Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 515
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(10): 1793-1805.e17, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35483372

ABSTRACT

The lack of tools to observe drug-target interactions at cellular resolution in intact tissue has been a major barrier to understanding inĀ vivo drug actions. Here, we develop clearing-assisted tissue click chemistry (CATCH) to optically image covalent drug targets in intact mammalian tissues. CATCH permits specific and robust in situ fluorescence imaging of target-bound drug molecules at subcellular resolution and enables the identification of target cell types. Using well-established inhibitors of endocannabinoid hydrolases and monoamine oxidases, direct or competitive CATCH not only reveals distinct anatomical distributions and predominant cell targets of different drug compounds in the mouse brain but also uncovers unexpected differences in drug engagement across and within brain regions, reflecting rare cell types, as well as dose-dependent target shifts across tissue, cellular, and subcellular compartments that are not accessible by conventional methods. CATCH represents a valuable platform for visualizingĀ in vivoĀ interactions of small molecules in tissue.


Subject(s)
Click Chemistry , Optical Imaging , Animals , Brain , Drug Delivery Systems , Mammals , Mice , Optical Imaging/methods
2.
Cell ; 182(4): 1009-1026.e29, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32730809

ABSTRACT

Electrophilic compounds originating from nature or chemical synthesis have profound effects on immune cells. These compounds are thought to act by cysteine modification to alter the functions of immune-relevant proteins; however, our understanding of electrophile-sensitive cysteines in the human immune proteome remains limited. Here, we present a global map of cysteines in primary human TĀ cells that are susceptible to covalent modification by electrophilic small molecules. More than 3,000 covalently liganded cysteines were found on functionally and structurally diverse proteins, including many that play fundamental roles in immunology. We further show that electrophilic compounds can impair TĀ cell activation by distinct mechanisms involving the direct functional perturbation and/or degradation of proteins. Our findings reveal a rich content of ligandable cysteines in human TĀ cells and point to electrophilic small molecules as a fertile source for chemical probes and ultimately therapeutics that modulate immunological processes and their associated disorders.


Subject(s)
Cysteine/metabolism , Ligands , T-Lymphocytes/metabolism , Acetamides/chemistry , Acetamides/pharmacology , Acrylamides/chemistry , Acrylamides/pharmacology , Cells, Cultured , Humans , Inhibitor of Apoptosis Proteins/metabolism , Lymphocyte Activation/drug effects , Protein-Tyrosine Kinases/metabolism , Proteolysis/drug effects , Proteome/chemistry , Proteome/metabolism , Stereoisomerism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/metabolism
3.
Cell ; 173(4): 815-817, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727668

ABSTRACT

Matching genetically defined cancer states to drugs that specifically target these states is a principal goal of personalized oncology medicine. In this issue, McMillan etĀ al. show how large-scale chemical screening coupled to deep molecular profiling can identify mechanistically diverse druggable vulnerabilities for genetic subtypes of lung cancers.


Subject(s)
Gene Expression Profiling , Lung Neoplasms , Humans , Molecular Targeted Therapy , Precision Medicine
4.
Cell ; 168(3): 527-541.e29, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111073

ABSTRACT

Advances in the synthesis and screening of small-molecule libraries have accelerated the discovery of chemical probes for studying biological processes. Still, only a small fraction of the human proteome has chemical ligands. Here, we describe a platform that marries fragment-based ligand discovery with quantitative chemical proteomics to map thousands of reversible small molecule-protein interactions directly in human cells, many of which can be site-specifically determined. We show that fragment hitsĀ can be advanced to furnish selective ligands that affect the activity of proteins heretofore lackingĀ chemical probes. We further combine fragment-based chemical proteomics with phenotypic screening to identify small molecules that promote adipocyte differentiation by engaging the poorly characterized membrane protein PGRMC2. Fragment-based screening in human cells thus provides an extensive proteome-wide map of protein ligandability and facilitates the coordinated discovery of bioactive small molecules and their molecular targets.


