Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Anal Sci ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847963

ABSTRACT

Green leafy vegetables are essential for a balanced diet, providing vital nutrients for overall well-being. However, concerns arise due to contamination with toxic substances, such as arsenic, posing risks to food safety and human health. This study analyzes inorganic (iAs), monomethyl (MMA), and dimethyl arsenic (DMA) in specific leafy vegetables (Amaranthus tricolor L., Corchorus olitorius L., Cordia myxa L., Hibiscus sabdariffa L., Ipomoea batatas (L.) Lam., Moringa oleifera Lam., and Spinacia oleracea L.) grown in the heavily polluted Ambagarh Chouki region, Chhattisgarh, India. Concentrations of DMA, MMA, and iAs ranged from 0 to 155, 0 to 7, and 131 to 3579 mg·kg-1, respectively. The health quotient (HQ) for iAs ranged between 0.37 and 3.78, with an average value of 2.58 ± 1.08.

2.
Cell Metab ; 17(2): 291-302, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395175

ABSTRACT

Posttranslational modifications play central roles in myriad biological pathways including circadian regulation. We employed a circadian proteomic approach to demonstrate that circadian timing of phosphorylation is a critical factor in regulating complex GSK3ß-dependent pathways and identified O-GlcNAc transferase (OGT) as a substrate of GSK3ß. Interestingly, OGT activity is regulated by GSK3ß; hence, OGT and GSK3ß exhibit reciprocal regulation. Modulating O-GlcNAcylation levels alter circadian period length in both mice and Drosophila; conversely, protein O-GlcNAcylation is circadianly regulated. Central clock proteins, Clock and Period, are reversibly modified by O-GlcNAcylation to regulate their transcriptional activities. In addition, O-GlcNAcylation of a region in PER2 known to regulate human sleep phase (S662-S674) competes with phosphorylation of this region, and this interplay is at least partly mediated by glucose levels. Together, these results indicate that O-GlcNAcylation serves as a metabolic sensor for clock regulation and works coordinately with phosphorylation to fine-tune circadian clock.


Subject(s)
Acetylglucosamine/metabolism , Circadian Clocks , Glucose/metabolism , Adenosine Triphosphate/analogs & derivatives , Amino Acid Sequence , Animals , CLOCK Proteins/chemistry , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Glycosylation , Humans , Mice , Molecular Sequence Data , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , Substrate Specificity , Transcription, Genetic , Transfection
3.
Nat Struct Mol Biol ; 19(11): 1108-15, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064645

ABSTRACT

Promoter-proximal pausing by RNA polymerase II (Pol II) ensures gene-specific regulation and RNA quality control. Structural considerations suggested a requirement for initiation-factor eviction in elongation-factor engagement and pausing of transcription complexes. Here we show that selective inhibition of Cdk7--part of TFIIH--increases TFIIE retention, prevents DRB sensitivity-inducing factor (DSIF) recruitment and attenuates pausing in human cells. Pause release depends on Cdk9-cyclin T1 (P-TEFb); Cdk7 is also required for Cdk9-activating phosphorylation and Cdk9-dependent downstream events--Pol II C-terminal domain Ser2 phosphorylation and histone H2B ubiquitylation--in vivo. Cdk7 inhibition, moreover, impairs Pol II transcript 3'-end formation. Cdk7 thus acts through TFIIE and DSIF to establish, and through P-TEFb to relieve, barriers to elongation: incoherent feedforward that might create a window to recruit RNA-processing machinery. Therefore, cyclin-dependent kinases govern Pol II handoff from initiation to elongation factors and cotranscriptional RNA maturation.


