ACS Chemical BiologyPub Date : 2025-05-16Epub Date: 2025-04-14DOI: 10.1021/acschembio.5c00097
Sebastian Mandel, Thomas Hanke, Sebastian Mathea, Deep Chatterjee, Hayuningbudi Saraswati, Benedict-Tilman Berger, Martin Peter Schwalm, Satoshi Yamamoto, Michiko Tawada, Terufumi Takagi, Mahmood Ahmed, Sandra Röhm, Ana Corrionero, Patricia Alfonso, Maria Baena, Lewis Elson, Amelie Menge, Andreas Krämer, Raquel Pereira, Susanne Müller, Daniela S Krause, Stefan Knapp
{"title":"Repurposing of the RIPK1-Selective Benzo[1,4]oxazepin-4-one Scaffold for the Development of a Type III LIMK1/2 Inhibitor.","authors":"Sebastian Mandel, Thomas Hanke, Sebastian Mathea, Deep Chatterjee, Hayuningbudi Saraswati, Benedict-Tilman Berger, Martin Peter Schwalm, Satoshi Yamamoto, Michiko Tawada, Terufumi Takagi, Mahmood Ahmed, Sandra Röhm, Ana Corrionero, Patricia Alfonso, Maria Baena, Lewis Elson, Amelie Menge, Andreas Krämer, Raquel Pereira, Susanne Müller, Daniela S Krause, Stefan Knapp","doi":"10.1021/acschembio.5c00097","DOIUrl":"10.1021/acschembio.5c00097","url":null,"abstract":"<p><p>Benzoxazepinones have been extensively studied as exclusively selective RIP kinase 1 inhibitors. This scaffold binds to an allosteric pocket created by an αC-out/DFG-out conformation. This inactive conformation results in a large expansion of the kinase back pocket, a conformation that has also been reported for LIM kinases. Scaffold hopping is common in the design of orthosteric kinase inhibitors but has not been explored in the design of allosteric inhibitors, mainly due to the typically exclusive selectivity of type III inhibitors. Here, we hypothesized that the shared structural properties of LIMKs and RIPKs could lead to novel type III LIMK inhibitors using the benzoxazepinone scaffold. We report the discovery of a novel LIMK1/2 inhibitor that relies on this scaffold-based approach. The discovered compound <b>10</b> showed low nanomolar potency on LIMK1/2 and exceptional selectivity, as confirmed by a comprehensive selectivity panel with residual RIPK activity as the only off-target. The study provides one of the few examples for scaffold hopping for allosteric inhibitors, which are usually associated with exclusive target selectivity.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"1087-1098"},"PeriodicalIF":3.5,"publicationDate":"2025-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12090182/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Chemically Induced Nuclear Pore Complex Protein Degradation via TRIM21.","authors":"Xiaomei Li, Qingyang Wang, Anping Guo, Yaping Qiu, Qiuxia Chen, You Li, Lanjun Zhang, Yaxin Guo, Xiaoyun Meng, Shiqian Li, Guizhi Liu, Liyun Zhang, Jian Liu, Xianyang Li, Longying Cai, Xuemin Cheng, Chuan Liu, Xiaotao Wang, Andrew Wood, James Murray, Guansai Liu, Jin Li, Xiaodong Huang, Dengfeng Dou","doi":"10.1021/acschembio.4c00833","DOIUrl":"10.1021/acschembio.4c00833","url":null,"abstract":"<p><p>Despite the exciting progress of bifunctional degrader molecules, also known as proteolysis-targeting chimeras (PROTACs), the rapidly expanding field is still significantly hampered by the lack of available E3 ligase ligands. Our research bridges this gap by uncovering a series of small-molecule ligands to the E3 ligase TRIM21 through DNA-Encoded Library (DEL) technology. We confirmed their interaction with TRIM21 using crystallography and demonstrated their antiproliferative effects across various cancer cell types. Furthermore, proteomic studies identified that the mRNA Export Factor GLE1 and the Nuclear Pore Complex Protein NUP155 were significantly downregulated on TRIM21 ligand treatment. This degradation required TRIM21 and was ubiquitin-proteasome-dependent. More specifically, NUP155 was the primary target for the TRIM21 ligands, while GLE1 was considered a passenger target on initial degradation of NUP155. Using immunofluorescence techniques, we further demonstrated that the degradation of GLE1 and NUP155 proteins impaired the integrity of the nuclear envelope, leading to cell death. Highlighted by this research, a novel mode of action has been discovered for the TRIM21 E3 ligase ligand, acting as a monovalent degrader that triggers de novo interaction with functional complex proteins and induces their degradation.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"1020-1028"},"PeriodicalIF":3.5,"publicationDate":"2025-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jillian M Hagel, Limei Chang, Jing Li, Xue Chen, Lisa Yu, Jonathan A Gallant, Peter J Facchini