Subject(s)
Drug Discovery/methods , Proteomics/methods , Adipocytes/cytology , Cell Differentiation , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Hydrolases/chemistry , Ligands , Membrane Proteins/antagonists & inhibitors , Oxidoreductases/chemistry , Protein Binding , Receptors, Progesterone/antagonists & inhibitors , Small Molecule Libraries
5.
Cell ; 168(3): 517-526.e18, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111075

ABSTRACT

The gut microbiota modulate host biology in numerous ways, but little is known about the molecular mediators of these interactions. Previously, we found a widely distributed family of nonribosomal peptide synthetase gene clusters in gut bacteria. Here, by expressing a subset of these clusters in Escherichia coli or Bacillus subtilis, we show that they encode pyrazinones and dihydropyrazinones. At least one of the 47 clusters is present in 88% of the National Institutes of Health Human Microbiome Project (NIH HMP) stool samples, and they are transcribed under conditions of host colonization. We present evidence that the active form of these molecules is the initially released peptide aldehyde, which bears potent protease inhibitory activity and selectively targets a subset of cathepsins in human cell proteomes. Our findings show that an approach combining bioinformatics, synthetic biology, and heterologous gene cluster expression can rapidly expand our knowledge of the metabolic potential of the microbiota while avoiding the challenges of cultivating fastidious commensals.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Microbiota , Peptide Synthases/metabolism , Pyrazines/metabolism , Animals , Bacillus subtilis/genetics , Bacteria/classification , Bacteria/genetics , Escherichia coli/genetics , Feces/microbiology , Humans , Peptide Synthases/genetics , Phylogeny
6.
Cell ; 171(3): 696-709.e23, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28965760

ABSTRACT

The transcription factor NRF2 is a master regulator of the cellular antioxidant response, and it is often genetically activated in non-small-cell lung cancers (NSCLCs) by, for instance, mutations in the negative regulator KEAP1. While direct pharmacological inhibition of NRF2 has proven challenging, its aberrant activation rewires biochemical networks in cancer cells that may create special vulnerabilities. Here, we use chemical proteomics to map druggable proteins that are selectively expressed in KEAP1-mutant NSCLC cells. Principal among these is NR0B1, an atypical orphan nuclear receptor that we show engages in a multimeric protein complex to regulate the transcriptional output of KEAP1-mutant NSCLC cells. We further identify small molecules that covalently target a conserved cysteine within the NR0B1 protein interaction domain, and we demonstrate that these compounds disrupt NR0B1 complexes and impair the anchorage-independent growth of KEAP1-mutant cancer cells. Our findings designate NR0B1 as a druggable transcriptional regulator that supports NRF2-dependent lung cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Proteome/analysis , Transcriptome , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cysteine/metabolism , DAX-1 Orphan Nuclear Receptor/metabolism , Gene Regulatory Networks , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Ligands , Lung Neoplasms/metabolism
7.
Cell ; 166(1): 126-39, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27368101

ABSTRACT

The HIF transcription factor promotes adaptation toĀ hypoxia and stimulates the growth of certain cancers, including triple-negative breast cancer (TNBC). The HIFα subunit is usually prolyl-hydroxylated by EglN family members under normoxic conditions, causing its rapid degradation. We confirmed that TNBC cells secrete glutamate, which we found is both necessary and sufficient for the paracrine induction of HIF1α in such cells under normoxic conditions. Glutamate inhibits the xCT glutamate-cystine antiporter, leading to intracellular cysteine depletion. EglN1, the main HIFα prolyl-hydroxylase, undergoes oxidative self-inactivation in the absence of cysteine both in biochemical assays and in cells, resulting in HIF1α accumulation. Therefore, EglN1 senses both oxygen and cysteine.