Subject(s)
Cyclin-Dependent Kinases/physiology , RNA Polymerase II/metabolism , Transcription Elongation, Genetic/physiology , Transcription Initiation, Genetic/physiology , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinases/metabolism , HCT116 Cells , Histones/metabolism , Humans , Immunoblotting , Nuclear Proteins/metabolism , Phosphorylation , Transcription Factors/metabolism , Transcription Factors, TFII/metabolism , Transcriptional Elongation Factors , Ubiquitination , Cyclin-Dependent Kinase-Activating Kinase
4.
PLoS Genet ; 8(8): e1002935, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22927831

ABSTRACT

The cyclin-dependent kinases (CDKs) that promote cell-cycle progression are targets for negative regulation by signals from damaged or unreplicated DNA, but also play active roles in response to DNA lesions. The requirement for activity in the face of DNA damage implies that there are mechanisms to insulate certain CDKs from checkpoint inhibition. It remains difficult, however, to assign precise functions to specific CDKs in protecting genomic integrity. In mammals, Cdk2 is active throughout S and G2 phases, but Cdk2 protein is dispensable for survival, owing to compensation by other CDKs. That plasticity obscured a requirement for Cdk2 activity in proliferation of human cells, which we uncovered by replacement of wild-type Cdk2 with a mutant version sensitized to inhibition by bulky adenine analogs. Here we show that transient, selective inhibition of analog-sensitive (AS) Cdk2 after exposure to ionizing radiation (IR) enhances cell-killing. In extracts supplemented with an ATP analog used preferentially by AS kinases, Cdk2(as) phosphorylated the Nijmegen Breakage Syndrome gene product Nbs1-a component of the conserved Mre11-Rad50-Nbs1 complex required for normal DNA damage repair and checkpoint signaling-dependent on a consensus CDK recognition site at Ser432. In vivo, selective inhibition of Cdk2 delayed and diminished Nbs1-Ser432 phosphorylation during S phase, and mutation of Ser432 to Ala or Asp increased IR-sensitivity. Therefore, by chemical genetics, we uncovered both a non-redundant requirement for Cdk2 activity in response to DNA damage and a specific target of Cdk2 within the DNA repair machinery.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , DNA Damage/radiation effects , Nuclear Proteins/metabolism , Radiation, Ionizing , Acid Anhydride Hydrolases , Cell Cycle , DNA Repair , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , MRE11 Homologue Protein , Phosphorylation
5.
PLoS One ; 7(6): e40000, 2012.
Article in English | MEDLINE | ID: mdl-22768194

ABSTRACT

Protein kinase C iota is required for various cell biological processes including epithelial tissue polarity and organ morphogenesis. To gain mechanistic insight into different roles of this kinase, it is essential to identify specific substrate proteins in their cellular context. The analog-sensitive kinase method provides a powerful tool for the identification of kinase substrates under in vivo conditions. However, it has remained a major challenge to establish screens based on this method in multicellular model organisms. Here, we report the methodology for in vivo conditions using the analog-sensitive kinase method in a genetically-tractable vertebrate model organism, the zebrafish. With this approach, kinase substrates can uniquely be labeled in the developing zebrafish embryo using bulky ATPγS analogs which results in the thiophosphorylation of substrates. The labeling of kinase substrates with a thiophosphoester epitope differs from phosphoesters that are generated by all other kinases and allows for an enrichment of thiophosphopeptides by immunoaffinity purification. This study provides the foundation for using the analog-sensitive kinase method in the context of complex vertebrate development, physiology, or disease.


Subject(s)
Enzyme Assays/methods , Isoenzymes/metabolism , Protein Kinase C/metabolism , Zebrafish/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Embryo, Nonmammalian/enzymology , Isoenzymes/chemistry , Molecular Sequence Data , Mutant Proteins/chemistry , Phosphorylation , Protein Kinase C/chemistry , Substrate Specificity , Sulfhydryl Compounds/metabolism , Zebrafish/embryology
6.
Mol Cell ; 44(6): 878-92, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22137581

