51
|
Kundu K, Costa F, Huber M, Reth M, Backofen R. Semi-supervised prediction of SH2-peptide interactions from imbalanced high-throughput data. PLoS One 2013; 8:e62732. [PMID: 23690949 PMCID: PMC3656881 DOI: 10.1371/journal.pone.0062732] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/22/2013] [Indexed: 01/08/2023] Open
Abstract
Src homology 2 (SH2) domains are the largest family of the peptide-recognition modules (PRMs) that bind to phosphotyrosine containing peptides. Knowledge about binding partners of SH2-domains is key for a deeper understanding of different cellular processes. Given the high binding specificity of SH2, in-silico ligand peptide prediction is of great interest. Currently however, only a few approaches have been published for the prediction of SH2-peptide interactions. Their main shortcomings range from limited coverage, to restrictive modeling assumptions (they are mainly based on position specific scoring matrices and do not take into consideration complex amino acids inter-dependencies) and high computational complexity. We propose a simple yet effective machine learning approach for a large set of known human SH2 domains. We used comprehensive data from micro-array and peptide-array experiments on 51 human SH2 domains. In order to deal with the high data imbalance problem and the high signal-to-noise ration, we casted the problem in a semi-supervised setting. We report competitive predictive performance w.r.t. state-of-the-art. Specifically we obtain 0.83 AUC ROC and 0.93 AUC PR in comparison to 0.71 AUC ROC and 0.87 AUC PR previously achieved by the position specific scoring matrices (PSSMs) based SMALI approach. Our work provides three main contributions. First, we showed that better models can be obtained when the information on the non-interacting peptides (negative examples) is also used. Second, we improve performance when considering high order correlations between the ligand positions employing regularization techniques to effectively avoid overfitting issues. Third, we developed an approach to tackle the data imbalance problem using a semi-supervised strategy. Finally, we performed a genome-wide prediction of human SH2-peptide binding, uncovering several findings of biological relevance. We make our models and genome-wide predictions, for all the 51 SH2-domains, freely available to the scientific community under the following URLs: http://www.bioinf.uni-freiburg.de/Software/SH2PepInt/SH2PepInt.tar.gz and http://www.bioinf.uni-freiburg.de/Software/SH2PepInt/Genome-wide-predictions.tar.gz, respectively.
Collapse
Affiliation(s)
- Kousik Kundu
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
- Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany
| | - Fabrizio Costa
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, University Clinic, RWTH Aachen University, Aachen, Germany
| | - Michael Reth
- Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany
- Department of Molecular Immunology, Max Planck Institute of Immunology, Freiburg, Germany
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
- Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany
- Centre for Biological Systems Analysis (ZBSA), University of Freiburg, Freiburg, Germany
- Center for non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg, Denmark
- * E-mail:
| |
Collapse
|
52
|
Zhao B, Tan PH, Li SSC, Pei D. Systematic characterization of the specificity of the SH2 domains of cytoplasmic tyrosine kinases. J Proteomics 2013; 81:56-69. [PMID: 23313216 DOI: 10.1016/j.jprot.2012.12.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 12/18/2012] [Accepted: 12/29/2012] [Indexed: 01/26/2023]
Abstract
Cytoplasmic tyrosine kinases (CTK) generally contain a Src-homology 2 (SH2) domain, whose role in the CTK family is not fully understood. Here we report the determination of the specificity of 25 CTK SH2 domains by screening one-bead-one-compound (OBOC) peptide libraries. Based on the peptide sequences selected by the SH2 domains, we built Support Vector Machine (SVM) models for the prediction of binding ligands for the SH2 domains. These models yielded support for the progressive phosphorylation model for CTKs in which the overlapping specificity of the CTK SH2 and kinase domains has been proposed to facilitate targeting of the CTK substrates with at least two potential phosphotyrosine (pTyr) sites. We curated 93 CTK substrates with at least two pTyr sites catalyzed by the same CTK, and showed that 71% of these substrates had at least two pTyr sites predicted to bind a common CTK SH2 domain. More importantly, we found 34 instances where there was at least one pTyr site predicted to be recognized by the SH2 domain of the same CTK, suggesting that the SH2 and kinase domains of the CTKs may cooperate to achieve progressive phosphorylation of a protein substrate. This article is part of a Special Issue entitled: From protein structures to clinical applications.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Biochemistry and Siebens-Drake Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada N6A 5C1