Subject(s)
Breast Neoplasms/metabolism , Cysteine/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Paracrine Communication , Triple Negative Breast Neoplasms/metabolism , Amino Acid Transport System y+/metabolism , Animals , Glutamic Acid/metabolism , Humans , MCF-7 Cells , Mice
8.
Mol Cell ; 83(10): 1725-1742.e12, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37084731

ABSTRACT

Most human proteins lack chemical probes, and several large-scale and generalizable small-molecule binding assays have been introduced to address this problem. How compounds discovered in such "binding-first" assays affect protein function, nonetheless, often remains unclear. Here, we describe a "function-first" proteomic strategy that uses size exclusion chromatography (SEC) to assess the global impact of electrophilic compounds on protein complexes in human cells. Integrating the SEC data with cysteine-directed activity-based protein profiling identifies changes in protein-protein interactions that are caused by site-specific liganding events, including the stereoselective engagement of cysteines in PSME1 and SF3B1 that disrupt the PA28 proteasome regulatory complex and stabilize a dynamic state of the spliceosome, respectively. Our findings thus show how multidimensional proteomic analysis of focused libraries of electrophilic compounds can expedite the discovery of chemical probes with site-specific functional effects on protein complexes in human cells.


Subject(s)
Proteomics , Transcription Factors , Humans , Proteomics/methods , Cysteine/metabolism , Ligands
9.
Cell ; 161(7): 1668-80, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091042

ABSTRACT

Lipids play central roles in physiology and disease, where their structural, metabolic, and signaling functions often arise from interactions with proteins. Here, we describe a set of lipid-based chemical proteomic probes and their global interaction map in mammalian cells. These interactions involve hundreds of proteins from diverse functional classes and frequently occur at sites of drug action. We determine the target profiles for several drugs across the lipid-interaction proteome, revealing that its ligandable content extends far beyond traditionally defined categories of druggable proteins. In further support of this finding, we describe a selective ligand for the lipid-binding protein nucleobindin-1 (NUCB1) and show that this compound perturbs the hydrolytic and oxidative metabolism of endocannabinoids in cells. The described chemical proteomic platform thus provides an integrated path to both discover and pharmacologically characterize a wide range of proteins that participate in lipid pathways in cells.


Subject(s)
Lipid Metabolism , Proteins/analysis , Proteins/metabolism , Animals , Calcium-Binding Proteins/analysis , Cell Line, Tumor , DNA-Binding Proteins/analysis , Drug Evaluation, Preclinical , Eicosanoids/metabolism , Endocannabinoids/metabolism , HEK293 Cells , Humans , Lipid Metabolism/drug effects , Mice , Nerve Tissue Proteins/analysis , Nucleobindins , Proteome/analysis , Proteome/metabolism , Small Molecule Libraries/pharmacology
10.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046436

ABSTRACT

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Subject(s)
Drug Discovery , Small Molecule Libraries , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , National Institutes of Health (U.S.) , United States
11.
Annu Rev Biochem ; 83: 341-77, 2014.
Article in English | MEDLINE | ID: mdl-24905785

ABSTRACT

Eukaryotic and prokaryotic organisms possess huge numbers of uncharacterized enzymes. Selective inhibitors offer powerful probes for assigning functions to enzymes in native biological systems. Here, we discuss how the chemical proteomic platform activity-based protein profiling (ABPP) can be implemented to discover selective and in vivo-active inhibitors for enzymes. We further describe how these inhibitors have been used to delineate the biochemical and cellular functions of enzymes, leading to the discovery of metabolic and signaling pathways that make important contributions to human physiology and disease. These studies demonstrate the value of selective chemical probes as drivers of biological inquiry.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Design , Enzyme Inhibitors/chemistry , Proteomics/methods , Animals , Binding, Competitive , Cell Line, Tumor , Drug Discovery , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Humans , Lactones/chemistry , Phenotype , Photochemistry/methods , Proteome
12.
Annu Rev Pharmacol Toxicol ; 64: 507-526, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-37722721

