Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
mBio ; 13(1): e0322621, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35073739

ABSTRACT

The majority of drug discovery efforts against herpesviruses have focused on nucleoside analogs that target viral DNA polymerases, agents that are associated with dose-limiting toxicity and/or a narrow spectrum of activity. We are pursuing a strategy based on targeting two-metal ion-dependent (TMID) viral enzymes. This family of enzymes consists of structurally related proteins that share common active sites containing conserved carboxylates predicted to coordinate divalent cations essential for catalysis. Compounds that target TMID enzymes, such as HIV integrase and influenza endoribonuclease, have been successfully developed for clinical use. HIV integrase inhibitors have been reported to inhibit replication of herpes simplex virus (HSV) and other herpesviruses; however, the molecular targets of their antiviral activities have not been identified. We employed a candidate-based approach utilizing several two-metal-directed chemotypes and the potential viral TMID enzymatic targets in an effort to correlate target-based activity with antiviral potency. The panel of compounds tested included integrase inhibitors, the anti-influenza agent baloxavir, three natural products previously shown to exhibit anti-HSV activity, and two 8-hydroxyquinolines (8-HQs), AK-157 and AK-166, from our in-house program. The integrase inhibitors exhibited weak overall anti-HSV-1 activity, while the 8-HQs were shown to inhibit both HSV-1 and cytomegalovirus (CMV). Target-based analysis demonstrated that none of the antiviral compounds acted by inhibiting ICP8, contradicting previous reports. On the other hand, baloxavir inhibited the proofreading exonuclease of HSV polymerase, while AK-157 and AK-166 inhibited the alkaline exonuclease UL12. In addition, AK-157 also inhibited the catalytic activity of the HSV polymerase, which provides an opportunity to potentially develop dual-targeting agents against herpesviruses. IMPORTANCE Human herpesviruses (HHVs) establish lifelong latent infections, which undergo periodic reactivation and remain a major cause of morbidity and mortality, especially in immunocompromised individuals. Currently, HHV infections are treated primarily with agents that target viral DNA polymerase, including nucleoside analogs; however, long-term treatment can be complicated by the development of drug resistance. New therapies with novel modes of action would be important not only for the treatment of resistant viruses but also for use in combination therapy to reduce dose-limiting toxicities and potentially eliminate infection. Since many essential HHV proteins are well conserved, inhibitors of novel targets would ideally exhibit broad-spectrum activity against multiple HHVs.


Subject(s)
HIV Integrase Inhibitors , Herpesviridae , Herpesvirus 1, Human , Humans , Antiviral Agents/pharmacology , Nucleosides/pharmacology , Herpesvirus 1, Human/physiology , HIV Integrase Inhibitors/pharmacology , DNA-Directed DNA Polymerase/genetics , Exonucleases/pharmacology , Virus Replication
2.
Bioorg Med Chem Lett ; 53: 128414, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34666187

ABSTRACT

S-Palmitoylation is a reversible post-translational lipid modification that regulates protein trafficking and signaling. The enzymatic depalmitoylation of proteins is inhibited by the beta-lactones Palmostatin M and B, which have been found to target several serine hydrolases. In efforts to better understand the mechanism of action of Palmostatin M, we describe herein the synthesis, chemical proteomic analysis, and functional characterization of analogs of this compound. We identify Palmostatin M analogs that maintain inhibitory activity in N-Ras depalmitoylation assays while displaying complementary reactivity across the serine hydrolase class as measured by activity-based protein profiling. Active Palmostatin M analogs inhibit the recently characterized ABHD17 subfamily of depalmitoylating enzymes, while sparing other candidate depalmitoylases such as LYPLA1 and LYPLA2. These findings improve our understanding of the structure-activity relationship of Palmostatin M and refine the set of serine hydrolase targets relevant to the compound's effects on N-Ras palmitoylation dynamics.


Subject(s)
Lactones/analysis , Propiolactone/analogs & derivatives , Proteomics , Sulfones/analysis , ras Proteins/metabolism , Humans , Lactones/metabolism , Lactones/pharmacology , Molecular Structure , Propiolactone/analysis , Propiolactone/metabolism , Propiolactone/pharmacology , Sulfones/metabolism , Sulfones/pharmacology , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry
3.
Sci Rep ; 9(1): 17106, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31745118

ABSTRACT

Many years ago, the natural secondary metabolite SF2312, produced by the actinomycete Micromonospora, was reported to display broad spectrum antibacterial properties against both Gram-positive and Gram-negative bacteria. Recent studies have revealed that SF2312, a natural phosphonic acid, functions as a potent inhibitor of human enolase. The mechanism of SF2312 inhibition of bacterial enolase and its role in bacterial growth and reproduction, however, have remained elusive. In this work, we detail a structural analysis of E. coli enolase bound to both SF2312 and its oxidized imide-form. Our studies support a model in which SF2312 acts as an analog of a high energy intermediate formed during the catalytic process. Biochemical, biophysical, computational and kinetic characterization of these compounds confirm that altering features characteristic of a putative carbanion (enolate) intermediate significantly reduces the potency of enzyme inhibition. When SF2312 is combined with fosfomycin in the presence of glucose-6 phosphate, significant synergy is observed. This suggests the two agents could be used as a potent combination, targeting distinct cellular mechanism for the treatment of bacterial infections. Together, our studies rationalize the structure-activity relationships for these phosphonates and validate enolase as a promising target for antibiotic discovery.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Organophosphonates/pharmacology , Phosphopyruvate Hydratase/antagonists & inhibitors , Phosphopyruvate Hydratase/metabolism , Pyrrolidinones/pharmacology , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Phosphopyruvate Hydratase/chemistry , Protein Conformation , Sequence Homology , Structure-Activity Relationship
4.
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
5.
Acta Crystallogr Sect E Struct Rep Online ; 67(Pt 12): o3151, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22199677

ABSTRACT

In the cation of the title mol-ecular salt, C(18)H(13)N(4)O(+)·Cl(-)·H(2)O, the dihedral angle between the mean planes of the 1,10-phenanthroline system and the phenol ring is 14.40 (19)°. The crystal packing is stabilized by O-H⋯O hydrogen bonds, weak N-H⋯Cl and O-H⋯Cl inter-molecular inter-actions and π-π stacking inter-actions [centroid-centroid distance = 3.6944 (13) and 3.9702 (12) Å].

SELECTION OF CITATIONS
SEARCH DETAIL
...