Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nat Chem Biol ; 19(11): 1309-1319, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37248412

ABSTRACT

With an eye toward expanding chemistries used for covalent ligand discovery, we elaborated an umpolung strategy that exploits the 'polarity reversal' of sulfur when cysteine is oxidized to sulfenic acid, a widespread post-translational modification, for selective bioconjugation with C-nucleophiles. Here we present a global map of a human sulfenome that is susceptible to covalent modification by members of a nucleophilic fragment library. More than 500 liganded sulfenic acids were identified on proteins across diverse functional classes, and, of these, more than 80% were not targeted by electrophilic fragment analogs. We further show that members of our nucleophilic fragment library can impair functional protein-protein interactions involved in nuclear oncoprotein transport and DNA damage repair. Our findings reveal a vast expanse of ligandable sulfenic acids in the human proteome and highlight the utility of nucleophilic small molecules in the fragment-based covalent ligand discovery pipeline, presaging further opportunities using non-traditional chemistries for targeting proteins.


Subject(s)
Cysteine , Sulfenic Acids , Humans , Cysteine/metabolism , Ligands , Proteome/metabolism , Protein Processing, Post-Translational
2.
Nat Chem Biol ; 18(8): 904-912, 2022 08.
Article in English | MEDLINE | ID: mdl-35864333

ABSTRACT

Chemoproteomics has emerged as a key technology to expand the functional space in complex proteomes for probing fundamental biology and for discovering new small-molecule-based therapies. Here we report a modification-centric computational tool termed pChem to provide a streamlined pipeline for unbiased performance assessment of chemoproteomic probes. The pipeline starts with an experimental setting for isotopically coding probe-derived modifications that can be automatically recognized by pChem, with masses accurately calculated and sites precisely localized. pChem exports on-demand reports by scoring the profiling efficiency, modification homogeneity and proteome-wide residue selectivity of a tested probe. The performance and robustness of pChem were benchmarked by applying it to eighteen bioorthogonal probes. These analyses reveal that the formation of unexpected probe-derived modifications can be driven by endogenous reactive metabolites (for example, bioactive aldehydes and glutathione). pChem is a powerful and user-friendly tool that aims to facilitate the development of probes for the ever-growing field of chemoproteomics.


Subject(s)
Aldehydes , Proteome , Aldehydes/chemistry , Proteome/metabolism
3.
Nat Commun ; 12(1): 1415, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658510

ABSTRACT

Post-translational changes in the redox state of cysteine residues can rapidly and reversibly alter protein functions, thereby modulating biological processes. The nematode C. elegans is an ideal model organism for studying cysteine-mediated redox signaling at a network level. Here we present a comprehensive, quantitative, and site-specific profile of the intrinsic reactivity of the cysteinome in wild-type C. elegans. We also describe a global characterization of the C. elegans redoxome in which we measured changes in three major cysteine redox forms after H2O2 treatment. Our data revealed redox-sensitive events in translation, growth signaling, and stress response pathways, and identified redox-regulated cysteines that are important for signaling through the p38 MAP kinase (MAPK) pathway. Our in-depth proteomic dataset provides a molecular basis for understanding redox signaling in vivo, and will serve as a valuable and rich resource for the field of redox biology.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cysteine/metabolism , Animals , Antioxidants/metabolism , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Oxidation-Reduction , Proteomics/methods , Signal Transduction , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Nature ; 589(7843): 620-626, 2021 01.
Article in English | MEDLINE | ID: mdl-33408414

ABSTRACT

Adhesion G-protein-coupled receptors (GPCRs) are a major family of GPCRs, but limited knowledge of their ligand regulation or structure is available1-3. Here we report that glucocorticoid stress hormones activate adhesion G-protein-coupled receptor G3 (ADGRG3; also known as GPR97)4-6, a prototypical adhesion GPCR. The cryo-electron microscopy structures of GPR97-Go complexes bound to the anti-inflammatory drug beclomethasone or the steroid hormone cortisol revealed that glucocorticoids bind to a pocket within the transmembrane domain. The steroidal core of glucocorticoids is packed against the 'toggle switch' residue W6.53, which senses the binding of a ligand and induces activation of the receptor. Active GPR97 uses a quaternary core and HLY motif to fasten the seven-transmembrane bundle and to mediate G protein coupling. The cytoplasmic side of GPR97 has an open cavity, where all three intracellular loops interact with the Go protein, contributing to the high basal activity of GRP97. Palmitoylation at the cytosolic tail of the Go protein was found to be essential for efficient engagement with GPR97 but is not observed in other solved GPCR complex structures. Our work provides a structural basis for ligand binding to the seven-transmembrane domain of an adhesion GPCR and subsequent G protein coupling.