ABSTRACT

Recent advances in chemical, molecular, and genetic approaches have provided us with an unprecedented capacity to identify drug-target interactions across the whole proteome and genome. Meanwhile, rapid developments of single-cell and spatial omics technologies are revolutionizing our understanding of the molecular architecture of biological systems. However, a significant gap remains in how we align our understanding of drug actions, traditionally based on molecular affinities, with the in vivo cellular and spatial tissue heterogeneity revealed by these newer techniques. Here, we review state-of-the-art methods for profiling drug-target interactions and emerging multiomics tools to delineate the tissue heterogeneity at single-cell resolution. Highlighting the recent technical advances enabling high-resolution, multiplexable in situ small-molecule drug imaging (clearing-assisted tissue click chemistry, or CATCH), we foresee the integration of single-cell and spatial omics platforms, data, and concepts into the future framework of defining and understanding in vivo drug-target interactions and mechanisms of actions.


Subject(s)
Drug Delivery Systems , Proteome , Humans , Technology
13.
Cell ; 150(2): 426-40, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22817901

ABSTRACT

Caspase proteases are principal mediators of apoptosis, where they cleave hundreds of proteins. Phosphorylation also plays an important role in apoptosis, although the extent to which proteolytic and phosphorylation pathways crosstalk during programmed cell death remains poorly understood. Using a quantitative proteomic platform that integrates phosphorylation sites into the topographical maps of proteins, we identify a cohort of over 500 apoptosis-specific phosphorylation events and show that they are enriched on cleaved proteins and clustered around sites of caspase proteolysis. We find that caspase cleavage can expose new sites for phosphorylation, and, conversely, that phosphorylation at the +3 position of cleavage sites can directly promote substrate proteolysis by caspase-8. This study provides a global portrait of the apoptotic phosphoproteome, revealing heretofore unrecognized forms of functional crosstalk between phosphorylation and caspase proteolytic pathways that lead to enhanced rates of protein cleavage and the unveiling of new sites for phosphorylation.


Subject(s)
Apoptosis , Proteome/analysis , Proteome/metabolism , Caspases/metabolism , Humans , Models, Molecular , Phosphorylation , Proteolysis , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 121(41): e2415934121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39356664

ABSTRACT

The propeller-shaped blades of the PIEZO1 and PIEZO2 ion channels partition into the plasma membrane and respond to indentation or stretching of the lipid bilayer, thus converting mechanical forces into signals that can be interpreted by cells, in the form of calcium flux and changes in membrane potential. While PIEZO channels participate in diverse physiological processes, from sensing the shear stress of blood flow in the vasculature to detecting touch through mechanoreceptors in the skin, the molecular details that enable these mechanosensors to tune their responses over a vast dynamic range of forces remain largely uncharacterized. To survey the molecular landscape surrounding PIEZO channels at the cell surface, we employed a mass spectrometry-based proteomic approach to capture and identify extracellularly exposed proteins in the vicinity of PIEZO1. This PIEZO1-proximal interactome was enriched in surface proteins localized to cell junctions and signaling hubs within the plasma membrane. Functional screening of these interaction candidates by calcium imaging and electrophysiology in an overexpression system identified the adhesion molecule CADM1/SynCAM that slows the inactivation kinetics of PIEZO1 with little effect on PIEZO2. Conversely, we found that CADM1 knockdown accelerates inactivation of endogenous PIEZO1 in Neuro-2a cells. Systematic deletion of CADM1 domains indicates that the transmembrane region is critical for the observed effects on PIEZO1, suggesting that modulation of inactivation is mediated by interactions in or near the lipid bilayer.


