Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nat Chem Biol ; 20(3): 365-372, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37828400

ABSTRACT

Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.


Subject(s)
Adaptor Proteins, Signal Transducing , Membrane Proteins , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Biological Assay , Cytosol , Immunity, Innate , Ligands , Membrane Proteins/metabolism
2.
Nat Chem Biol ; 18(4): 412-421, 2022 04.
Article in English | MEDLINE | ID: mdl-35210618

ABSTRACT

Many diseases are driven by proteins that are aberrantly ubiquitinated and degraded. These diseases would be therapeutically benefited by targeted protein stabilization (TPS). Here we present deubiquitinase-targeting chimeras (DUBTACs), heterobifunctional small molecules consisting of a deubiquitinase recruiter linked to a protein-targeting ligand, to stabilize the levels of specific proteins degraded in a ubiquitin-dependent manner. Using chemoproteomic approaches, we discovered the covalent ligand EN523 that targets a non-catalytic allosteric cysteine C23 in the K48-ubiquitin-specific deubiquitinase OTUB1. We showed that a DUBTAC consisting of our EN523 OTUB1 recruiter linked to lumacaftor, a drug used to treat cystic fibrosis that binds ΔF508-cystic fibrosis transmembrane conductance regulator (CFTR), robustly stabilized ΔF508-CFTR protein levels, leading to improved chloride channel conductance in human cystic fibrosis bronchial epithelial cells. We also demonstrated stabilization of the tumor suppressor kinase WEE1 in hepatoma cells. Our study showcases covalent chemoproteomic approaches to develop new induced proximity-based therapeutic modalities and introduces the DUBTAC platform for TPS.


Subject(s)
Cystic Fibrosis , Chimera/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/therapeutic use , Humans , Ligands , Ubiquitin/metabolism
3.
Chembiochem ; 24(11): e202300116, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37069799

ABSTRACT

While vaccines and antivirals are now being deployed for the current SARS-CoV-2 pandemic, we require additional antiviral therapeutics to not only effectively combat SARS-CoV-2 and its variants, but also future coronaviruses. All coronaviruses have relatively similar genomes that provide a potential exploitable opening to develop antiviral therapies that will be effective against all coronaviruses. Among the various genes and proteins encoded by all coronaviruses, one particularly "druggable" or relatively easy-to-drug target is the coronavirus Main Protease (3CLpro or Mpro), an enzyme that is involved in cleaving a long peptide translated by the viral genome into its individual protein components that are then assembled into the virus to enable viral replication in the cell. Inhibiting Mpro with a small-molecule antiviral would effectively stop the ability of the virus to replicate, providing therapeutic benefit. In this study, we have utilized activity-based protein profiling (ABPP)-based chemoproteomic approaches to discover and further optimize cysteine-reactive pyrazoline-based covalent inhibitors for the SARS-CoV-2 Mpro. Structure-guided medicinal chemistry and modular synthesis of di- and tri-substituted pyrazolines bearing either chloroacetamide or vinyl sulfonamide cysteine-reactive warheads enabled the expedient exploration of structure-activity relationships (SAR), yielding nanomolar potency inhibitors against Mpro from not only SARS-CoV-2, but across many other coronaviruses. Our studies highlight promising chemical scaffolds that may contribute to future pan-coronavirus inhibitors.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Cysteine , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Molecular Docking Simulation
4.
J Am Chem Soc ; 144(2): 701-708, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34994556

ABSTRACT

Proteolysis-targeting chimeras (PROTACs), heterobifunctional compounds that consist of protein-targeting ligands linked to an E3 ligase recruiter, have arisen as a powerful therapeutic modality for targeted protein degradation (TPD). Despite the popularity of TPD approaches in drug discovery, only a small number of E3 ligase recruiters are available for the >600 E3 ligases that exist in human cells. Here, we have discovered a cysteine-reactive covalent ligand, EN106, that targets FEM1B, an E3 ligase recently discovered as the critical component of the cellular response to reductive stress. By targeting C186 in FEM1B, EN106 disrupts recognition of the key reductive stress substrate of FEM1B, FNIP1. We further establish that EN106 can be used as a covalent recruiter for FEM1B in TPD applications by demonstrating that a PROTAC linking EN106 to the BET bromodomain inhibitor JQ1 or the kinase inhibitor dasatinib leads to the degradation of BRD4 and BCR-ABL, respectively. Our study showcases a covalent ligand that targets a natural E3 ligase-substrate binding site and highlights the utility of covalent ligand screening in expanding the arsenal of E3 ligase recruiters suitable for TPD applications.