{"title":"Bioproduction of a Large-Scale Library of Tryptamine Derivatives for Neuropsychiatric Drug Screening.","authors":"Jillian M Hagel, Limei Chang, Jing Li, Xue Chen, Lisa Yu, Jonathan A Gallant, Peter J Facchini","doi":"10.1021/acschembio.4c00857","DOIUrl":"https://doi.org/10.1021/acschembio.4c00857","url":null,"abstract":"<p><p>Drug screening programs targeting novel indolethylamines with pharmacological properties suitable for the treatment of psychiatric and central nervous system disorders benefit from the availability of large compound libraries normally prepared using synthetic chemistry. Bioproduction strategies based on microbial metabolic engineering and fermentation generally fail to achieve the throughput, scale, or versatility of synthetic chemistry owing, in part, to a lack of efficient and promiscuous enzymes. Moreover, synthetic biology rarely extends to the purification of targeted products, which is an essential component of synthetic chemistry and drug screening regimes. A lattice of biosynthetic routes beginning with endogenous tryptophan or exogenous indole derivatives were engineered in <i>Escherichia coli</i> using heterologous genes encoding enzymes sourced from plants, mushrooms, microbes and animals. Twelve tryptophan decarboxylase candidates were screened and highly versatile top-performers from <i>Bacillus atrophaeus</i> and the gut microbiome species <i>Clostridium sporogenes</i> were identified. Seven halogenases, three tryptophan synthase β-subunits, six <i>N</i>-methyltransferases, five regioselective prenyltransferases, a cytochrome P450 oxidoreductase 5-hydroxylase, an <i>N</i>-acetyltransferase, a 4-<i>O</i>-kinase and various accessory proteins were also tested. These enzymes were used in various combinations and permutations to build <i>E. coli</i> strains capable of 344 putative biotransformations, which resulted in the formation of 279 products with only 63 targeted compounds not detected. A set of 17 novel <i>N</i>-acetylated derivatives were selected for upscaled culturing and purification to ≥95% from 0.5 to 1 L of the fermentation broth, which yielded ∼6-80 mg of each molecule. The potential of each compound for bioactivity at 14 different receptors or transporters with established or purported involvement in neuropsychiatric diseases was tested using a single ligand concentration. Nearly all the <i>N</i>-acetylated compounds showed interaction with the melatonin (MT<sub>1</sub>) receptor, and several molecules showed interaction with serotonergic receptors 5-HT<sub>2B</sub>, 5-HT<sub>2C</sub>, and 5-HT<sub>7</sub>. Overall, we show that bio-fermentation is useful in the large-scale screening of molecules with potential in drug development.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144074835","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
C Taylor Dederich, Livia S Lazarus, Abby D Benninghoff, Lisa M Berreau
{"title":"Localized Light-Triggered CO Delivery: Comparing the Amount of CO Delivered and Cellular Toxicity.","authors":"C Taylor Dederich, Livia S Lazarus, Abby D Benninghoff, Lisa M Berreau","doi":"10.1021/acschembio.4c00884","DOIUrl":"https://doi.org/10.1021/acschembio.4c00884","url":null,"abstract":"<p><p>Molecules that enable the controlled delivery of carbon monoxide (CO) in biological environments are of significant current interest to probe the beneficial roles of CO for humans. Assumptions regarding the ability of molecules to reliably deliver CO continue to impact the field, including in work involving non-metal CO delivery motifs. Flavonols are drawing increasing interest as light-triggered CO release motifs due to their ease of synthesis, functionalization, and fluorescence trackability. Importantly, the light-driven CO release properties of flavonols depend on their structure and must be fully evaluated under various conditions to understand the relationship between the amount of CO delivered and the induced biological effects. Herein, we use a family of amine-functionalized π-extended flavonols to demonstrate that structural differences result in differing interactions with biomolecules, cellular uptake, and changes in subcellular localization, which can affect the amount of CO delivered intracellularly. This results in differences in the CO-induced cellular toxicity.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144074838","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Julie S Valastyan, Emilee E Shine, Robert A Mook, Bonnie L Bassler