ABSTRACT

The energy-sensing AMP-activated protein kinase (AMPK) is activated by low nutrient levels. Functions of AMPK, other than its role in cellular metabolism, are just beginning to emerge. Here we use a chemical genetics screen to identify direct substrates of AMPK in human cells. We find that AMPK phosphorylates 28 previously unidentified substrates, several of which are involved in mitosis and cytokinesis. We identify the residues phosphorylated by AMPK in vivo in several substrates, including protein phosphatase 1 regulatory subunit 12C (PPP1R12C) and p21-activated protein kinase (PAK2). AMPK-induced phosphorylation is necessary for PPP1R12C interaction with 14-3-3 and phosphorylation of myosin regulatory light chain. Both AMPK activity and PPP1R12C phosphorylation are increased in mitotic cells and are important for mitosis completion. These findings suggest that AMPK coordinates nutrient status with mitosis completion, which may be critical for the organism's response to low nutrients during development, or in adult stem and cancer cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic/genetics , Mitosis/genetics , AMP-Activated Protein Kinases/genetics , Adenosine Triphosphate/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Myosin Light Chains/metabolism , Phosphorylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Substrate Specificity , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
7.
Mol Cell ; 42(5): 624-36, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21658603

ABSTRACT

Multiple cyclin-dependent kinases (CDKs) control eukaryotic cell division, but assigning specific functions to individual CDKs remains a challenge. During the mammalian cell cycle, Cdk2 forms active complexes before Cdk1, but lack of Cdk2 protein does not block cell-cycle progression. To detect requirements and define functions for Cdk2 activity in human cells when normal expression levels are preserved, and nonphysiologic compensation by other CDKs is prevented, we replaced the wild-type kinase with a version sensitized to specific inhibition by bulky adenine analogs. The sensitizing mutation also impaired a noncatalytic function of Cdk2 in restricting assembly of cyclin A with Cdk1, but this defect could be corrected by both inhibitory and noninhibitory analogs. This allowed either chemical rescue or selective antagonism of Cdk2 activity in vivo, to uncover a requirement in cell proliferation, and nonredundant, rate-limiting roles in restriction point passage and S phase entry.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 2/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Cell Line , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/genetics , G1 Phase/drug effects , G1 Phase/physiology , Humans , Protein Structure, Tertiary , S Phase/drug effects , S Phase/physiology
8.
J Biol Chem ; 285(15): 11093-9, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20157113

ABSTRACT

The reversible association and dissociation of a metabolic multi-enzyme complex participating in de novo purine biosynthesis, the purinosome, was demonstrated in live cells to respond to the levels of purine nucleotides in the culture media. We also took advantage of in vitro proteomic scale studies of cellular substrates of human protein kinases (e.g. casein kinase II (CK2) and Akt), that implicated several de novo purine biosynthetic enzymes as kinase substrates. Here, we successfully identified that purinosome formation in vivo was significantly promoted in HeLa cells by the addition of small-molecule CK2-specific inhibitors (i.e. 4,5,6,7-tetrabromo-1H-benzimidazole, 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole, tetrabromocinammic acid, 4,4',5,5',6,6'-hexahydroxydiphenic acid 2,2',6,6'-dilactone (ellagic acid) as well as by silencing the endogenous human CK2alpha catalytic subunit with small interfering RNA. However, 4,5,6,7-tetrabromobenzotriazole, another CK2-specific inhibitor, triggered the dissociation of purinosome clusters in HeLa cells. Although the mechanism by which 4,5,6,7-tetrabromobenzotriazole affects purinosome clustering is not clear, we were capable of chemically reversing purinosome formation in cells by the sequential addition of two CK2 inhibitors. Collectively, we provide compelling cellular evidence that CK2-mediated pathways reversibly regulate purinosome assembly, and thus the purinosome may be one of the ultimate targets of kinase inhibitors.


Subject(s)
Casein Kinase II/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Casein Kinase II/metabolism , Catalysis , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Gene Silencing , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Models, Biological , Models, Chemical , Purines/chemistry , RNA Interference , RNA, Small Interfering/metabolism
9.
Curr Protoc Chem Biol ; 2(1): 15-36, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-23836541

ABSTRACT

Mapping kinase-substrate interactions demands robust methods to rapidly and unequivocally identify substrates from complex protein mixtures. Toward this goal, we present a method in which a kinase, engineered to utilize synthetic ATPγS analogs, specifically thiophosphorylates its substrates in a complex lysate. The thiophosphate label provides a bio-orthogonal tag that can be used to affinity purify and identify labeled proteins. Following the labeling reaction, proteins are digested with trypsin; thiol-containing peptides are then covalently captured and non-thiol-containing peptides are washed from the resin. Oxidation-promoted hydrolysis, at sites of thiophosphorylation, releases phosphopeptides for analysis by tandem mass spectrometry. By incorporating two specificity gates-kinase engineering and peptide affinity purification-this method yields high-confidence substrate identifications. This method gives both the identity of the substrates and phosphorylation-site localization. With this information, investigators can analyze the biological significance of the phosphorylation mark immediately following confirmation of the kinase-substrate relationship. Here, we provide an optimized version of this technique to further enable widespread utilization of this technology. Curr. Protoc. Chem Biol. 2:15-36. © 2010 by John Wiley & Sons, Inc.