Subject(s)
Acetamides/chemistry , Cell Cycle Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligase Complexes/metabolism , Animals , Azepines/chemistry , Binding Sites , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , Cysteine/chemistry , Dasatinib/chemistry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Humans , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Triazoles/chemistry , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/genetics
5.
J Am Chem Soc ; 144(50): 22890-22901, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36484997

ABSTRACT

Activity-based protein profiling (ABPP) is a versatile strategy for identifying and characterizing functional protein sites and compounds for therapeutic development. However, the vast majority of ABPP methods for covalent drug discovery target highly nucleophilic amino acids such as cysteine or lysine. Here, we report a methionine-directed ABPP platform using Redox-Activated Chemical Tagging (ReACT), which leverages a biomimetic oxidative ligation strategy for selective methionine modification. Application of ReACT to oncoprotein cyclin-dependent kinase 4 (CDK4) as a representative high-value drug target identified three new ligandable methionine sites. We then synthesized a methionine-targeting covalent ligand library bearing a diverse array of heterocyclic, heteroatom, and stereochemically rich substituents. ABPP screening of this focused library identified 1oxF11 as a covalent modifier of CDK4 at an allosteric M169 site. This compound inhibited kinase activity in a dose-dependent manner on purified protein and in breast cancer cells. Further investigation of 1oxF11 found prominent cation-π and H-bonding interactions stabilizing the binding of this fragment at the M169 site. Quantitative mass-spectrometry studies validated 1oxF11 ligation of CDK4 in breast cancer cell lysates. Further biochemical analyses revealed cross-talk between M169 oxidation and T172 phosphorylation, where M169 oxidation prevented phosphorylation of the activating T172 site on CDK4 and blocked cell cycle progression. By identifying a new mechanism for allosteric methionine redox regulation on CDK4 and developing a unique modality for its therapeutic intervention, this work showcases a generalizable platform that provides a starting point for engaging in broader chemoproteomics and protein ligand discovery efforts to find and target previously undruggable methionine sites.


Subject(s)
Breast Neoplasms , Methionine , Humans , Female , Cyclin-Dependent Kinase 4/metabolism , Ligands , Phosphorylation , Oxidation-Reduction , Racemethionine/metabolism
6.
Nat Chem Biol ; 16(11): 1189-1198, 2020 11.
Article in English | MEDLINE | ID: mdl-32572277

ABSTRACT

Molecular glues are an intriguing therapeutic modality that harness small molecules to induce interactions between proteins that typically do not interact. However, such molecules are rare and have been discovered fortuitously, thus limiting their potential as a general strategy for therapeutic intervention. We postulated that natural products bearing one or more electrophilic sites may be an unexplored source of new molecular glues, potentially acting through multicovalent attachment. Using chemoproteomic platforms, we show that members of the manumycin family of polyketides, which bear multiple potentially reactive sites, target C374 of the putative E3 ligase UBR7 in breast cancer cells, and engage in molecular glue interactions with the neosubstrate tumor-suppressor TP53, leading to p53 transcriptional activation and cell death. Our results reveal an anticancer mechanism of this natural product family, and highlight the potential for combining chemoproteomics and multicovalent natural products for the discovery of new molecular glues.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Polyenes/chemistry , Polyketides/chemistry , Polyunsaturated Alkamides/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cross-Linking Reagents/chemistry , Drug Discovery , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Molecular Conformation , Molecular Structure , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , Static Electricity , Structure-Activity Relationship , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
7.
Nat Chem Biol ; 15(7): 747-755, 2019 07.
Article in English | MEDLINE | ID: mdl-31209351