{"title":"Inhibitors of the PqsR Quorum-Sensing Receptor Reveal Differential Roles for PqsE and RhlI in Control of Phenazine Production.","authors":"Julie S Valastyan, Emilee E Shine, Robert A Mook, Bonnie L Bassler","doi":"10.1021/acschembio.5c00114","DOIUrl":"10.1021/acschembio.5c00114","url":null,"abstract":"<p><p><i>Pseudomonas aeruginosa</i> is a leading cause of hospital-acquired infections and it is resistant to many current antibiotic therapies, making development of new antimicrobial treatments imperative. The cell-to-cell communication process called quorum sensing controls <i>P. aeruginosa</i> pathogenicity. Quorum sensing relies on the production, release, and group-wide detection of extracellular signal molecules called autoinducers. Quorum sensing enables bacteria to synchronize group behaviors. <i>P. aeruginosa</i> possesses multiple quorum-sensing systems that control overlapping regulons, including some required for virulence and biofilm formation. Interventions that target <i>P. aeruginosa</i> quorum-sensing receptors are considered a fruitful avenue to pursue for new therapeutic advances. Here, we developed a <i>P. aeruginosa</i> strain that carries a bioluminescent reporter fused to a target promoter that is controlled by two <i>P. aeruginosa</i> quorum-sensing receptors. The receptors are PqsR, which binds and responds to the autoinducer called PQS (2-heptyl-3-hydroxy-4(1<i>H</i>)-quinolone) and RhlR, which binds and responds to the autoinducer called C4-HSL (C4-homoserine lactone). We used this reporter strain to screen >100,000 compounds with the aim of identifying inhibitors of either or both the PqsR and RhlR quorum-sensing receptors. We report results for 30 PqsR inhibitors from this screen. All of the identified compounds inhibit PqsR with IC<sub>50</sub> values in the nanomolar to low micromolar range and they are readily docked into the autoinducer binding site of the PqsR crystal structure, suggesting they function competitively. The majority of hits identified are not structurally related to previously reported PqsR inhibitors. Recently, RhlR was shown to rely on the accessory protein PqsE for full function. Specifically, RhlR controls different subsets of genes depending on whether or not it is bound to PqsE, however, the consequences of differential regulation on the quorum-sensing output response have not been defined. PqsR regulates <i>pqsE</i>. That feature of the system enabled us to exploit our new set of PqsR inhibitors to show that RhlR requires PqsE to activate the biosynthetic genes for pyocyanin, a key <i>P. aeruginosa</i> virulence factor, while C4-HSL is dispensable. These results highlight the promise of inhibition of PqsR as a possible <i>P. aeruginosa</i> therapeutic to suppress production of factors under RhlR-PqsE control.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950966","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ally Thompson, Nehru Viji Sankaranarayanan, John E Chittum, Virendrasinh Mahida, Sharath S Vishweshwara, Rakesh Raigawali, Saurabh Anand, Raghavendra Kikkeri, Umesh R Desai
{"title":"Identification of an Unnatural Sulfated Monosaccharide as a High-Affinity Ligand for Pan-Variant Targeting of SARS-CoV-2 Spike Glycoprotein.","authors":"Ally Thompson, Nehru Viji Sankaranarayanan, John E Chittum, Virendrasinh Mahida, Sharath S Vishweshwara, Rakesh Raigawali, Saurabh Anand, Raghavendra Kikkeri, Umesh R Desai","doi":"10.1021/acschembio.5c00206","DOIUrl":"https://doi.org/10.1021/acschembio.5c00206","url":null,"abstract":"<p><p>Identifying smaller sulfated glycan fragments that recognize target proteins with high affinity is highly challenging. In this work, we show that microarray screening of 53 small glycan fragments helped identify distinct sulfated monosaccharide to tetrasaccharide fragments that bind to multiple isoforms of SARS-CoV-2 spike glycoprotein (SgP) with high affinity. Our library consisted of natural and unnatural glycan sequences with a wide range of sulfation levels. The unnatural features arose from the presence of phosphate or fluoro groups on the natural sulfated GAG scaffold as well as sulfate modification of idose fragments that were monomer to tetramer long. None of the natural glycans yielded much promise, which probably conveys the importance of the polymeric glycosaminoglycan chain in SgP biology. However, the unnatural idose fragments with sulfation at the 2, 3, 4, and 6 positions displayed high affinities (100-500 nM) for wild-type, Delta, and Omicron variants of SgP. The unnatural sulfated idose monosaccharide is the smallest molecule known to date that can be classified as a high-affinity, pan-variant fragment. This fragment is expected to serve as the lead for the design of pan-variant ligands with sub-nM inhibition potency.