| | | | | | | |
Collapse
|
53
|
Tanaka H, Akagi KI, Oneyama C, Tanaka M, Sasaki Y, Kanou T, Lee YH, Yokogawa D, Dobenecker MW, Nakagawa A, Okada M, Ikegami T. Identification of a new interaction mode between the Src homology 2 domain of C-terminal Src kinase (Csk) and Csk-binding protein/phosphoprotein associated with glycosphingolipid microdomains. J Biol Chem 2013; 288:15240-54. [PMID: 23548896 DOI: 10.1074/jbc.m112.439075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proteins with Src homology 2 (SH2) domains play major roles in tyrosine kinase signaling. Structures of many SH2 domains have been studied, and the regions involved in their interactions with ligands have been elucidated. However, these analyses have been performed using short peptides consisting of phosphotyrosine followed by a few amino acids, which are described as the canonical recognition sites. Here, we report the solution structure of the SH2 domain of C-terminal Src kinase (Csk) in complex with a longer phosphopeptide from the Csk-binding protein (Cbp). This structure, together with biochemical experiments, revealed the existence of a novel binding region in addition to the canonical phosphotyrosine 314-binding site of Cbp. Mutational analysis of this second region in cells showed that both canonical and novel binding sites are required for tumor suppression through the Cbp-Csk interaction. Furthermore, the data indicate an allosteric connection between Cbp binding and Csk activation that arises from residues in the βB/βC loop of the SH2 domain.
Collapse
Affiliation(s)
- Hiroaki Tanaka
- Institute for Protein Research, Osaka University, 3-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Yeung MCL, Yam VWW. Phosphate derivative-induced supramolecular assembly and NIR-emissive behaviour of alkynylplatinum(ii) terpyridine complexes for real-time monitoring of enzymatic activities. Chem Sci 2013. [DOI: 10.1039/c3sc50383f] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
55
|
Kaneko T, Joshi R, Feller SM, Li SS. Phosphotyrosine recognition domains: the typical, the atypical and the versatile. Cell Commun Signal 2012; 10:32. [PMID: 23134684 PMCID: PMC3507883 DOI: 10.1186/1478-811x-10-32] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/09/2012] [Indexed: 12/21/2022] Open
Abstract
SH2 domains are long known prominent players in the field of phosphotyrosine recognition within signaling protein networks. However, over the years they have been joined by an increasing number of other protein domain families that can, at least with some of their members, also recognise pTyr residues in a sequence-specific context. This superfamily of pTyr recognition modules, which includes substantial fractions of the PTB domains, as well as much smaller, or even single member fractions like the HYB domain, the PKCδ and PKCθ C2 domains and RKIP, represents a fascinating, medically relevant and hence intensely studied part of the cellular signaling architecture of metazoans. Protein tyrosine phosphorylation clearly serves a plethora of functions and pTyr recognition domains are used in a similarly wide range of interaction modes, which encompass, for example, partner protein switching, tandem recognition functionalities and the interaction with catalytically active protein domains. If looked upon closely enough, virtually no pTyr recognition and regulation event is an exact mirror image of another one in the same cell. Thus, the more we learn about the biology and ultrastructural details of pTyr recognition domains, the more does it become apparent that nature cleverly combines and varies a few basic principles to generate a sheer endless number of sophisticated and highly effective recognition/regulation events that are, under normal conditions, elegantly orchestrated in time and space. This knowledge is also valuable when exploring pTyr reader domains as diagnostic tools, drug targets or therapeutic reagents to combat human diseases.