ABSTRACT

Nimbolide, a terpenoid natural product derived from the Neem tree, impairs cancer pathogenicity; however, the direct targets and mechanisms by which nimbolide exerts its effects are poorly understood. Here, we used activity-based protein profiling (ABPP) chemoproteomic platforms to discover that nimbolide reacts with a novel functional cysteine crucial for substrate recognition in the E3 ubiquitin ligase RNF114. Nimbolide impairs breast cancer cell proliferation in-part by disrupting RNF114-substrate recognition, leading to inhibition of ubiquitination and degradation of tumor suppressors such as p21, resulting in their rapid stabilization. We further demonstrate that nimbolide can be harnessed to recruit RNF114 as an E3 ligase in targeted protein degradation applications and show that synthetically simpler scaffolds are also capable of accessing this unique reactive site. Our study highlights the use of ABPP platforms in uncovering unique druggable modalities accessed by natural products for cancer therapy and targeted protein degradation applications.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biological Products/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Limonins/pharmacology , Proteolysis/drug effects , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Biological Products/chemistry , Biological Products/isolation & purification , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Limonins/chemistry , Limonins/isolation & purification , Ubiquitin-Protein Ligases
8.
Nat Chem Biol ; 15(2): 179-188, 2019 02.
Article in English | MEDLINE | ID: mdl-30643281

ABSTRACT

The identification of activating mutations in NOTCH1 in 50% of T cell acute lymphoblastic leukemia has generated interest in elucidating how these mutations contribute to oncogenic transformation and in targeting the pathway. A phenotypic screen identified compounds that interfere with trafficking of Notch and induce apoptosis via an endoplasmic reticulum (ER) stress mechanism. Target identification approaches revealed a role for SLC39A7 (ZIP7), a zinc transport family member, in governing Notch trafficking and signaling. Generation and sequencing of a compound-resistant cell line identified a V430E mutation in ZIP7 that confers transferable resistance to the compound NVS-ZP7-4. NVS-ZP7-4 altered zinc in the ER, and an analog of the compound photoaffinity labeled ZIP7 in cells, suggesting a direct interaction between the compound and ZIP7. NVS-ZP7-4 is the first reported chemical tool to probe the impact of modulating ER zinc levels and investigate ZIP7 as a novel druggable node in the Notch pathway.


Subject(s)
Cation Transport Proteins/genetics , Endoplasmic Reticulum Stress/physiology , Receptor, Notch1/genetics , Animals , Apoptosis , Carrier Proteins/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Cell Line , Cell Transformation, Neoplastic , Endoplasmic Reticulum/physiology , Humans , Mutation , Protein Transport , Receptor, Notch1/physiology , Signal Transduction , Zinc/metabolism
10.
J Cell Sci ; 128(6): 1217-29, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25616894

ABSTRACT

A new cyclic decadepsipeptide was isolated from Chaetosphaeria tulasneorum with potent bioactivity on mammalian and yeast cells. Chemogenomic profiling in S. cerevisiae indicated that the Sec61 translocon complex, the machinery for protein translocation and membrane insertion at the endoplasmic reticulum, is the target. The profiles were similar to those of cyclic heptadepsipeptides of a distinct chemotype (including HUN-7293 and cotransin) that had previously been shown to inhibit cotranslational translocation at the mammalian Sec61 translocon. Unbiased, genome-wide mutagenesis followed by full-genome sequencing in both fungal and mammalian cells identified dominant mutations in Sec61p (yeast) or Sec61α1 (mammals) that conferred resistance. Most, but not all, of these mutations affected inhibition by both chemotypes, despite an absence of structural similarity. Biochemical analysis confirmed inhibition of protein translocation into the endoplasmic reticulum of both co- and post-translationally translocated substrates by both chemotypes, demonstrating a mechanism independent of a translating ribosome. Most interestingly, both chemotypes were found to also inhibit SecYEG, the bacterial Sec61 translocon homolog. We suggest 'decatransin' as the name for this new decadepsipeptide translocation inhibitor.


Subject(s)
Biological Products/pharmacology , Endoplasmic Reticulum/drug effects , Membrane Proteins/metabolism , Protein Transport/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Ascomycota/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , HCT116 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Polymorphism, Single Nucleotide/genetics , SEC Translocation Channels , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
11.
Nat Chem Biol ; 11(7): 511-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26030728

ABSTRACT

Spinal muscular atrophy (SMA), which results from the loss of expression of the survival of motor neuron-1 (SMN1) gene, represents the most common genetic cause of pediatric mortality. A duplicate copy (SMN2) is inefficiently spliced, producing a truncated and unstable protein. We describe herein a potent, orally active, small-molecule enhancer of SMN2 splicing that elevates full-length SMN protein and extends survival in a severe SMA mouse model. We demonstrate that the molecular mechanism of action is via stabilization of the transient double-strand RNA structure formed by the SMN2 pre-mRNA and U1 small nuclear ribonucleic protein (snRNP) complex. The binding affinity of U1 snRNP to the 5' splice site is increased in a sequence-selective manner, discrete from constitutive recognition. This new mechanism demonstrates the feasibility of small molecule-mediated, sequence-selective splice modulation and the potential for leveraging this strategy in other splicing diseases.