10.
Anal Biochem ; 397(2): 139-43, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19819215

ABSTRACT

Histidine-aspartic acid phosphotransfer pathways are central components of prokaryotic signal transduction pathways and are also found in many eukaryotes. Tools to study histidine kinases, however, are currently quite limited. In this article, we present a new tool to study histidine-aspartic acid phosphotransfer pathways. We show that many histidine kinases will accept ATPgammaS as a substrate to form a stable thiophosphohistidine even when they do not form stable phosphohistidines using the natural substrate ATP. An antibody that has previously been used to detect thiophosphorylated serine, threonine, and tyrosine residues is shown to recognize thiophosphohistidine and thiophosphoaspartic acid residues. Histidine kinase autothiophosphorylation is regulated by other protein sensor domains in the same way as autophosphorylation, and thiophosphate is transferred to downstream aspartic acid containing response regulators.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Epitopes/chemistry , Protein Kinases/analysis , Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Cloning, Molecular , Escherichia coli/enzymology , Histidine/analogs & derivatives , Histidine/analysis , Histidine/immunology , Histidine Kinase
11.
Mol Cell ; 36(3): 393-404, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19917248

ABSTRACT

A preference for homologs over sister chromatids in homologous recombination is a fundamental difference in meiotic versus mitotic cells. In budding yeast, the bias for interhomolog recombination in meiosis requires the Dmc1 recombinase and the meiosis-specific kinase Mek1, which suppresses engagement of sister chromatids by the mitotic recombinase Rad51. Here, a combination of proteomic, biochemical, and genetic approaches has identified an additional role for Mek1 in inhibiting the activity of the Rad51 recombinase through phosphorylation of its binding partner, Rad54. Rad54 phosphorylation of threonine 132 attenuates complex formation with Rad51, and a negative charge at this position reduces Rad51 function in vitro and in vivo. Thus, Mek1 phosphorylation provides a dynamic means of controlling recombination partner choice in meiosis in two ways: (1) it reduces Rad51 activity through inhibition of Rad51/Rad54 complex formation, and (2) it suppresses Rad51-mediated strand invasion of sister chromatids via a Rad54-independent mechanism.


Subject(s)
DNA Repair Enzymes/metabolism , MAP Kinase Kinase 1/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division , DNA Breaks, Double-Stranded , DNA Helicases , DNA Repair , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Immunoblotting , MAP Kinase Kinase 1/genetics , Mass Spectrometry , Meiosis , Mutation , Phosphorylation , Protein Binding , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Spores, Fungal/genetics , Threonine/metabolism
12.
Mol Cell Biol ; 29(17): 4852-63, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19581288

ABSTRACT

The Saccharomyces cerevisiae kinase Bur1 is involved in coupling transcription elongation to chromatin modification, but not all important Bur1 targets in the elongation complex are known. Using a chemical genetics strategy wherein Bur1 kinase was engineered to be regulated by a specific inhibitor, we found that Bur1 phosphorylates the Spt5 C-terminal repeat domain (CTD) both in vivo and in isolated elongation complexes in vitro. Deletion of the Spt5 CTD or mutation of the Spt5 serines targeted by Bur1 reduces recruitment of the PAF complex, which functions to recruit factors involved in chromatin modification and mRNA maturation to elongating polymerase II (Pol II). Deletion of the Spt5 CTD showed the same defect in PAF recruitment as rapid inhibition of Bur1 kinase activity, and this Spt5 mutation led to a decrease in histone H3K4 trimethylation. Brief inhibition of Bur1 kinase activity in vivo also led to a significant decrease in phosphorylation of the Pol II CTD at Ser-2, showing that Bur1 also contributes to Pol II Ser-2 phosphorylation. Genetic results suggest that Bur1 is essential for growth because it targets multiple factors that play distinct roles in transcription.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinases/metabolism , Macromolecular Substances/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Chromosomal Proteins, Non-Histone/genetics , Cyclin-Dependent Kinases/genetics , Humans , Mutation , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics
13.
Mol Cell ; 33(6): 738-51, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19328067