Collapse
Affiliation(s)
- Tomonori Kaneko
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| | | | | | | |
Collapse
|
56
|
Kaneko T, Huang H, Cao X, Li X, Li C, Voss C, Sidhu SS, Li SSC. Superbinder SH2 Domains Act as Antagonists of Cell Signaling. Sci Signal 2012; 5:ra68. [DOI: 10.1126/scisignal.2003021] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
57
|
Georghiou G, Kleiner RE, Pulkoski-Gross M, Liu DR, Seeliger MA. Highly specific, bisubstrate-competitive Src inhibitors from DNA-templated macrocycles. Nat Chem Biol 2012; 8:366-74. [PMID: 22344177 PMCID: PMC3307835 DOI: 10.1038/nchembio.792] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 12/01/2011] [Indexed: 01/21/2023]
Abstract
Protein kinases are attractive therapeutic targets, but their high sequence and structural conservation complicates the development of specific inhibitors. We recently discovered from a DNA-templated macrocycle library inhibitors with unusually high selectivity among Src-family kinases. Starting from these compounds, we developed and characterized in molecular detail potent macrocyclic inhibitors of Src kinase and its cancer-associated gatekeeper mutant. We solved two co-crystal structures of macrocycles bound to Src kinase. These structures reveal the molecular basis of the combined ATP- and substrate peptide-competitive inhibitory mechanism and the remarkable kinase specificity of the compounds. The most potent compounds inhibit Src activity in cultured mammalian cells. Our work establishes that macrocycles can inhibit protein kinases through a bi-substrate competitive mechanism with high potency and exceptional specificity, reveals the precise molecular basis for their desirable properties, and provides new insights into the development of Src-specific inhibitors with potential therapeutic relevance.
Collapse
Affiliation(s)
- George Georghiou
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | | | | | | | | |
Collapse
|
58
|
Chou MF, Schwartz D. Biological sequence motif discovery using motif-x. CURRENT PROTOCOLS IN BIOINFORMATICS 2011; Chapter 13:13.15.1-13.15.24. [PMID: 21901740 DOI: 10.1002/0471250953.bi1315s35] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Web-based motif-x program provides a simple interface to extract statistically significant motifs from large data sets, such as MS/MS post-translational modification data and groups of proteins that share a common biological function. Users upload data files and download results using common Web browsers on essentially any Web-compatible computer. Once submitted, data analyses are performed rapidly on an associated high-speed computer cluster and they produce both syntactic and image-based motif results and statistics. The protocols presented demonstrate the use of motif-x in three common user scenarios.
Collapse
Affiliation(s)
- Michael F Chou
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
59
|
Zheng Y, Asara JM, Tyner AL. Protein-tyrosine kinase 6 promotes peripheral adhesion complex formation and cell migration by phosphorylating p130 CRK-associated substrate. J Biol Chem 2011; 287:148-158. [PMID: 22084245 DOI: 10.1074/jbc.m111.298117] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Protein-tyrosine kinase 6 (PTK6) is a non-myristoylated intracellular tyrosine kinase evolutionarily related to Src kinases. Aberrant PTK6 expression and intracellular localization have been detected in human prostate tumors. In the PC3 prostate cancer cell line, the pool of endogenous activated PTK6, which is phosphorylated on tyrosine residue 342, is localized at the membrane. Expression of ectopic membrane-targeted PTK6 led to dramatic morphology changes and formation of peripheral adhesion complexes in PC3 cells. Peripheral adhesion complex formation was dependent upon PTK6 kinase activity. We demonstrated that p130 CRK-associated substrate (p130CAS) is a novel direct substrate of PTK6, and it works as a crucial adapter protein in inducing peripheral adhesion complexes. Activation of ERK5 downstream of p130CAS was indispensable for this process. Knockdown of endogenous PTK6 led to reduced cell migration and p130CAS phosphorylation, whereas knockdown of p130CAS attenuated oncogenic signaling induced by membrane-targeted PTK6, including ERK5 and AKT activation. Expression of membrane-targeted PTK6 promoted cell migration, which could be impaired by knockdown of p130CAS or ERK5. Our study reveals a novel function for PTK6 at the plasma membrane and suggests that the PTK6-p130CAS-ERK5 signaling cascade plays an important role in cancer cell migration and invasion.