Subject(s)
Ion Channels , Ion Channels/metabolism , Ion Channels/genetics , Humans , Cell Adhesion Molecule-1/metabolism , Cell Adhesion Molecule-1/genetics , Cell Membrane/metabolism , HEK293 Cells , Proteomics/methods , Mechanotransduction, Cellular , Animals
15.
Nat Chem Biol ; 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294320

ABSTRACT

More than half of the ~20,000 protein-encoding human genes have paralogs. Chemical proteomics has uncovered many electrophile-sensitive cysteines that are exclusive to subsets of paralogous proteins. Here we explore whether such covalent compound-cysteine interactions can be used to discover ligandable pockets in paralogs lacking the cysteine. Leveraging the covalent ligandability of C109 in the cyclin CCNE2, we substituted the corresponding residue in paralog CCNE1 to cysteine (N112C) and found through activity-based protein profiling that this mutant reacts stereoselectively and site-specifically with tryptoline acrylamides. We then converted the tryptoline acrylamide-CCNE1-N112C interaction into in vitro NanoBRET (bioluminescence resonance energy transfer) and in cellulo activity-based protein profiling assays capable of identifying compounds that reversibly inhibit both the N112C mutant and wild-type CCNE1:CDK2 (cyclin-dependent kinase 2) complexes. X-ray crystallography revealed a cryptic allosteric pocket at the CCNE1:CDK2 interface adjacent to N112 that binds the reversible inhibitors. Our findings, thus, show how electrophile-cysteine interactions mapped by chemical proteomics can extend the understanding of protein ligandability beyond covalent chemistry.

16.
Nat Methods ; 19(3): 341-352, 2022 03.
Article in English | MEDLINE | ID: mdl-35228727

ABSTRACT

Proteomics has revealed that the ~20,000 human genes engender a far greater number of proteins, or proteoforms, that are diversified in large part by post-translational modifications (PTMs). How such PTMs affect protein structure and function is an active area of research but remains technically challenging to assess on a proteome-wide scale. Here, we describe a chemical proteomic method to quantitatively relate serine/threonine phosphorylation to changes in the reactivity of cysteine residues, a parameter that can affect the potential for cysteines to be post-translationally modified or engaged by covalent drugs. Leveraging the extensive high-stoichiometry phosphorylation occurring in mitotic cells, we discover numerous cysteines that exhibit phosphorylation-dependent changes in reactivity on diverse proteins enriched in cell cycle regulatory pathways. The discovery of bidirectional changes in cysteine reactivity often occurring in proximity to serine/threonine phosphorylation events points to the broad impact of phosphorylation on the chemical reactivity of proteins and the future potential to create small-molecule probes that differentially target proteoforms with PTMs.


Subject(s)
Cysteine , Proteomics , Cysteine/chemistry , Humans , Phosphorylation , Protein Processing, Post-Translational , Proteome/metabolism , Proteomics/methods , Serine , Threonine/metabolism
17.
Nat Chem Biol ; 19(3): 378-388, 2023 03.
Article in English | MEDLINE | ID: mdl-36782012

ABSTRACT

Ferroptosis is an iron-dependent form of cell death driven by oxidation of polyunsaturated fatty acid (PUFA) phospholipids. Large-scale genetic screens have uncovered a specialized role for PUFA ether phospholipids (ePLs) in promoting ferroptosis. Understanding of the enzymes involved in PUFA-ePL production, however, remains incomplete. Here we show, using a combination of pathway mining of genetic dependency maps, AlphaFold-guided structure predictions and targeted lipidomics, that the uncharacterized transmembrane protein TMEM164-the genetic ablation of which has been shown to protect cells from ferroptosis-is a cysteine active center enzyme that selectively transfers C20:4 acyl chains from phosphatidylcholine to lyso-ePLs to produce PUFA ePLs. Genetic deletion of TMEM164 across a set of ferroptosis-sensitive cancer cell lines caused selective reductions in C20:4 ePLs with minimal effects on C20:4 diacyl PLs, and this lipid profile produced a variable range of protection from ferroptosis, supportive of an important but contextualized role for C20:4 ePLs in this form of cell death.