Subject(s)
Alternative Splicing , Muscular Atrophy, Spinal/drug therapy , RNA, Double-Stranded/agonists , Ribonucleoprotein, U1 Small Nuclear/agonists , Small Molecule Libraries/pharmacology , Survival of Motor Neuron 2 Protein/metabolism , Animals , Binding Sites , Disease Models, Animal , Female , Gene Expression , Humans , Mice , Mice, Transgenic , Models, Molecular , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/pathology , Protein Binding/drug effects , Protein Stability/drug effects , Proteolysis , RNA Precursors/agonists , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Survival Analysis , Survival of Motor Neuron 2 Protein/chemistry , Survival of Motor Neuron 2 Protein/genetics
12.
Nat Chem Biol ; 10(8): 623-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24929529

ABSTRACT

Identification and validation of drug-resistant mutations can provide important insights into the mechanism of action of a compound. Here we demonstrate the feasibility of such an approach in mammalian cells using next-generation sequencing of drug-resistant clones and CRISPR-Cas9-mediated gene editing on two drug-target pairs, 6-thioguanine-HPRT1 and triptolide-ERCC3. We showed that disrupting functional HPRT1 allele or introducing ERCC3 point mutations by gene editing can confer drug resistance in cells.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Endonucleases/genetics , High-Throughput Nucleotide Sequencing/methods , Animals , Cell Line/drug effects , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Diterpenes/pharmacology , Drug Resistance/drug effects , Drug Resistance/genetics , Epoxy Compounds/pharmacology , HCT116 Cells , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Mammals , Phenanthrenes/pharmacology , Point Mutation , Reproducibility of Results , Thioguanine/pharmacology
13.
Nat Chem Biol ; 10(5): 343-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24633354

ABSTRACT

Hedgehog (Hh) signaling determines cell fate during development and can drive tumorigenesis. We performed a screen for new compounds that can impinge on Hh signaling downstream of Smoothened (Smo). A series of cyclohexyl-methyl aminopyrimidine chemotype compounds ('CMAPs') were identified that could block pathway signaling in a Smo-independent manner. In addition to inhibiting Hh signaling, the compounds generated inositol phosphates through an unknown GPCR. Correlation of GPCR mRNA expression levels with compound activity across cell lines suggested the target to be the orphan receptor GPR39. RNA interference or cDNA overexpression of GPR39 demonstrated that the receptor is necessary for compound activity. We propose a model in which CMAPs activate GPR39, which signals to the Gli transcription factors and blocks signaling. In addition to the discovery of GPR39 as a new target that impinges on Hh signaling, we report on small-molecule modulators of the receptor that will enable in vitro interrogation of GPR39 signaling in different cellular contexts.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Chromatography, Affinity , Proteomics , Signal Transduction , Tandem Mass Spectrometry
14.
Nature ; 461(7264): 614-20, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19759537

ABSTRACT

The stability of the Wnt pathway transcription factor beta-catenin is tightly regulated by the multi-subunit destruction complex. Deregulated Wnt pathway activity has been implicated in many cancers, making this pathway an attractive target for anticancer therapies. However, the development of targeted Wnt pathway inhibitors has been hampered by the limited number of pathway components that are amenable to small molecule inhibition. Here, we used a chemical genetic screen to identify a small molecule, XAV939, which selectively inhibits beta-catenin-mediated transcription. XAV939 stimulates beta-catenin degradation by stabilizing axin, the concentration-limiting component of the destruction complex. Using a quantitative chemical proteomic approach, we discovered that XAV939 stabilizes axin by inhibiting the poly-ADP-ribosylating enzymes tankyrase 1 and tankyrase 2. Both tankyrase isoforms interact with a highly conserved domain of axin and stimulate its degradation through the ubiquitin-proteasome pathway. Thus, our study provides new mechanistic insights into the regulation of axin protein homeostasis and presents new avenues for targeted Wnt pathway therapies.