Collapse
Affiliation(s)
- Yu Zheng
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center; Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115
| | - Angela L Tyner
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607.
| |
Collapse
|
60
|
Tyrosine phosphorylated c-Cbl regulates platelet functional responses mediated by outside-in signaling. Blood 2011; 118:5631-40. [PMID: 21967979 DOI: 10.1182/blood-2011-01-328807] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
c-Cbl protein functions as an E3 ligase and scaffolding protein, where 3 residues, Y700, Y731, and Y774, upon phosphorylation, have been shown to initiate several signaling cascades. In this study, we investigated the role of these phospho-tyrosine residues in the platelet functional responses after integrin engagement. We observed that c-Cbl Y700, Y731 and Y774 undergo phosphorylation upon platelet adhesion to immobilized fibrinogen, which was inhibited in the presence of PP2, a pan-src family kinase (SFK) inhibitor, suggesting that c-Cbl is phosphorylated downstream of SFKs. However, OXSI-2, a Syk inhibitor, significantly reduced c-Cbl phosphorylation at residues Y774 and Y700, without affecting Y731 phosphorylation. Interestingly, PP2 inhibited both platelet-spreading on fibrinogen as well as clot retraction, whereas OXSI-2 blocked only platelet-spreading, suggesting a differential role of these tyrosine residues. The physiologic role of c-Cbl and Y731 was studied using platelets from c-Cbl KO and c-Cbl(YF/YF) knock-in mice. c-Cbl KO and c-Cbl(YF/YF) platelets had a significantly reduced spreading over immobilized fibrinogen. Furthermore, clot retraction with c-Cbl KO and c-Cbl(YF/YF) platelets was drastically delayed. These results indicate that c-Cbl and particularly its phosphorylated residue Y731 plays an important role in platelet outside-in signaling contributing to platelet-spreading and clot retraction.
Collapse
|
61
|
Bogdelis A, Treinys R, Stankevičius E, Jurevičius J, Skeberdis VA. Src family protein tyrosine kinases modulate L-type calcium current in human atrial myocytes. Biochem Biophys Res Commun 2011; 413:116-21. [PMID: 21872572 DOI: 10.1016/j.bbrc.2011.08.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 08/15/2011] [Indexed: 10/17/2022]
Abstract
In the heart, L-type voltage dependent calcium channels (L-VDCC) provide Ca(2+) for the activation of contractile apparatus. The best described pathway for L-type Ca(2+) current (I(Ca,L)) modulation is the phosphorylation of calcium channels by cAMP-dependent protein kinase A (PKA), the activity of which is predominantly regulated in opposite manner by β-adrenergic (β-ARs) and muscarinic receptors. The role of other kinases is controversial and often depends on tissues and species used in the studies. In different studies the inhibitors of tyrosine kinases have been shown either to stimulate or inhibit, or even have a biphasic effect on I(Ca,L). Moreover, there is no clear picture about the route of activation and the site of action of cardiac Src family nonreceptor tyrosine kinases (Src-nPTKs). In the present study we used PP1, a selective inhibitor of Src-nPTKs, alone and together with different activators of I(Ca,L), and demonstrated that in human atrial myocytes (HAMs): (i) Src-nPTKs are activated concomitantly with activation of cAMP-signaling cascade; (ii) Src-nPTKs attenuate PKA-dependent stimulation of I(Ca,L) by inhibiting PKA activity; (iii) Gα(s) are not involved in the direct activation of Src-nPTKs. In this way, Src-nPTKs may provide a protecting mechanism against myocardial overload under conditions of increased sympathetic activity.
Collapse
Affiliation(s)
- Andrius Bogdelis
- Lithuanian University of Health Sciences, Institute of Cardiology, 50009 Kaunas, Lithuania
| | | | | | | | | |
Collapse
|
62
|
Christofk HR, Wu N, Cantley LC, Asara JM. Proteomic screening method for phosphopeptide motif binding proteins using peptide libraries. J Proteome Res 2011; 10:4158-64. [PMID: 21774532 DOI: 10.1021/pr200578n] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphopeptide binding domains mediate the directed and localized assembly of protein complexes essential to intracellular kinase signaling. To identify phosphopeptide binding proteins, we developed a proteomic screening method using immobilized partially degenerate phosphopeptide mixtures combined with SILAC and microcapillary LC-MS/MS. The method was used to identify proteins that specifically bound to phosphorylated peptide library affinity matrices, including pTyr, and the motifs pSer/pThr-Pro, pSer/pThr-X-X-X-pSer/pThr, pSer/pThr-Glu/Asp, or pSer/pThr-pSer/pThr in degenerate sequence contexts. Heavy and light SILAC lysates were applied to columns containing these phosphorylated and nonphosphorylated (control) peptide libraries respectively, and bound proteins were eluted, combined, digested, and analyzed by LC-MS/MS using a hybrid quadrupole-TOF mass spectrometer. Heavy/light peptide ion ratios were calculated, and peptides that yielded ratios greater than ∼3:1 were considered as being from potential phosphopeptide binding proteins since this ratio represents the lowest ratio from a known positive control. Many of those identified were known phosphopeptide-binding proteins, including the SH2 domain containing p85 subunit of PI3K bound to pTyr, 14-3-3 bound to pSer/pThr-Asp/Glu, polo-box domain containing PLK1 and Pin1 bound to pSer/pThr-Pro, and pyruvate kinase M2 binding to pTyr. Approximately half of the hits identified by the peptide library screens were novel. Protein domain enrichment analysis revealed that most pTyr hits contain SH2 domains, as expected, and to a lesser extent SH3, C1, STAT, Tyr phosphatase, Pkinase, C2, and PH domains; however, pSer/pThr motifs did not reveal enriched domains across hits.