Subject(s)
Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Glucocorticoids/chemistry , Glucocorticoids/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/ultrastructure , Binding Sites , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , Humans , Ligands , Lipoylation , Models, Molecular , Protein Binding , Receptors, G-Protein-Coupled/metabolism
5.
Bioorg Chem ; 107: 104536, 2021 02.
Article in English | MEDLINE | ID: mdl-33342565

ABSTRACT

Caffeic acid phenethyl ester (CAPE), a bioactive component extracted from propolis of honeybee hives, can inhibit hepatocellular carcinoma (HCC). In order to explore more stable CAPE derivatives, 25 compounds were designed, synthesized, and pharmacologically assessed in vitro and in vivo as anti-tumor agents in HCC. Compounds 8d, 8f, 8l, 8j, and 8k showed favorable antiproliferative activity than other compounds including CAPE in the HCC cell lines. Based on the result of QTRP (Quantitative Thiol Reactivity Profiling), epidermal growth factor receptor (EGFR) and C-terminal Src kinase (CSK) were supposed to the targets of 8f, which was confirmed by binding mode analysis. Furthermore, compounds 8f, 8l, 8j, 8k, 8g, and 8h showed potent inhibitory effects against both CSK and EGFR than other derivatives in an ADP-Glo™ kinase assay. The representative compound, 8f, potently inhibited various tumor growth in murine model including murine hepatocellular carcinoma H22, meanwhile downregulating the EGFR/AKT pathway and enhancing T cell proliferation through inhibition of CSK. Metabolic stability in vitro suggested 8f and 8k were more stable in mouse plasma than CAPE and susceptible to metabolism in liver microsomes. The overall excellent profile of compound 8f makes it a potential candidate for further preclinical investigation.


Subject(s)
Caffeic Acids/therapeutic use , Carcinoma, Hepatocellular/drug therapy , ErbB Receptors/antagonists & inhibitors , Liver Neoplasms/drug therapy , Phenylethyl Alcohol/analogs & derivatives , src-Family Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Caffeic Acids/chemistry , Caffeic Acids/pharmacology , Carcinoma, Hepatocellular/pathology , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Female , Liver Neoplasms/pathology , Male , Mice , Molecular Docking Simulation , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Structure-Activity Relationship , src-Family Kinases/metabolism
6.
J Cancer ; 11(21): 6356-6364, 2020.
Article in English | MEDLINE | ID: mdl-33033519

ABSTRACT

Radiotherapy is frequently applied for clinically localized prostate cancer while its efficacy could be significantly hindered by radioresistance. MicroRNAs (miRNAs) are important regulators in mediating cellular responses to ionizing radiation (IR), and strongly associate with radiosensitivity in many cancers. In this study, enhancement of radiosensitivity by miR-29b-3p was demonstrated in prostate cancer cell line LNCaP in vitro. Results showed that miR-29b-3p expression was significantly upregulated in response to IR from both X-rays and carbon ion irradiations. Knockdown of miR-29b-3p resulted in radioresistance while overexpression of miR-29b-3p led to increased radiosensitivity (showing reduced cell viability, suppressed cell proliferation and decreased colony formation). In addition, miR-29b-3p was found to directly target Wnt1-inducible-signaling protein 1 (WISP1). Inhibition of WISP1 facilitated the mitochondrial apoptosis pathway through suppressing Bcl-XL expression while activating caspase-3 and poly (ADP-ribose) polymerase (PARP). The results indicated that miR-29b-3p was a radiosensitizing miRNAs and could enhance radiosensitivity of LNCaP cells by targeting WISP1. These findings suggested a novel treatment to overcome radioresistance in prostate cancer patients, especially those with higher levels of the WISP1 expression.