Subject(s)
Repressor Proteins/metabolism , Signal Transduction/drug effects , Tankyrases/antagonists & inhibitors , Wnt Proteins/antagonists & inhibitors , Axin Protein , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteomics , Repressor Proteins/chemistry , Tankyrases/metabolism , Transcription, Genetic/drug effects , Ubiquitin/metabolism , Ubiquitination , Wnt Proteins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
15.
Proc Natl Acad Sci U S A ; 108(17): 6739-44, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21502533

ABSTRACT

The search for novel therapeutic interventions for viral disease is a challenging pursuit, hallmarked by the paucity of antiviral agents currently prescribed. Targeting of viral proteins has the inextricable challenge of rise of resistance. Safe and effective vaccines are not possible for many viral pathogens. New approaches are required to address the unmet medical need in this area. We undertook a cell-based high-throughput screen to identify leads for development of drugs to treat respiratory syncytial virus (RSV), a serious pediatric pathogen. We identified compounds that are potent (nanomolar) inhibitors of RSV in vitro in HEp-2 cells and in primary human bronchial epithelial cells and were shown to act postentry. Interestingly, two scaffolds exhibited broad-spectrum activity among multiple RNA viruses. Using the chemical matter as a probe, we identified the targets and identified a common cellular pathway: the de novo pyrimidine biosynthesis pathway. Both targets were validated in vitro and showed no significant cell cytotoxicity except for activity against proliferative B- and T-type lymphoid cells. Corollary to this finding was to understand the consequences of inhibition of the target to the host. An in vivo assessment for antiviral efficacy failed to demonstrate reduced viral load, but revealed microscopic changes and a trend toward reduced pyrimidine pools and findings in histopathology. We present here a discovery program that includes screen, target identification, validation, and druggability that can be broadly applied to identify and interrogate other host factors for antiviral effect starting from chemical matter of unknown target/mechanism of action.


Subject(s)
Antiviral Agents , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/metabolism , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Proliferation/drug effects , Chlorocebus aethiops , Dogs , Dose-Response Relationship, Drug , HeLa Cells , Humans , Jurkat Cells , Respiratory Syncytial Virus Infections/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology , Vero Cells
16.
ACS Chem Biol ; 19(6): 1260-1270, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38739449

ABSTRACT

Ophiobolin A (OPA) is a sesterterpenoid fungal natural product with broad anticancer activity. While OPA possesses multiple electrophilic moieties that can covalently react with nucleophilic amino acids on proteins, the proteome-wide targets and mechanism of OPA remain poorly understood in many contexts. In this study, we used covalent chemoproteomic platforms to map the proteome-wide reactivity of the OPA in a highly sensitive lung cancer cell line. Among several proteins that OPA engaged, we focused on two targets: lysine-72 of cytochrome c oxidase subunit 5A (COX5A) and cysteine-53 of mitochondrial hypoxia induced gene 1 domain family member 2A (HIGD2A). These two subunit proteins are part of complex IV (cytochrome C oxidase) within the electron transport chain and contributed significantly to the antiproliferative activity of OPA. OPA activated mitochondrial respiration in a COX5A- and HIGD2A-dependent manner, leading to an initial spike in mitochondrial ATP and heightened mitochondrial oxidative stress. OPA compromised mitochondrial membrane potential, ultimately leading to ATP depletion. We have used chemoproteomic strategies to discover a unique anticancer mechanism of OPA through activation of complex IV leading to compromised mitochondrial energetics and rapid cell death.


Subject(s)
Electron Transport Complex IV , Mitochondria , Sesterterpenes , Humans , Sesterterpenes/pharmacology , Sesterterpenes/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Electron Transport Complex IV/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Oxidative Stress/drug effects , Membrane Potential, Mitochondrial/drug effects , Adenosine Triphosphate/metabolism , Cell Proliferation/drug effects
17.
Antimicrob Agents Chemother ; 57(5): 2272-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23478965