Collapse
Affiliation(s)
- Heather R Christofk
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | | | | | | |
Collapse
|
63
|
Smejkal GB, Rivas-Morello C, Chang JHR, Freeman E, Trachtenberg AJ, Lazarev A, Ivanov AR, Kuo WP. Thermal stabilization of tissues and the preservation of protein phosphorylation states for two-dimensional gel electrophoresis. Electrophoresis 2011; 32:2206-15. [PMID: 21792998 DOI: 10.1002/elps.201100170] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Revised: 05/01/2011] [Accepted: 05/04/2011] [Indexed: 11/06/2022]
Abstract
2-DE is typically capable of discriminating proteins differing by a single phosphorylation or dephosphorylation event. However, a reliable representation of protein phosphorylation states as they occur in vivo requires that both phosphatases and kinases are rapidly and completely inactivated. Thermal stabilization of mouse cerebral cortex homogenates effectively inactivated these enzymes, as evidenced by comparison with unstabilized tissues where abscissal pI shifts were a common feature in 2-D gels. Of the 588 matched proteins separated on 2-D gels comparing stabilized and unstabilized tissues, 53 proteins exhibited greater than twofold differences in spot volume (ANOVA, p<0.05). Phosphoprotein-specific staining was corroborated by the identification of 16 phosphoproteins by nano-LC MS/MS and phosphotyrosine kinase activity assay.
Collapse
Affiliation(s)
- Gary B Smejkal
- Harvard Catalyst, The Harvard Clinical and Translational Science Center, Laboratory for Innovative Translational Technologies, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Ia KK, Jeschke GR, Deng Y, Kamaruddin MA, Williamson NA, Scanlon DB, Culvenor JG, Hossain MI, Purcell AW, Liu S, Zhu HJ, Catimel B, Turk BE, Cheng HC. Defining the substrate specificity determinants recognized by the active site of C-terminal Src kinase-homologous kinase (CHK) and identification of β-synuclein as a potential CHK physiological substrate. Biochemistry 2011; 50:6667-77. [PMID: 21699177 DOI: 10.1021/bi2001938] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
C-Terminal Src kinase-homologous kinase (CHK) exerts its tumor suppressor function by phosphorylating the C-terminal regulatory tyrosine of the Src-family kinases (SFKs). The phosphorylation suppresses their activity and oncogenic action. In addition to phosphorylating SFKs, CHK also performs non-SFK-related functions by phosphorylating other cellular protein substrates. To define these non-SFK-related functions of CHK, we used the "kinase substrate tracking and elucidation" method to search for its potential physiological substrates in rat brain cytosol. Our search revealed β-synuclein as a potential CHK substrate, and Y127 in β-synuclein as the preferential phosphorylation site. Using peptides derived from β-synuclein and positional scanning combinatorial peptide library screening, we defined the optimal substrate phosphorylation sequence recognized by the CHK active site to be E-x-[Φ/E/D]-Y-Φ-x-Φ, where Φ and x represent hydrophobic residues and any residue, respectively. Besides β-synuclein, cellular proteins containing motifs resembling this sequence are potential CHK substrates. Intriguingly, the CHK-optimal substrate phosphorylation sequence bears little resemblance to the C-terminal tail sequence of SFKs, indicating that interactions between the CHK active site and the local determinants near the C-terminal regulatory tyrosine of SFKs play only a minor role in governing specific phosphorylation of SFKs by CHK. Our results imply that recognition of SFKs by CHK is mainly governed by interactions between motifs located distally from the active site of CHK and determinants spatially separate from the C-terminal regulatory tyrosine in SFKs. Thus, besides assisting in the identification of potential CHK physiological substrates, our findings shed new light on how CHK recognizes SFKs and other protein substrates.