7.
Front Plant Sci ; 11: 777, 2020.
Article in English | MEDLINE | ID: mdl-32714340

ABSTRACT

In proteins, hydrogen peroxide (H2O2) reacts with redox-sensitive cysteines to form cysteine sulfenic acid, also known as S-sulfenylation. These cysteine oxidation events can steer diverse cellular processes by altering protein interactions, trafficking, conformation, and function. Previously, we had identified S-sulfenylated proteins by using a tagged proteinaceous probe based on the yeast AP-1-like (Yap1) transcription factor that specifically reacts with sulfenic acids and traps them through a mixed disulfide bond. However, the identity of the S-sulfenylated amino acid residues within a protein remained enigmatic. By using the same transgenic YAP1C probe, we present here a technological advancement to identify in situ sulfenylated cysteine sites in Arabidopsis thaliana cells under control condition and oxidative stress. Briefly, the total extract of transgenic YAP1C A. thaliana cells was initially purified on IgG-Sepharose beads, followed by a tryptic digest. Then, the mixed disulfide-linked peptides were further enriched at the peptide level on an anti-YAP1C-derived peptide (C598SEIWDR) antibody. Subsequent mass spectrometry analysis with pLink 2 identified 1,745 YAP1C cross-linked peptides, indicating sulfenylated cysteines in over 1,000 proteins. Approximately 55% of these YAP1C-linked cysteines had previously been reported as redox-sensitive cysteines (S-sulfenylation, S-nitrosylation, and reversibly oxidized cysteines). The presented methodology provides a noninvasive approach to identify sulfenylated cysteines in any species that can be genetically modified.

8.
Nat Protoc ; 15(9): 2891-2919, 2020 09.
Article in English | MEDLINE | ID: mdl-32690958

ABSTRACT

Cysteine is unique among all protein-coding amino acids, owing to its intrinsically high nucleophilicity. The cysteinyl thiol group can be covalently modified by a broad range of redox mechanisms or by various electrophiles derived from exogenous or endogenous sources. Measuring the response of protein cysteines to redox perturbation or electrophiles is critical for understanding the underlying mechanisms involved. Activity-based protein profiling based on thiol-reactive probes has been the method of choice for such analyses. We therefore adapted this approach and developed a new chemoproteomic platform, termed 'QTRP' (quantitative thiol reactivity profiling), that relies on the ability of a commercially available thiol-reactive probe IPM (2-iodo-N-(prop-2-yn-1-yl)acetamide) to covalently label, enrich and quantify the reactive cysteinome in cells and tissues. Here, we provide a detailed and updated workflow of QTRP that includes procedures for (i) labeling of the reactive cysteinome from cell or tissue samples (e.g., control versus treatment) with IPM, (ii) processing the protein samples into tryptic peptides and tagging the probe-modified peptides with isotopically labeled azido-biotin reagents containing a photo-cleavable linker via click chemistry reaction, (iii) capturing biotin-conjugated peptides with streptavidin beads, (iv) identifying and quantifying the photo-released peptides by mass spectrometry (MS)-based shotgun proteomics and (v) interpreting MS data by a streamlined informatic pipeline using a proteomics software, pFind 3, and an automatic post-processing algorithm. We also exemplified here how to use QTRP for mining H2O2-sensitive cysteines and for determining the intrinsic reactivity of cysteines in a complex proteome. We anticipate that this protocol should find broad applications in redox biology, chemical biology and the pharmaceutical industry. The protocol for sample preparation takes 3 d, whereas MS measurements and data analyses require 75 min and <30 min, respectively, per sample.


Subject(s)
Cysteine/metabolism , Proteomics/methods , HEK293 Cells , Humans , Oxidation-Reduction
9.
Cell Chem Biol ; 27(7): 858-865.e8, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32470320

ABSTRACT

The engineered ascorbate peroxidase (APEX) is a powerful tool for the proximity-dependent labeling of proteins and RNAs in live cells. Although widely use in mammalian cells, APEX applications in microorganisms have been hampered by the poor labeling efficiency of its biotin-phenol (BP) substrate. In this study, we sought to address this challenge by designing and screening a panel of alkyne-functionalized substrates. Our best probe, Alk-Ph, substantially improves APEX-labeling efficiency in intact yeast cells, as it is more cell wall-permeant than BP. Through a combination of protein-centric and peptide-centric chemoproteomic experiments, we have identified 165 proteins with a specificity of 94% in the yeast mitochondrial matrix. In addition, we have demonstrated that Alk-Ph is useful for proximity-dependent RNA labeling in yeast, thus expanding the scope of APEX-seq. We envision that this improved APEX-labeling strategy would set the stage for the large-scale mapping of spatial proteome and transcriptome in yeast.


