progress on the development of antifolate compounds is reviewed. AREAS COVERED: The evaluation of the patent literature during the period 2010 - 2013 focused on any compounds inhibiting recognized targets on the folate biosynthetic pathway. EXPERT OPINION: The folate pathway constitutes a well-validated and well-characterized set of targets; this pathway continues to elicit considerable enthusiasm for new drug discovery from both academic and industrial pharmaceutical research groups. Within the pathway, the enzymes dihydrofolate reductase and thymidylate synthase persist as the most attractive targets for new drug discovery for the treatment of cancer and infectious disease. Importantly, new potential targets for antifolates such as those on the purine biosynthetic pathway have been recently explored. The use of structure-based drug design is a major aspect in modern approaches to these drug targets.


Subject(s)
Folic Acid Antagonists/pharmacology , Patents as Topic , Animals , Drug Design , Drug Discovery , Folate Receptor 1/antagonists & inhibitors , Folic Acid Antagonists/therapeutic use , Humans , Tetrahydrofolate Dehydrogenase/physiology , Thymidylate Synthase/antagonists & inhibitors
20.
J Med Chem ; 57(6): 2643-56, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24568657

ABSTRACT

Species of Candida, primarily C. albicans and with increasing prevalence, C. glabrata, are responsible for the majority of fungal bloodstream infections that cause morbidity, especially among immune compromised patients. While the development of new antifungal agents that target the essential enzyme, dihydrofolate reductase (DHFR), in both Candida species would be ideal, previous attempts have resulted in antifolates that exhibit inconsistencies between enzyme inhibition and antifungal properties. In this article, we describe the evaluation of pairs of propargyl-linked antifolates that possess similar physicochemical properties but different shapes. All of these compounds are effective at inhibiting the fungal enzymes and the growth of C. glabrata; however, the inhibition of the growth of C. albicans is shape-dependent with extended para-linked compounds proving more effective than compact, meta-linked compounds. Using crystal structures of DHFR from C. albicans and C. glabrata bound to lead compounds, 13 new para-linked compounds designed to inhibit both species were synthesized. Eight of these compounds potently inhibit the growth of both fungal species with three compounds displaying dual MIC values less than 1 µg/mL. Analysis of the active compounds shows that shape and distribution of polar functionality is critical in achieving dual antifungal activity.


Subject(s)
Antifungal Agents , Candida albicans/drug effects , Candida glabrata/drug effects , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Morphinans/chemical synthesis , Morphinans/pharmacology , Chromatography, High Pressure Liquid , Crystallization , Crystallography, X-Ray , Folic Acid Antagonists/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Morphinans/chemistry , NADP/chemistry , Solubility , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/drug effects , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...