Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Nat Commun ; 14(1): 4798, 2023 08 09.
Article En | MEDLINE | ID: mdl-37558718

UBA1 is the primary E1 ubiquitin-activating enzyme responsible for generation of activated ubiquitin required for ubiquitination, a process that regulates stability and function of numerous proteins. Decreased or insufficient ubiquitination can cause or drive aging and many diseases. Therefore, a small-molecule enhancing UBA1 activity could have broad therapeutic potential. Here we report that auranofin, a drug approved for the treatment of rheumatoid arthritis, is a potent UBA1 activity enhancer. Auranofin binds to the UBA1's ubiquitin fold domain and conjugates to Cys1039 residue. The binding enhances UBA1 interactions with at least 20 different E2 ubiquitin-conjugating enzymes, facilitating ubiquitin charging to E2 and increasing the activities of seven representative E3s in vitro. Auranofin promotes ubiquitination and degradation of misfolded ER proteins during ER-associated degradation in cells at low nanomolar concentrations. It also facilitates outer mitochondrial membrane-associated degradation. These findings suggest that auranofin can serve as a much-needed tool for UBA1 research and therapeutic exploration.


Ubiquitin-Conjugating Enzymes , Ubiquitin , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Auranofin/pharmacology , Ubiquitination , Ubiquitin-Activating Enzymes/metabolism
2.
Mol Biol Cell ; 33(13): ar120, 2022 11 01.
Article En | MEDLINE | ID: mdl-36074076

RNF5 E3 ubiquitin ligase has multiple biological roles and has been linked to the development of severe diseases such as cystic fibrosis, acute myeloid leukemia, and certain viral infections, emphasizing the importance of discovering small-molecule RNF5 modulators for research and drug development. The present study describes the synthesis of a new benzo[b]thiophene derivative, FX12, that acts as a selective small-molecule inhibitor and degrader of RNF5. We initially identified the previously reported STAT3 inhibitor, Stattic, as an inhibitor of dislocation of misfolded proteins from the endoplasmic reticulum (ER) lumen to the cytosol in ER-associated degradation. A concise structure-activity relationship campaign (SAR) around the Stattic chemotype led to the synthesis of FX12, which has diminished activity in inhibition of STAT3 activation and retains dislocation inhibitory activity. FX12 binds to RNF5 and inhibits its E3 activity in vitro as well as promoting proteasomal degradation of RNF5 in cells. RNF5 as a molecular target for FX12 was supported by the facts that FX12 requires RNF5 to inhibit dislocation and negatively regulates RNF5 function. Thus, this study developed a small-molecule inhibitor and degrader of the RNF5 ubiquitin ligase, providing a chemical biology tool for RNF5 research and therapeutic development.


DNA-Binding Proteins , Ubiquitin , Cyclic S-Oxides , DNA-Binding Proteins/metabolism , Thiophenes/pharmacology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
ACS Pharmacol Transl Sci ; 5(7): 468-478, 2022 Jul 08.
Article En | MEDLINE | ID: mdl-35821746

The COVID-19 pandemic has had enormous health, economic, and social consequences. Vaccines have been successful in reducing rates of infection and hospitalization, but there is still a need for acute treatment of the disease. We investigate whether compounds that bind the human angiotensin-converting enzyme 2 (ACE2) protein can decrease SARS-CoV-2 replication without impacting ACE2's natural enzymatic function. Initial screening of a diversity library resulted in hit compounds active in an ACE2-binding assay, which showed little inhibition of ACE2 enzymatic activity (116 actives, success rate ∼4%), suggesting they were allosteric binders. Subsequent application of in silico techniques boosted success rates to ∼14% and resulted in 73 novel confirmed ACE2 binders with K d values as low as 6 nM. A subsequent SARS-CoV-2 assay revealed that five of these compounds inhibit the viral life cycle in human cells. Further effort is required to completely elucidate the antiviral mechanism of these ACE2-binders, but they present a valuable starting point for both the development of acute treatments for COVID-19 and research into the host-directed therapy.