Subject(s)
Ascorbate Peroxidases/metabolism , Molecular Probes/chemistry , Proteome/analysis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Alkynes/chemistry , Ascorbate Peroxidases/chemistry , Chromatography, High Pressure Liquid , Click Chemistry , Mitochondria/metabolism , Molecular Probes/chemical synthesis , Molecular Probes/metabolism , Phenol/chemistry , Proteome/metabolism , Proteomics , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Substrate Specificity , Tandem Mass Spectrometry
10.
Cell Host Microbe ; 27(4): 601-613.e7, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32272078

ABSTRACT

Plants deploy a variety of secondary metabolites to fend off pathogen attack. Although defense compounds are generally considered toxic to microbes, the exact mechanisms are often unknown. Here, we show that the Arabidopsis defense compound sulforaphane (SFN) functions primarily by inhibiting Pseudomonas syringae type III secretion system (TTSS) genes, which are essential for pathogenesis. Plants lacking the aliphatic glucosinolate pathway, which do not accumulate SFN, were unable to attenuate TTSS gene expression and exhibited increased susceptibility to P. syringae strains that cannot detoxify SFN. Chemoproteomics analyses showed that SFN covalently modified the cysteine at position 209 of HrpS, a key transcription factor controlling TTSS gene expression. Site-directed mutagenesis and functional analyses further confirmed that Cys209 was responsible for bacterial sensitivity to SFN in vitro and sensitivity to plant defenses conferred by the aliphatic glucosinolate pathway. Collectively, these results illustrate a previously unknown mechanism by which plants disarm a pathogenic bacterium.


Subject(s)
Arabidopsis/metabolism , Isothiocyanates/pharmacology , Pseudomonas syringae/drug effects , Type III Secretion Systems/drug effects , Bacterial Proteins/drug effects , Cysteine/drug effects , Cysteine/metabolism , Disease Resistance , Gene Expression Regulation, Bacterial , Isothiocyanates/metabolism , Plant Diseases/microbiology , Pseudomonas syringae/metabolism , Secondary Metabolism , Sulfoxides , Transcription Factors/drug effects , Type III Secretion Systems/genetics
11.
Antioxid Redox Signal ; 33(15): 1061-1076, 2020 11 20.
Article in English | MEDLINE | ID: mdl-31411056

ABSTRACT

Aims: Cysteine persulfidation (also called sulfhydration or sulfuration) has emerged as a potential redox mechanism to regulate protein functions and diverse biological processes in hydrogen sulfide (H2S) signaling. Due to its intrinsically unstable nature, working with this modification has proven to be challenging. Although methodological progress has expanded the inventory of persulfidated proteins, there is a continued need to develop methods that can directly and unequivocally identify persulfidated cysteine residues in complex proteomes. Results: A quantitative chemoproteomic method termed as low-pH quantitative thiol reactivity profiling (QTRP) was developed to enable direct site-specific mapping and reactivity profiling of proteomic persulfides and thiols in parallel. The method was first applied to cell lysates treated with NaHS, resulting in the identification of overall 1547 persulfidated sites on 994 proteins. Structural analysis uncovered unique consensus motifs that might define this distinct type of modification. Moreover, the method was extended to profile endogenous protein persulfides in cells expressing H2S-generating enzyme, mouse tissues, and human serum, which led to additional insights into mechanistic, structural, and functional features of persulfidation events, particularly on human serum albumin. Innovation and Conclusion: Low-pH QTRP represents the first method that enables direct and unbiased proteomic mapping of cysteine persulfidation. Our method allows to generate the most comprehensive inventory of persulfidated targets of NaHS so far and to perform the first analysis of in vivo persulfidation events, providing a valuable tool to dissect the biological functions of this important modification.