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143951377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Protease-Containing Nanobodies for Detecting and Manipulating Intracellular Antigens Using Antiviral Drugs.","authors":"Quan Le, John T Ngo","doi":"10.1021/acschembio.5c00176","DOIUrl":"https://doi.org/10.1021/acschembio.5c00176","url":null,"abstract":"<p><p>Tools to induce the formation of protein-protein interactions (PPIs) via small molecules are essential for investigating and engineering biological systems. Here we introduce a protease-based strategy for controlling the preservation of otherwise self-cleaving nanobodies. By inserting the hepatitis C virus NS3 cis-protease into the nanobody scaffold, we showed that the antigen-binding ability of these chimeric nanobodies can be controlled in a dose-dependent manner using NS3 inhibitors. We demonstrated the generalizability of this approach by designing and validating drug-controllable nanobodies targeting mCherry (LaM4), eGFP (LaG16), and the ALFA peptide tag. Additionally, we showed that an NS3-containing version of a nanobody targeting the β2-adrenergic receptor can control the endogenous G-protein-mediated signaling activity. Overall, we introduce new chemogenetic components for controlling intracellular PPIs using clinically approved antiviral drugs.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiangpeiyun Jin, Atharva S Kulkarni, Andrew C McAvoy, Neha Garg
{"title":"Antimicrobial Agent Trimethoprim Influences Chemical Interactions in Cystic Fibrosis Pathogens via the <i>ham</i> Gene Cluster.","authors":"Jiangpeiyun Jin, Atharva S Kulkarni, Andrew C McAvoy, Neha Garg","doi":"10.1021/acschembio.4c00562","DOIUrl":"https://doi.org/10.1021/acschembio.4c00562","url":null,"abstract":"<p><p>The fungus <i>Aspergillus fumigatus</i> and the bacterium <i>Burkholderia cenocepacia</i> cause fatal respiratory infections in immunocompromised humans and patients with lung disease, such as cystic fibrosis (CF). In dual infections, antagonistic interactions contribute to increased mortality. These interactions are further altered by the presence of antimicrobial and antifungal agents. However, studies performed to date on chemical interactions between clinical <i>B. cenocepacia</i> and <i>A. fumigatus</i> have focused on pathogens in isolation and do not include the most abundant chemical signal, i.e., clinically administered therapeutics, present in the lung. Here, we characterize small molecule-mediated interactions between <i>B. cenocepacia</i> and <i>A. fumigatus</i> and their shift in response to trimethoprim exposure by using metabolomics and mass spectrometry imaging. Using these methods, we report that the production of several small-molecule natural products of both the bacteria and the fungus is affected by cocultivation and exposure to trimethoprim. By systematic analysis of metabolomics data, we hypothesize that the <i>B. cenocepacia</i>-encoded <i>ham</i> gene cluster plays a role in the trimethoprim-mediated alteration of bacterial-fungal interactions. We support our findings by generating a genetically modified strain lacking the <i>ham</i> gene cluster and querying its interaction with <i>A. fumigatus</i>. Using comparative analyses of the extracts of wild-type and knockout strains, we report the inactivation of a bacterially produced antifungal compound, fragin, by <i>A. fumigatus</i>, which was verified by the addition of purified fragin to the <i>A. fumigatus</i> culture. Furthermore, we report that trimethoprim does not inhibit fungal growth, but affects the biochemical pathway for DHN-melanin biosynthesis, an important antifungal drug target, altering the pigmentation of the fungal conidia and is associated with modification of ergosterol to ergosteryl-3β-O-l-valine in coculture. This study demonstrates the impact of therapeutics on shaping microbial and fungal metabolomes, which influence interkingdom interactions and the expression of virulence factors. Our findings enhance the understanding of the complexity of chemical interactions between therapeutic compounds, bacteria, and fungi and may contribute to the development of selective treatments.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Severin Lechner, Shuyao Sha, Jigar Paras Sethiya, Patrycja Szczupak, Rafal Dolot, Santosh Lomada, Amirhossein Sakhteman, Johanna Tushaus, Polina Prokofeva, Michael Krauss, Ferdinand Breu, Katharina Vögerl, Martin Morgenstern, Martin Hrabě de Angelis, Volker Haucke, Thomas Wieland, Carston Wagner, Guillaume Médard, Franz Bracher, Bernhard Kuster