4.
bioRxiv ; 2022 Mar 16.
Article En | MEDLINE | ID: mdl-35313579

The COVID-19 pandemic has had enormous health, economic, and social consequences. Vaccines have been successful in reducing rates of infection and hospitalization, but there is still a need for an acute treatment for the disease. We investigate whether compounds that bind the human ACE2 protein can interrupt SARS-CoV-2 replication without damaging ACE2’s natural enzymatic function. Initial compounds were screened for binding to ACE2 but little interruption of ACE2 enzymatic activity. This set of compounds was extended by application of quantitative structure-activity analysis, which resulted in 512 virtual hits for further confirmatory screening. A subsequent SARS-CoV-2 replication assay revealed that five of these compounds inhibit SARS-CoV-2 replication in human cells. Further effort is required to completely determine the antiviral mechanism of these compounds, but they serve as a strong starting point for both development of acute treatments for COVID-19 and research into the mechanism of infection.

5.
ACS Pharmacol Transl Sci ; 4(5): 1675-1688, 2021 Oct 08.
Article En | MEDLINE | ID: mdl-34608449

The National Center for Advancing Translational Sciences (NCATS) has been actively generating SARS-CoV-2 high-throughput screening data and disseminates it through the OpenData Portal (https://opendata.ncats.nih.gov/covid19/). Here, we provide a hybrid approach that utilizes NCATS screening data from the SARS-CoV-2 cytopathic effect reduction assay to build predictive models, using both machine learning and pharmacophore-based modeling. Optimized models were used to perform two iterative rounds of virtual screening to predict small molecules active against SARS-CoV-2. Experimental testing with live virus provided 100 (∼16% of predicted hits) active compounds (efficacy > 30%, IC50 ≤ 15 µM). Systematic clustering analysis of active compounds revealed three promising chemotypes which have not been previously identified as inhibitors of SARS-CoV-2 infection. Further investigation resulted in the identification of allosteric binders to host receptor angiotensin-converting enzyme 2; these compounds were then shown to inhibit the entry of pseudoparticles bearing spike protein of wild-type SARS-CoV-2, as well as South African B.1.351 and UK B.1.1.7 variants.

6.
J Med Chem ; 64(13): 9431-9443, 2021 07 08.
Article En | MEDLINE | ID: mdl-34184537

The majority of FDA-approved HCV therapeutics target the viral replicative machinery. An automated high-throughput phenotypic screen identified several small molecules as potent inhibitors of hepatitis C virus replication. Here, we disclose the discovery and optimization of a 4-aminopiperidine (4AP) scaffold targeting the assembly stages of the HCV life cycle. The original screening hit (1) demonstrates efficacy in the HCVcc assay but does not show potency prior to or during viral replication. Colocalization and infectivity studies indicate that the 4AP chemotype inhibits the assembly and release of infectious HCV. Compound 1 acts synergistically with FDA-approved direct-acting antiviral compounds Telaprevir and Daclatasvir, as well as broad spectrum antivirals Ribavirin and cyclosporin A. Following an SAR campaign, several derivatives of the 4AP series have been identified with increased potency against HCV, reduced in vitro toxicity, as well as improved in vitro and in vivo ADME properties.


Antiviral Agents/pharmacology , Drug Discovery , Hepacivirus/drug effects , Piperidines/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Male , Mice , Microbial Sensitivity Tests , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Rats , Structure-Activity Relationship , Virus Replication/drug effects
7.
J Med Chem ; 64(4): 2291-2309, 2021 02 25.
Article En | MEDLINE | ID: mdl-33573376

A novel diazaspiro[3.4]octane series was identified from a Plasmodium falciparum whole-cell high-throughput screening campaign. Hits displayed activity against multiple stages of the parasite lifecycle, which together with a novel sp3-rich scaffold provided an attractive starting point for a hit-to-lead medicinal chemistry optimization and biological profiling program. Structure-activity-relationship studies led to the identification of compounds that showed low nanomolar asexual blood-stage activity (<50 nM) together with strong gametocyte sterilizing properties that translated to transmission-blocking activity in the standard membrane feeding assay. Mechanistic studies through resistance selection with one of the analogues followed by whole-genome sequencing implicated the P. falciparum cyclic amine resistance locus in the mode of resistance.


Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Spiro Compounds/pharmacology , Animals , Anopheles/drug effects , Antimalarials/chemical synthesis , Antimalarials/metabolism , Female , Germ Cells/drug effects , High-Throughput Screening Assays , Humans , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Rats , Spiro Compounds/chemical synthesis , Spiro Compounds/metabolism , Structure-Activity Relationship
8.
Sci Rep ; 11(1): 2121, 2021 01 22.
Article En | MEDLINE | ID: mdl-33483532

The spread of Plasmodium falciparum parasites resistant to most first-line antimalarials creates an imperative to enrich the drug discovery pipeline, preferably with curative compounds that can also act prophylactically. We report a phenotypic quantitative high-throughput screen (qHTS), based on concentration-response curves, which was designed to identify compounds active against Plasmodium liver and asexual blood stage parasites. Our qHTS screened over 450,000 compounds, tested across a range of 5 to 11 concentrations, for activity against Plasmodium falciparum asexual blood stages. Active compounds were then filtered for unique structures and drug-like properties and subsequently screened in a P. berghei liver stage assay to identify novel dual-active antiplasmodial chemotypes. Hits from thiadiazine and pyrimidine azepine chemotypes were subsequently prioritized for resistance selection studies, yielding distinct mutations in P. falciparum cytochrome b, a validated antimalarial drug target. The thiadiazine chemotype was subjected to an initial medicinal chemistry campaign, yielding a metabolically stable analog with sub-micromolar potency. Our qHTS methodology and resulting dataset provides a large-scale resource to investigate Plasmodium liver and asexual blood stage parasite biology and inform further research to develop novel chemotypes as causal prophylactic antimalarials.


Antimalarials/pharmacology , High-Throughput Screening Assays/methods , Liver/drug effects , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Drug Evaluation, Preclinical/methods , Hep G2 Cells , Humans , Liver/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Protective Agents/chemistry , Protective Agents/pharmacology , Reproducibility of Results , Structure-Activity Relationship , Thiadiazines/chemistry , Thiadiazines/pharmacology
9.
ChemMedChem ; 16(4): 654-661, 2021 02 17.
Article En | MEDLINE | ID: mdl-33211374

Bacterial expression of ß-lactamases, which hydrolyze ß-lactam antibiotics, contributes to the growing threat of antibacterial drug resistance. Metallo-ß-lactamases, such as NDM-1, use catalytic zinc ions in their active sites and hydrolyze nearly all clinically available ß-lactam antibiotics. Inhibitors of metallo-ß-lactamases are urgently needed to overcome this resistance mechanism. Zinc-binding compounds are promising leads for inhibitor development, as many NDM-1 inhibitors contain zinc-binding pharmacophores. Here, we evaluated 13 chelating agents containing benzimidazole and benzoxazole scaffolds as NDM-1 inhibitors. Six of the compounds showed potent inhibitory activity with IC50 values as low as 0.38 µM, and several compounds restored the meropenem susceptibility of NDM-1-expressing E. coli. Spectroscopic and docking studies suggest ternary complex formation as the mechanism of inhibition, making these compounds promising for development as NDM-1 inhibitors.


Anti-Bacterial Agents/pharmacology , Chelating Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , beta-Lactamases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Zinc/chemistry , Zinc/pharmacology
10.
Nat Microbiol ; 5(12): 1532-1541, 2020 12.
Article En | MEDLINE | ID: mdl-32868923

Fluoxazolevir is an aryloxazole-based entry inhibitor of hepatitis C virus (HCV). We show that fluoxazolevir inhibits fusion of HCV with hepatic cells by binding HCV envelope protein 1 to prevent fusion. Nine of ten fluoxazolevir resistance-associated substitutions are in envelope protein 1, and four are in a putative fusion peptide. Pharmacokinetic studies in mice, rats and dogs revealed that fluoxazolevir localizes to the liver. A 4-week intraperitoneal regimen of fluoxazolevir in humanized chimeric mice infected with HCV genotypes 1b, 2a or 3 resulted in a 2-log reduction in viraemia, without evidence of drug resistance. In comparison, daclatasvir, an approved HCV drug, suppressed more than 3 log of viraemia but is associated with the emergence of resistance-associated substitutions in mice. Combination therapy using fluoxazolevir and daclatasvir cleared HCV genotypes 1b and 3 in mice. Fluoxazolevir combined with glecaprevir and pibrentasvir was also effective in clearing multidrug-resistant HCV replication in mice. Fluoxazolevir may be promising as the next generation of combination drug cocktails for HCV treatment.