Subject(s)
Acyltransferases , Phospholipid Ethers , Acyltransferases/metabolism , Phospholipid Ethers/pharmacology , Phospholipids/chemistry , Phosphatidylcholines , Oxidation-Reduction
18.
Nat Chem Biol ; 19(7): 825-836, 2023 07.
Article in English | MEDLINE | ID: mdl-36864190

ABSTRACT

Much of the human proteome is involved in mRNA homeostasis, but most RNA-binding proteins lack chemical probes. Here we identify electrophilic small molecules that rapidly and stereoselectively decrease the expression of transcripts encoding the androgen receptor and its splice variants in prostate cancer cells. We show by chemical proteomics that the compounds engage C145 of the RNA-binding protein NONO. Broader profiling revealed that covalent NONO ligands suppress an array of cancer-relevant genes and impair cancer cell proliferation. Surprisingly, these effects were not observed in cells genetically disrupted for NONO, which were instead resistant to NONO ligands. Reintroduction of wild-type NONO, but not a C145S mutant, restored ligand sensitivity in NONO-disrupted cells. The ligands promoted NONO accumulation in nuclear foci and stabilized NONO-RNA interactions, supporting a trapping mechanism that may prevent compensatory action of paralog proteins PSPC1 and SFPQ. These findings show that NONO can be co-opted by covalent small molecules to suppress protumorigenic transcriptional networks.


Subject(s)
DNA-Binding Proteins , Transcriptome , Male , Humans , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA
19.
Nat Chem Biol ; 19(11): 1320-1330, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37783940

ABSTRACT

Covalent chemistry represents an attractive strategy for expanding the ligandability of the proteome, and chemical proteomics has revealed numerous electrophile-reactive cysteines on diverse human proteins. Determining which of these covalent binding events affect protein function, however, remains challenging. Here we describe a base-editing strategy to infer the functionality of cysteines by quantifying the impact of their missense mutation on cancer cell proliferation. The resulting atlas, which covers more than 13,800 cysteines on more than 1,750 cancer dependency proteins, confirms the essentiality of cysteines targeted by covalent drugs and, when integrated with chemical proteomic data, identifies essential, ligandable cysteines in more than 160 cancer dependency proteins. We further show that a stereoselective and site-specific ligand targeting an essential cysteine in TOE1 inhibits the nuclease activity of this protein through an apparent allosteric mechanism. Our findings thus describe a versatile method and valuable resource to prioritize the pursuit of small-molecule probes with high function-perturbing potential.


Subject(s)
Cysteine , Neoplasms , Humans , Cysteine/chemistry , Proteomics , Gene Editing , Proteome/chemistry , Neoplasms/genetics , Nuclear Proteins
20.
Nat Chem Biol ; 19(7): 815-824, 2023 07.
Article in English | MEDLINE | ID: mdl-36823351

ABSTRACT

Creatine kinases (CKs) provide local ATP production in periods of elevated energetic demand, such as during rapid anabolism and growth. Thus, creatine energetics has emerged as a major metabolic liability in many rapidly proliferating cancers. Whether CKs can be targeted therapeutically is unknown because no potent or selective CK inhibitors have been developed. Here we leverage an active site cysteine present in all CK isoforms to develop a selective covalent inhibitor of creatine phosphagen energetics, CKi. Using deep chemoproteomics, we discover that CKi selectively engages the active site cysteine of CKs in cells. A co-crystal structure of CKi with creatine kinase B indicates active site inhibition that prevents bidirectional phosphotransfer. In cells, CKi and its analogs rapidly and selectively deplete creatine phosphate, and drive toxicity selectively in CK-dependent acute myeloid leukemia. Finally, we use CKi to uncover an essential role for CKs in the regulation of proinflammatory cytokine production in macrophages.


Subject(s)
Creatine Kinase , Creatine , Creatine Kinase/chemistry , Creatine Kinase/metabolism , Creatine/pharmacology , Cysteine , Phosphotransferases , Protein Isoforms
SELECTION OF CITATIONS
SEARCH DETAIL