ABSTRACT

Cyclin-dependent kinases (CDKs) are subunits of transcription factor (TF) IIH and positive transcription elongation factor b (P-TEFb). To define their functions, we mutated the TFIIH-associated kinase Mcs6 and P-TEFb homologs Cdk9 and Lsk1 of fission yeast, making them sensitive to inhibition by bulky purine analogs. Selective inhibition of Mcs6 or Cdk9 blocks cell division, alters RNA polymerase (Pol) II carboxyl-terminal domain (CTD) phosphorylation, and represses specific, overlapping subsets of transcripts. At a common target gene, both CDKs must be active for normal Pol II occupancy, and Spt5-a CDK substrate and regulator of elongation-accumulates disproportionately to Pol II when either kinase is inhibited. In contrast, Mcs6 activity is sufficient-and necessary-to recruit the Cdk9/Pcm1 (mRNA cap methyltransferase) complex. In vitro, phosphorylation of the CTD by Mcs6 stimulates subsequent phosphorylation by Cdk9. We propose that TFIIH primes the CTD and promotes recruitment of P-TEFb/Pcm1, serving to couple elongation and capping of select pre-mRNAs.


Subject(s)
Positive Transcriptional Elongation Factor B/genetics , RNA Caps/genetics , Schizosaccharomyces/metabolism , Transcription Factor TFIIH/genetics , Transcription, Genetic , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Positive Transcriptional Elongation Factor B/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Transcription Factor TFIIH/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Cyclin-Dependent Kinase-Activating Kinase
14.
Mol Cell ; 32(5): 662-72, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-19061641

ABSTRACT

In metazoans, different cyclin-dependent kinases (CDKs) bind preferred cyclin partners to coordinate cell division. Here, we investigate these preferences in human cells and show that cyclin A assembles with Cdk1 only after complex formation with Cdk2 reaches a plateau during late S and G2 phases. To understand the basis for Cdk2's competitive advantage, despite Cdk1's greater abundance, we dissect their activation pathways by chemical genetics. Cdk1 and Cdk2 are activated by kinetically distinct mechanisms, even though they share the same CDK-activating kinase (CAK), Cdk7. We recapitulate cyclin A's selectivity for Cdk2 in extracts and override it with a yeast CAK that phosphorylates monomeric Cdk1, redirecting Cdk1 into a pathway normally restricted to Cdk2. Conversely, upon Cdk7 inhibition in vivo, cyclin B, which normally binds Cdk1 nearly exclusively, is diverted to Cdk2. Therefore, differential ordering of common activation steps promotes CDK-cyclin specificity, with Cdk7 acting catalytically to enforce fidelity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , Animals , Cell Cycle , Cell Extracts , Cyclin-Dependent Kinases/metabolism , Enzyme Activation , Enzyme Stability , HCT116 Cells , Humans , Models, Biological , Phosphates/metabolism , Phosphorylation , Phosphothreonine/metabolism , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Cyclin-Dependent Kinase-Activating Kinase
15.
Nat Methods ; 4(6): 511-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17486086

ABSTRACT

The ubiquitous nature of protein phosphorylation makes it challenging to map kinase-substrate relationships, which is a necessary step toward defining signaling network architecture. To trace the activity of individual kinases, we developed a semisynthetic reaction scheme, which results in the affinity tagging of substrates of the kinase in question. First, a kinase, engineered to use a bio-orthogonal ATPgammaS analog, catalyzes thiophosphorylation of its direct substrates. Second, alkylation of thiophosphorylated serine, threonine or tyrosine residues creates an epitope for thiophosphate ester-specific antibodies. We demonstrated the generality of semisynthetic epitope construction with 13 diverse kinases: JNK1, p38alpha MAPK, Erk1, Erk2, Akt1, PKCdelta, PKCepsilon, Cdk1/cyclinB, CK1, Cdc5, GSK3beta, Src and Abl. Application of this approach, in cells isolated from a mouse that expressed endogenous levels of an analog-specific (AS) kinase (Erk2), allowed purification of a direct Erk2 substrate.