Collapse
Affiliation(s)
- Kim K Ia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Recruitment interactions can override catalytic interactions in determining the functional identity of a protein kinase. Proc Natl Acad Sci U S A 2011; 108:9809-14. [PMID: 21628578 DOI: 10.1073/pnas.1016337108] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The yeast Saccharomyces cerevisae has four distinct mitogen-activated protein kinase kinases (MAPKKs), each of which has a distinct functional identity characterized by communication with specific upstream and downstream partners to form distinct functional pathways. These four kinases belong to one family, sharing closely related catalytic domains. How have these four related kinases diverged to take on four distinct functional roles? The specificity of an enzyme for a particular substrate is often thought to reside in differences in the catalytic domain. However, many kinases, including MAPKKs, have modular interaction domains and motifs that have been shown to play an important role in determining the specificity of kinases through recruitment to specific partners and complexes. Here we probe the relative importance of catalytic domain interactions versus recruitment interactions in defining the functional identity of MAPKKs by asking whether we can use recruitment interactions to force other MAPKK catalytic domains to play the functional role of the mating MAPKK, Ste7. We find that two alternative MAPKKs, Pbs2 and Mkk2, can be forced to functionally replace the mating MAPKK Ste7, but only if the proper set of recruitment interactions are grafted onto their catalytic domains. These results show that within a family of kinases, recruitment interactions can play a dominant role in defining functional identity, and is consistent with a model in which new kinase functions can arise through recombination of existing catalytic domains with new interaction modules.
Collapse
|
66
|
Curtiss ML, Hostager BS, Stepniak E, Singh M, Manhica N, Knisz J, Traver G, Rennert PD, Colgan JD, Rothman PB. Fyn binds to and phosphorylates T cell immunoglobulin and mucin domain-1 (Tim-1). Mol Immunol 2011; 48:1424-31. [PMID: 21513984 DOI: 10.1016/j.molimm.2011.03.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 03/25/2011] [Accepted: 03/28/2011] [Indexed: 12/30/2022]
Abstract
The gene encoding T cell immunoglobulin and mucin domain-1 (Tim-1) is linked to atopy and asthma susceptibility in mice and humans. Tim-1 is a transmembrane protein expressed on activated lymphocytes and appears to have a role as a co-stimulatory receptor in T cells. The protein has not been shown to have enzymatic activity but contains a site within its cytoplasmic tail predicted to be a target for tyrosine kinases. Here, we show that Tim-1 can associate with the kinase Fyn, a member of the Src family of tyrosine kinases. This association does not require Fyn's kinase activity and is independent of the phosphorylation of a conserved tyrosine present within the cytoplasmic tail of Tim-1. Fyn is necessary for phosphorylation of this tyrosine in Tim-1 and the phosphorylation of Tim-1 varies with the levels of Fyn present in cells. These data suggest a role for Fyn in the signaling downstream of Tim-1.
Collapse
Affiliation(s)
- Miranda L Curtiss
- Department of Internal Medicine, University of Iowa Carver College of Medicine. 200 Hawkins Drive, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Regulation of SRC family kinases in human cancers. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:865819. [PMID: 21776389 PMCID: PMC3135246 DOI: 10.1155/2011/865819] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 02/08/2011] [Indexed: 11/22/2022]
Abstract
The nonreceptor protein tyrosine kinase Src plays a crucial role in the signal transduction pathways involved in cell division, motility, adhesion, and survival in both normal and cancer cells. Although the Src family kinases (SFKs) are activated in various types of cancers, the exact mechanisms through which they contribute to the progression of individual tumors remain to be defined. The activation of Src in human cancers may occur through a variety of mechanisms that include domain interaction and structural remodeling in response to various activators or upstream kinases and phosphatastes. Because of Src's prominent roles in invasion and tumor progression, epithelial-to-mesenchymal transition, angiogenesis, and the development of metastasis, Src is a promising target for cancer therapy. Several small molecule inhibitors of Src are currently being investigated in clinical trials. In this article, we will summarize the mechanisms regulating Src kinase activity in normal and cancer cells and discuss the status of Src inhibitor development against various types of cancers.