Subject(s)
Cysteine/metabolism , Protein Processing, Post-Translational , Proteomics/methods , Sulfides/metabolism , Cysteine/chemistry , Humans , Hydrogen Sulfide/metabolism , Hydrogen-Ion Concentration , Oxidation-Reduction , Proteome , Structure-Activity Relationship , Sulfhydryl Compounds/metabolism
12.
Proc Natl Acad Sci U S A ; 116(42): 21256-21261, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31578252

ABSTRACT

Hydrogen peroxide (H2O2) is an important messenger molecule for diverse cellular processes. H2O2 oxidizes proteinaceous cysteinyl thiols to sulfenic acid, also known as S-sulfenylation, thereby affecting the protein conformation and functionality. Although many proteins have been identified as S-sulfenylation targets in plants, site-specific mapping and quantification remain largely unexplored. By means of a peptide-centric chemoproteomics approach, we mapped 1,537 S-sulfenylated sites on more than 1,000 proteins in Arabidopsis thaliana cells. Proteins involved in RNA homeostasis and metabolism were identified as hotspots for S-sulfenylation. Moreover, S-sulfenylation frequently occurred on cysteines located at catalytic sites of enzymes or on cysteines involved in metal binding, hinting at a direct mode of action for redox regulation. Comparison of human and Arabidopsis S-sulfenylation datasets provided 155 conserved S-sulfenylated cysteines, including Cys181 of the Arabidopsis MITOGEN-ACTIVATED PROTEIN KINASE4 (AtMAPK4) that corresponds to Cys161 in the human MAPK1, which has been identified previously as being S-sulfenylated. We show that, by replacing Cys181 of recombinant AtMAPK4 by a redox-insensitive serine residue, the kinase activity decreased, indicating the importance of this noncatalytic cysteine for the kinase mechanism. Altogether, we quantitatively mapped the S-sulfenylated cysteines in Arabidopsis cells under H2O2 stress and thereby generated a comprehensive view on the S-sulfenylation landscape that will facilitate downstream plant redox studies.


Subject(s)
Arabidopsis/metabolism , Proteins/metabolism , Sulfhydryl Compounds/metabolism , Catalytic Domain/physiology , Cysteine/metabolism , Humans , Hydrogen Peroxide/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Oxidation-Reduction , RNA/metabolism , Serine/metabolism , Signal Transduction/physiology , Sulfenic Acids/metabolism
13.
Nat Commun ; 10(1): 1076, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842418

ABSTRACT

Phagocytes, including neutrophils and macrophages, have been suggested to function in a cooperative way in the initial phase of inflammatory responses, but their interaction and integration in the resolution of inflammation and tissue repair remain unclear. Here we show that neutrophils have crucial functions in liver repair by promoting the phenotypic conversion of pro-inflammatory Ly6ChiCX3CR1lo monocytes/macrophages to pro-resolving Ly6CloCX3CR1hi macrophages. Intriguingly, reactive oxygen species (ROS), expressed predominantly by neutrophils, are important mediators that trigger this phenotypic conversion to promote liver repair. Moreover, this conversion is prevented by the depletion of neutrophils via anti-Ly6G antibody, genetic deficiency of granulocyte colony-stimulating factor, or genetic deficiency of NADPH oxidase 2 (Nox2). By contrast, adoptive transfer of WT rather than Nox2-/- neutrophils rescues the impaired phenotypic conversion of macrophages in neutrophil-depleted mice. Our findings thus identify an intricate cooperation between neutrophils and macrophages that orchestrate resolution of inflammation and tissue repair.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Liver Regeneration/immunology , Macrophages/immunology , Neutrophils/immunology , Reactive Oxygen Species/metabolism , Acetaminophen/toxicity , Adoptive Transfer/methods , Animals , Bone Marrow Transplantation , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Humans , Liver/immunology , Liver/pathology , Macrophages/metabolism , Male , Mice , Mice, Knockout , NADPH Oxidase 2/genetics , NADPH Oxidase 2/immunology , Neutrophils/metabolism , Neutrophils/transplantation , Primary Cell Culture , Transplantation Chimera
14.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 35(6): 964-969, 2018 12 25.
Article in Chinese | MEDLINE | ID: mdl-30583324