Subject(s)
Epitopes/chemistry , Epitopes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Haptens/chemistry , Haptens/metabolism , Adenosine Triphosphate/analogs & derivatives , Amino Acid Sequence , Animals , Epitopes/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Duplication , Haptens/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Isotope Labeling/methods , Mice , Mice, Knockout , Organothiophosphates/chemistry , Organothiophosphates/metabolism , Substrate Specificity
16.
ACS Chem Biol ; 1(3): 139-40, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-17163659

ABSTRACT

A recent study analyzed the transcriptional effects induced by a panel of non-steroidal glucocorticoid receptor modulators. The authors discover patterns of cell-, gene-, and mechanism-specific regulation, with implications for development of improved anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Genomics , Receptors, Glucocorticoid/genetics , Transcription, Genetic , Animals , Homeostasis , Ligands , Models, Biological , Receptors, Cytoplasmic and Nuclear/genetics , Selective Estrogen Receptor Modulators/pharmacokinetics , Substrate Specificity
17.
J Am Chem Soc ; 127(15): 5288-9, 2005 Apr 20.
Article in English | MEDLINE | ID: mdl-15826144

ABSTRACT

Protein phosphorylation is a major mechanism of post-translational protein modification used to control cellular signaling. A challenge in phosphoproteomics is to identify the direct substrates of each protein kinase. Herein, we describe a chemical strategy for delivery of a bio-orthogonal affinity tag to the substrates of an individual protein kinase. The kinase of interest is engineered to transfer a phosphorothioate moiety to phosphoacceptor hydroxyl groups on direct substrates. In a second nonenzymatic step, the introduced phosphorothioate is alkylated with p-nitrobenzylmesylate (PNBM). Antibodies directed against the alkylated phosphorothioate epitope recognize these labeled substrates, but not alkylation products of other cellular nucleophiles. This strategy is demonstrated with Cdk1/cyclinB substrates using ELISA, western blotting, and immunoprecipitation in the context of whole cell lysates.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Affinity Labels/chemistry , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/chemistry , Histones/chemistry , Immunoconjugates/chemistry , Protein-Tyrosine Kinases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Affinity Labels/metabolism , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , HeLa Cells , Histones/isolation & purification , Histones/metabolism , Humans , Immunoconjugates/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunoglobulins/chemistry , Immunoglobulins/metabolism , Mesylates/chemistry , Protein-Tyrosine Kinases/isolation & purification , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
18.
J Am Chem Soc ; 126(30): 9160-1, 2004 Aug 04.
Article in English | MEDLINE | ID: mdl-15281787

ABSTRACT

The reversible phosphorylation of proteins is one of the most important mechanisms for the regulation of signal transduction cascades. Recently, there has been substantial progress made in the identification of new phosphoproteins and phosphorylation sites. Unfortunately, there are very few methods available that allow this information to be used to identify the upstream kinase responsible for the phosphorylation event. Herein, we describe a new method that allows the cross-linking of a substrate of interest to its upstream kinase. This method relies upon a novel, mechanism-based cross-linker and the replacement of the phosphorylated residue with a cysteine residue. The application of this method to a number of kinase-peptide substrate pairs is described.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine/analogs & derivatives , Cross-Linking Reagents/chemistry , Protein Kinases/chemistry , Protein Kinases/metabolism , Adenosine/chemistry , Adenosine/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding Sites , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/metabolism , Cysteine/chemistry , Cysteine/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Serine/chemistry , Serine/metabolism , Substrate Specificity , Threonine/chemistry , Threonine/metabolism , o-Phthalaldehyde/chemistry , o-Phthalaldehyde/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...