Collapse
|
68
|
Kaneko T, Sidhu SS, Li SSC. Evolving specificity from variability for protein interaction domains. Trends Biochem Sci 2011; 36:183-90. [PMID: 21227701 DOI: 10.1016/j.tibs.2010.12.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 12/07/2010] [Accepted: 12/08/2010] [Indexed: 12/18/2022]
Abstract
An important question in modular domain-peptide interactions, which play crucial roles in many biological processes, is how the diverse specificities exhibited by different members of a domain family are encoded in a common scaffold. Analysis of the Src homology (SH) 2 family has revealed that its specificity is determined, in large part, by the configuration of surface loops that regulate ligand access to binding pockets. In a distinct manner, SH3 domains employ loops for ligand recognition. The PDZ domain, in contrast, achieves specificity by co-evolution of binding-site residues. Thus, the conformational and sequence variability afforded by surface loops and binding sites provides a general mechanism by which to encode the wide spectrum of specificities observed for modular protein interaction domains.
Collapse
Affiliation(s)
- Tomonori Kaneko
- Department of Biochemistry and the Siebens-Drake Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | | | | |
Collapse
|
69
|
Amanchy R, Kandasamy K, Mathivanan S, Periaswamy B, Reddy R, Yoon WH, Joore J, Beer MA, Cope L, Pandey A. Identification of Novel Phosphorylation Motifs Through an Integrative Computational and Experimental Analysis of the Human Phosphoproteome. JOURNAL OF PROTEOMICS & BIOINFORMATICS 2011; 4:22-35. [PMID: 21720494 PMCID: PMC3124146 DOI: 10.4172/jpb.1000163] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein phosphorylation occurs in certain sequence/structural contexts that are still incompletely understood. The amino acids surrounding the phosphorylated residues are important in determining the binding of the kinase to the protein sequence. Upon phosphorylation these sequences also determine the binding of certain domains that specifically bind to phosphorylated sequences. Thus far, such 'motifs' have been identified through alignment of a limited number of well identified kinase substrates. RESULTS: Experimentally determined phosphorylation sites from Human Protein Reference Database were used to identify 1,167 novel serine/threonine or tyrosine phosphorylation motifs using a computational approach. We were able to statistically validate a number of these novel motifs based on their enrichment in known phosphopeptides datasets over phosphoserine/threonine/tyrosine peptides in the human proteome. There were 299 novel serine/threonine or tyrosine phosphorylation motifs that were found to be statistically significant. Several of the novel motifs that we identified computationally have subsequently appeared in large datasets of experimentally determined phosphorylation sites since we initiated our analysis. Using a peptide microarray platform, we have experimentally evaluated the ability of casein kinase I to phosphorylate a subset of the novel motifs discovered in this study. Our results demonstrate that it is feasible to identify novel phosphorylation motifs through large phosphorylation datasets. Our study also establishes peptide microarrays as a novel platform for high throughput kinase assays and for the validation of consensus motifs. Finally, this extended catalog of phosphorylation motifs should assist in a systematic study of phosphorylation networks in signal transduction pathways.