Antiviral Agents/administration & dosage , Hepacivirus/drug effects , Hepatitis C/drug therapy , Virus Internalization/drug effects , Animals , Carbamates/administration & dosage , Disease Models, Animal , Dogs , Drug Therapy, Combination , Genotype , Hepacivirus/classification , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C/virology , Humans , Imidazoles/administration & dosage , Male , Mice , Pyrrolidines/administration & dosage , Rats , Rats, Sprague-Dawley , Valine/administration & dosage , Valine/analogs & derivatives , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
11.
Cell Chem Biol ; 27(7): 780-792.e5, 2020 07 16.
Article En | MEDLINE | ID: mdl-32386595

Chlorcyclizine (CCZ) is a potent hepatitis C virus (HCV) entry inhibitor, but its molecular mechanism is unknown. Here, we show that CCZ directly targets the fusion peptide of HCV E1 and interferes with the fusion process. Generation of CCZ resistance-associated substitutions of HCV in vitro revealed six missense mutations in the HCV E1 protein, five being in the putative fusion peptide. A viral fusion assay demonstrated that CCZ blocked HCV entry at the membrane fusion step and that the mutant viruses acquired resistance to CCZ's action in blocking membrane fusion. UV cross-linking of photoactivatable CCZ-diazirine-biotin in both HCV-infected cells and recombinant HCV E1/E2 protein demonstrated direct binding to HCV E1 glycoprotein. Mass spectrometry analysis revealed that CCZ cross-linked to an E1 sequence adjacent to the putative fusion peptide. Docking simulations demonstrate a putative binding model, wherein CCZ binds to a hydrophobic pocket of HCV E1 and forms extensive interactions with the fusion peptide.


Hepacivirus/metabolism , Piperazines/chemistry , Viral Envelope Proteins/antagonists & inhibitors , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Binding Sites , Biotin/chemistry , Diazomethane/chemistry , Drug Resistance, Viral/drug effects , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Humans , Membrane Fusion/drug effects , Molecular Docking Simulation , Piperazines/metabolism , Piperazines/pharmacology , Ultraviolet Rays , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects
12.
Bioorg Med Chem Lett ; 26(4): 1196-9, 2016 Feb 15.
Article En | MEDLINE | ID: mdl-26804234

Several novel compounds have been identified that inhibit the replication of hepatitis C virus in a replicon assay with EC50 values as low as 0.6 µM. Lead compounds were modified to investigate the possible role that zinc binding may play in inhibitor efficacy. In addition, the structure-activity relationship was explored to increase inhibitor efficacy and possibly identify favorable interactions within the currently unknown inhibitor binding pocket. The rationale for inhibitor design and biological results are presented herein.


Antiviral Agents/chemistry , Hepacivirus/physiology , Zinc/metabolism , Antiviral Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Humans , Inhibitory Concentration 50 , Naphthols/chemistry , Naphthols/pharmacology , Protein Binding , Quinolines/chemistry , Quinolines/pharmacology , Structure-Activity Relationship , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Zinc/chemistry
13.
J Porphyr Phthalocyanines ; 19(4): 547-572, 2015 Apr.
Article En | MEDLINE | ID: mdl-26640361

Advances in chlorin synthetic chemistry now enable the de novo preparation of diverse chlorin-containing molecular architectures. Five distinct molecular designs have been explored here, including hydrophobic bioconjugatable (oxo)chlorins; a hydrophilic bioconjugatable chlorin; a trans-ethynyl/iodochlorin building block; a set of chlorins bearing electron-rich (methoxy, dimethylamino, methylthio) groups at the 3-position; and a set of ten 3,13-disubstituted chlorins chiefly bearing groups with extended π-moieties. Altogether 23 new chlorins (17 targets, 6 intermediates) have been prepared. The challenge associated with molecular designs that encompass the combination of "hydrophilic, bioconjugatable and wavelength-tunable" chiefly resides in the nature of the hydrophilic unit.

14.
Bioorg Med Chem Lett ; 24(2): 609-12, 2014 Jan 15.
Article En | MEDLINE | ID: mdl-24360997

The bacterial natural product UK-1 and several structural analogs inhibit replication of the hepatitis C virus in the replicon assay, with IC50 values as low as 0.50 µM. The NS3 helicase has been identified as a possible target of inhibition for several of these compounds, while the remaining inhibitors act via an undetermined mechanism. Gel shift assays suggest that helicase inhibition is a direct result of inhibitor-enzyme binding as opposed to direct RNA binding, and the ATPase activity of NS3 is not affected. The syntheses and biological results are presented herein.


Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Virus Replication/drug effects , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Dose-Response Relationship, Drug , Hepacivirus/physiology , Humans , RNA Helicases/antagonists & inhibitors , RNA Helicases/physiology , Virus Replication/physiology
...