{"title":"Serendipitous and Systematic Chemoproteomic Discovery of MBLAC2, HINT1, and NME1-4 Inhibitors from Histone Deacetylase-Targeting Pharmacophores.","authors":"Severin Lechner, Shuyao Sha, Jigar Paras Sethiya, Patrycja Szczupak, Rafal Dolot, Santosh Lomada, Amirhossein Sakhteman, Johanna Tushaus, Polina Prokofeva, Michael Krauss, Ferdinand Breu, Katharina Vögerl, Martin Morgenstern, Martin Hrabě de Angelis, Volker Haucke, Thomas Wieland, Carston Wagner, Guillaume Médard, Franz Bracher, Bernhard Kuster","doi":"10.1021/acschembio.5c00108","DOIUrl":"https://doi.org/10.1021/acschembio.5c00108","url":null,"abstract":"<p><p>Metalloenzyme inhibitors often incorporate a hydroxamic acid moiety to bind the bivalent metal ion cofactor within the enzyme's active site. Recently, inhibitors of Zn<sup>2+</sup>-dependent histone deacetylases (HDACs), including clinically advanced drugs, have been identified as potent inhibitors of the metalloenzyme MBLAC2. However, selective chemical probes for MBLAC2, which are essential for studying its inhibitory effects, have not yet been reported. To discover highly selective MBLAC2 inhibitors, we conducted chemoproteomic target deconvolution and selectivity profiling of a library of hydroxamic acid-type molecules and other metal-chelating compounds. This screen revealed MBLAC2 as a frequent off-target of supposedly selective HDAC inhibitors, including the HDAC6 inhibitor SW-100. Profiling a focused library of SW-100-related phenylhydroxamic acids led to identifying two compounds, KV-65 and KV-79, which exhibit nanomolar binding affinity for MBLAC2 and over 60-fold selectivity compared to HDACs. Interestingly, some phenylhydroxamic acids were found to bind additional off-targets. We identified KV-30 as the first drug-like inhibitor of the histidine triad nucleotide-binding protein HINT1 and confirmed its mode of inhibition through a cocrystal structure analysis. Furthermore, we report the discovery of the first inhibitors for the undrugged nucleoside diphosphate kinases NME1, NME2, NME3, and NME4. Overall, this study maps the target and off-target landscape of 53 metalloenzyme inhibitors, providing the first selective MBLAC2 inhibitors. Additionally, the discovery of pharmacophores for NME1-4 and HINT1 establishes a foundation for the future design of potent and selective inhibitors for these targets.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-05-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ACS Chemical BiologyPub Date : 2025-05-05DOI: 10.1021/acschembio.5c0015810.1021/acschembio.5c00158
Hengrui Liu, Farhad Forouhar, Russell Saneto and Brent R. Stockwell*,
{"title":"Selective Small-Molecule Activator of Patient-Derived GPX4 Variant","authors":"Hengrui Liu, Farhad Forouhar, Russell Saneto and Brent R. Stockwell*, ","doi":"10.1021/acschembio.5c0015810.1021/acschembio.5c00158","DOIUrl":"https://doi.org/10.1021/acschembio.5c00158https://doi.org/10.1021/acschembio.5c00158","url":null,"abstract":"<p >Glutathione peroxidase 4 (GPX4) is distinguished from other members of the GPX family as being the enzyme capable of reducing phospholipid hydroperoxides within cellular membranes and therefore protecting cells from ferroptosis, a form of iron-driven cell death involving lipid peroxidation. We previously identified a homozygous point mutation in the <i>GPX4</i> gene, resulting in an R152H coding mutation and a substantial loss of GPX4 enzymatic activity, in patients with Sedaghatian-type spondylometaphyseal dysplasia (SSMD), an ultrarare progressive disorder. To explore whether selective binding and correction of the loss of enzyme activity observed with this variant is possible, we screened 2.8 billion compounds in a DNA-encoded chemical library and identified compounds with remarkably selective binding affinities with the R152H variant (GPX4<sup>R152H</sup>) over wild-type (GPX4<sup>WT</sup>). Our structural optimization of these compounds led to the identification of analogues with improved potency for R152H GPX4. The most promising compounds selectively restored the enzyme activity of GPX4<sup>R152H</sup> and specifically increased the viability of fibroblast and lymphoblast cells developed from an SSMD patient with the homozygous R152H variation but not control cells from a healthy parent or HEK293T cells undergoing ferroptosis induced by a wild-type GPX4 inhibitor. This approach represents a low-cost, high-throughput, and generalizable approach to identify targeted small-molecule therapeutics for missense variants, which features the potential to be broadly applied to diseases that bear point mutations on crucial proteins, including cancers.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":"20 5","pages":"1107–1122 1107–1122"},"PeriodicalIF":3.5,"publicationDate":"2025-05-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144067841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}