ABSTRACT

Nucleic acid aptamer is an oligonucleotide sequence screened by the exponential enrichment ligand system evolution technology (SELEX). Previous studies have shown that nucleic acid aptamer has a good application prospect in tumor diagnosis and treatment. Therefore, we reviewed the selection and identification of nucleic acid aptamer of lung cancer cells in recent years, and discussed the effect of aptamer as targeting drugs and targeting vectors on the diagnosis of tumors, which provide a new idea for early diagnosis and treatment of tumor.

15.
Proc Natl Acad Sci U S A ; 115(34): E7978-E7986, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30082411

ABSTRACT

The metabolic and redox state changes during the transition from an arrested oocyte to a totipotent embryo remain uncharacterized. Here, we applied state-of-the-art, integrated methodologies to dissect these changes in Drosophila We demonstrate that early embryos have a more oxidized state than mature oocytes. We identified specific alterations in reactive cysteines at a proteome-wide scale as a result of this metabolic and developmental transition. Consistent with a requirement for redox change, we demonstrate a role for the ovary-specific thioredoxin Deadhead (DHD). dhd-mutant oocytes are prematurely oxidized and exhibit meiotic defects. Epistatic analyses with redox regulators link dhd function to the distinctive redox-state balance set at the oocyte-to-embryo transition. Crucially, global thiol-redox profiling identified proteins whose cysteines became differentially modified in the absence of DHD. We validated these potential DHD substrates by recovering DHD-interaction partners using multiple approaches. One such target, NO66, is a conserved protein that genetically interacts with DHD, revealing parallel functions. As redox changes also have been observed in mammalian oocytes, we hypothesize a link between developmental control of this cell-cycle transition and regulation by metabolic cues. This link likely operates both by general redox state and by changes in the redox state of specific proteins. The redox proteome defined here is a valuable resource for future investigation of the mechanisms of redox-modulated control at the oocyte-to-embryo transition.


Subject(s)
Cell Cycle/physiology , Cysteine/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development/physiology , Oocytes/metabolism , Animals , Cysteine/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Embryo, Nonmammalian/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Oocytes/cytology , Oxidation-Reduction , Thioredoxins/genetics , Thioredoxins/metabolism
16.
Anal Chem ; 90(1): 794-800, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29207244

ABSTRACT

Proteins can undergo oxidative cleavage by in vitro metal-catalyzed oxidation (MCO) in either the α-amidation or the diamide pathway. However, whether oxidative cleavage of polypeptide-chain occurs in biological systems remains unexplored. We describe a chemoproteomic approach to globally and site-specifically profile electrophilic protein degradants formed from peptide backbone cleavages in human proteomes, including the known N-terminal α-ketoacyl products and >1000 unexpected N-terminal formyl products. Strikingly, such cleavages predominantly occur at the carboxyl side of lysine (K) and arginine (R) residues across native proteomes in situ, while MCO-induced oxidative cleavages randomly distribute on peptide/protein sequences in vitro. Furthermore, ionizing radiation-induced reactive oxygen species (ROS) also generate random oxidative cleavages in situ. These findings suggest that the endogenous formation of N-formyl and N-α-ketoacyl degradants in biological systems is more likely regulated by a previously unknown mechanism with a trypsin-like specificity, rather than the random oxidative damage as previously thought. More generally, our study highlights the utility of quantitative chemoproteomics in combination with unrestricted search tools as a viable strategy to discover unexpected chemical modifications of proteins labeled with active-based probes.