ABSTRACT

High-throughput phenotypic screening against the yeast Saccharomyces cerevisiae revealed a series of triazolopyrimidine-sulfonamide compounds with broad-spectrum antifungal activity, no significant cytotoxicity, and low protein binding. To elucidate the target of this series, we have applied a chemogenomic profiling approach using the S. cerevisiae deletion collection. All compounds of the series yielded highly similar profiles that suggested acetolactate synthase (Ilv2p, which catalyzes the first common step in branched-chain amino acid biosynthesis) as a possible target. The high correlation with profiles of known Ilv2p inhibitors like chlorimuron-ethyl provided further evidence for a similar mechanism of action. Genome-wide mutagenesis in S. cerevisiae identified 13 resistant clones with 3 different mutations in the catalytic subunit of acetolactate synthase that also conferred cross-resistance to established Ilv2p inhibitors. Mapping of the mutations into the published Ilv2p crystal structure outlined the chlorimuron-ethyl binding cavity, and it was possible to dock the triazolopyrimidine-sulfonamide compound into this pocket in silico. However, fungal growth inhibition could be bypassed through supplementation with exogenous branched-chain amino acids or by the addition of serum to the medium in all of the fungal organisms tested except for Aspergillus fumigatus. Thus, these data support the identification of the triazolopyrimidine-sulfonamide compounds as inhibitors of acetolactate synthase but suggest that targeting may be compromised due to the possibility of nutrient bypass in vivo.


Subject(s)
Acetolactate Synthase/antagonists & inhibitors , Antifungal Agents/pharmacology , Pyrimidines/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Sulfonamides/pharmacology , Sulfonylurea Compounds/pharmacology , Acetolactate Synthase/chemistry , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Amino Acids, Branched-Chain/metabolism , Amino Acids, Branched-Chain/pharmacology , Antifungal Agents/chemistry , Catalytic Domain/drug effects , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutation , Protein Binding , Pyrimidines/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Serum/chemistry , Serum/metabolism , Sulfonamides/chemistry , Sulfonylurea Compounds/chemistry
18.
Nat Chem Biol ; 7(9): 639-47, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822274

ABSTRACT

Cephalostatin 1, OSW-1, ritterazine B and schweinfurthin A are natural products that potently, and in some cases selectively, inhibit the growth of cultured human cancer cell lines. The cellular targets of these small molecules have yet to be identified. We have discovered that these molecules target oxysterol binding protein (OSBP) and its closest paralog, OSBP-related protein 4L (ORP4L)--proteins not known to be involved in cancer cell survival. OSBP and the ORPs constitute an evolutionarily conserved protein superfamily, members of which have been implicated in signal transduction, lipid transport and lipid metabolism. The functions of OSBP and the ORPs, however, remain largely enigmatic. Based on our findings, we have named the aforementioned natural products ORPphilins. Here we used ORPphilins to reveal new cellular activities of OSBP. The ORPphilins are powerful probes of OSBP and ORP4L that will be useful in uncovering their cellular functions and their roles in human diseases.


Subject(s)
Biological Products/pharmacology , Cholestenones/pharmacology , Neoplasms/metabolism , Phenazines/pharmacology , Receptors, Steroid/metabolism , Saponins/pharmacology , Spiro Compounds/pharmacology , Steroids/pharmacology , Biological Products/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cholestenones/antagonists & inhibitors , Humans , Hydroxycholesterols/pharmacology , Lipid Metabolism/drug effects , Phenazines/antagonists & inhibitors , Receptors, Steroid/genetics , Saponins/antagonists & inhibitors , Signal Transduction/drug effects , Sphingomyelins/biosynthesis , Spiro Compounds/antagonists & inhibitors , Steroids/antagonists & inhibitors , Stilbenes/antagonists & inhibitors , Stilbenes/pharmacology
20.
Nat Rev Drug Discov ; 22(7): 585-603, 2023 07.
Article in English | MEDLINE | ID: mdl-37173515

ABSTRACT

During the coronavirus disease 2019 (COVID-19) pandemic, a wave of rapid and collaborative drug discovery efforts took place in academia and industry, culminating in several therapeutics being discovered, approved and deployed in a 2-year time frame. This article summarizes the collective experience of several pharmaceutical companies and academic collaborations that were active in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antiviral discovery. We outline our opinions and experiences on key stages in the small-molecule drug discovery process: target selection, medicinal chemistry, antiviral assays, animal efficacy and attempts to pre-empt resistance. We propose strategies that could accelerate future efforts and argue that a key bottleneck is the lack of quality chemical probes around understudied viral targets, which would serve as a starting point for drug discovery. Considering the small size of the viral proteome, comprehensively building an arsenal of probes for proteins in viruses of pandemic concern is a worthwhile and tractable challenge for the community.


Subject(s)
COVID-19 , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , SARS-CoV-2 , Drug Discovery , Pandemics
SELECTION OF CITATIONS
SEARCH DETAIL