Subject(s)
Peptide Fragments/chemistry , Proteome/chemistry , Proteomics/methods , Arginine/chemistry , HeLa Cells , Humans , Hydrazines/chemistry , Lysine/chemistry , Oxidation-Reduction , Peptide Fragments/chemical synthesis , Proteolysis
18.
Cell Chem Biol ; 24(11): 1416-1427.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28988947

ABSTRACT

Electrophilic groups, such as Michael acceptors, expoxides, are common motifs in natural products (NPs). Electrophilic NPs can act through covalent modification of cysteinyl thiols on functional proteins, and exhibit potent cytotoxicity and anti-inflammatory/cancer activities. Here we describe a new chemoproteomic strategy, termed multiplexed thiol reactivity profiling (MTRP), and its use in target discovery of electrophilic NPs. We demonstrate the utility of MTRP by identifying cellular targets of gambogic acid, an electrophilic NP that is currently under evaluation in clinical trials as anticancer agent. Moreover, MTRP enables simultaneous comparison of seven structurally diversified α,ß-unsaturated γ-lactones, which provides insights into the relative proteomic reactivity and target preference of diverse structural scaffolds coupled to a common electrophilic motif and reveals various potential druggable targets with liganded cysteines. We anticipate that this new method for thiol reactivity profiling in a multiplexed manner will find broad application in redox biology and drug discovery.


Subject(s)
Biological Products/chemistry , Sulfhydryl Compounds/chemistry , Cell Line, Tumor , Chaperonin 60/antagonists & inhibitors , Chaperonin 60/metabolism , Chromatography, High Pressure Liquid , Cysteine/chemistry , Humans , Lactones/chemistry , Lactones/metabolism , Tandem Mass Spectrometry , Xanthones/chemistry , Xanthones/metabolism
19.
Chem Res Toxicol ; 30(10): 1797-1803, 2017 10 16.
Article in English | MEDLINE | ID: mdl-28960974

ABSTRACT

Reactive metabolites (RM) formed from bioactivation of drugs can covalently modify liver proteins and cause mechanism-based inactivation of major cytochrome P450 (CYP450) enzymes. Risk of bioactivation of a test compound is routinely examined as part of lead optimization efforts in drug discovery. Here we described a chemoproteomic platform to assess in vitro and in vivo bioactivation potential of drugs. This platform enabled us to determine reactivity of thousands of proteomic cysteines toward RMs of diclofenac formed in human liver microsomes and living animals. We pinpointed numerous reactive cysteines as the targets of RMs of diclofenac, including the active (heme-binding) sites on several key CYP450 isoforms (1A2, 2E1 and 3A4 for human, 2C39 and 3A11 for mouse). This general platform should be applied to other drugs, drug candidates, and xenobiotics with potential hepatoxicity, including environmental organic substances, bioactive natural products, and traditional Chinese medicine.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/adverse effects , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/adverse effects , Microsomes, Liver/drug effects , Proteomics , Xenobiotics/adverse effects , Animals , Cytochrome P-450 Enzyme Inhibitors/metabolism , Diclofenac/metabolism , Dose-Response Relationship, Drug , Humans , Mice , Microsomes, Liver/metabolism , Xenobiotics/metabolism
20.
Mol Cell Proteomics ; 16(10): 1815-1828, 2017 10.
Article in English | MEDLINE | ID: mdl-28827280

ABSTRACT

Protein cysteinyl residues are the mediators of hydrogen peroxide (H2O2)-dependent redox signaling. However, site-specific mapping of the selectivity and dynamics of these redox reactions in cells poses a major analytical challenge. Here we describe a chemoproteomic platform to systematically and quantitatively analyze the reactivity of thousands of cysteines toward H2O2 in human cells. We identified >900 H2O2-sensitive cysteines, which are defined as the H2O2-dependent redoxome. Although redox sites associated with antioxidative and metabolic functions are consistent, most of the H2O2-dependent redoxome varies dramatically between different cells. Structural analyses reveal that H2O2-sensitive cysteines are less conserved than their redox-insensitive counterparts and display distinct sequence motifs, structural features, and potential for crosstalk with lysine modifications. Notably, our chemoproteomic platform also provides an opportunity to predict oxidation-triggered protein conformational changes. The data are freely accessible as a resource at http://redox.ncpsb.org/OXID/.


Subject(s)
Cysteine/chemistry , Hydrogen Peroxide/chemistry , Proteome/analysis , Proteomics/methods , Amino Acid Motifs , Cell Line, Tumor , Computer Simulation , Cysteine/analysis , HEK293 Cells , Hep G2 Cells , Humans , Hydrogen Peroxide/analysis , Lysine/analysis , Lysine/chemistry , Oxidation-Reduction , Protein Conformation , Proteome/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...