Collapse
Affiliation(s)
- Ramars Amanchy
- McKusick-Nathans Institute of Genetic Medicine and the Departments of Biological Chemistry and Oncology, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Kumaran Kandasamy
- McKusick-Nathans Institute of Genetic Medicine and the Departments of Biological Chemistry and Oncology, Johns Hopkins University, Baltimore, Maryland 21205, USA
- Institute of Bioinformatics, International Tech Park, Bangalore 560066, India
| | - Suresh Mathivanan
- Institute of Bioinformatics, International Tech Park, Bangalore 560066, India
| | | | - Raghunath Reddy
- Institute of Bioinformatics, International Tech Park, Bangalore 560066, India
| | - Wan-Hee Yoon
- McKusick-Nathans Institute of Genetic Medicine and the Departments of Biological Chemistry and Oncology, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Jos Joore
- Pepscan Systems, Edelhertweg 15, 8219 PH Lelystad, The Netherlands
| | - Michael A Beer
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Leslie Cope
- Sidney Kimmel Comprehensive Cancer Center and the Department of Biostatistics, Bloomberg School of Public Health, and Johns Hopkins University, Baltimore, Maryland, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine and the Departments of Biological Chemistry and Oncology, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
70
|
Groveman BR, Xue S, Marin V, Xu J, Ali MK, Bienkiewicz EA, Yu XM. Roles of the SH2 and SH3 domains in the regulation of neuronal Src kinase functions. FEBS J 2010; 278:643-53. [PMID: 21199370 DOI: 10.1111/j.1742-4658.2010.07985.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Previous studies demonstrated that intra-domain interactions between Src family kinases (SFKs), stabilized by binding of the phosphorylated C-terminus to the SH2 domain and/or binding of the SH2 kinase linker to the SH3 domain, lock the molecules in a closed conformation, disrupt the kinase active site, and inactivate SFKs. Here we report that the up-regulation of N-methyl-D-aspartate receptors (NMDARs) induced by expression of constitutively active neuronal Src (n-Src), in which the C-terminus tyrosine is mutated to phenylalanine (n-Src/Y535F), is significantly reduced by dysfunctions of the SH2 and/or SH3 domains of the protein. Furthermore, we found that dysfunctions of SH2 and/or SH3 domains reduce auto-phosphorylation of the kinase activation loop, depress kinase activity, and decrease NMDAR phosphorylation. The SH2 domain plays a greater regulatory role than the SH3 domain. Our data also show that n-Src binds directly to the C-terminus of the NMDAR NR2A subunit in vitro, with a K(D) of 108.2 ± 13.3 nM. This binding is not Src kinase activity-dependent, and dysfunctions of the SH2 and/or SH3 domains do not significantly affect the binding. These data indicate that the SH2 and SH3 domains may function to promote the catalytic activity of active n-Src, which is important in the regulation of NMDAR functions.
Collapse
Affiliation(s)
- Bradley R Groveman
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Kim EM, Kim J, Kim YG, Lee P, Shin DS, Kim M, Hahn JS, Lee YS, Kim BG. Development of high-throughput phosphorylation profiling method for identification of Ser/Thr kinase specificity. J Pept Sci 2010; 17:392-7. [DOI: 10.1002/psc.1312] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 09/07/2010] [Accepted: 09/14/2010] [Indexed: 01/21/2023]
|
72
|
Knowlton ML, Selfors LM, Wrobel CN, Gu TL, Ballif BA, Gygi SP, Polakiewicz R, Brugge JS. Profiling Y561-dependent and -independent substrates of CSF-1R in epithelial cells. PLoS One 2010; 5:e13587. [PMID: 21049007 PMCID: PMC2964295 DOI: 10.1371/journal.pone.0013587] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 08/08/2010] [Indexed: 01/06/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) activate multiple downstream cytosolic tyrosine kinases following ligand stimulation. SRC family kinases (SFKs), which are recruited to activated RTKs through SH2 domain interactions with RTK autophosphorylation sites, are targets of many subfamilies of RTKs. To date, there has not been a systematic analysis of the downstream substrates of such receptor-activated SFKs. Here, we conducted quantitative mass spectrometry utilizing stable isotope labeling (SILAC) analysis to profile candidate SRC-substrates induced by the CSF-1R tyrosine kinase by comparing the phosphotyrosine-containing peptides from cells expressing either CSF-1R or a mutant form of this RTK that is unable to bind to SFKs. This analysis identified previously uncharacterized changes in tyrosine phosphorylation induced by CSF-1R in mammary epithelial cells as well as a set of candidate substrates dependent on SRC recruitment to CSF-1R. Many of these candidates may be direct SRC targets as the amino acids flanking the phosphorylation sites in these proteins are similar to known SRC kinase phosphorylation motifs. The putative SRC-dependent proteins include known SRC substrates as well as previously unrecognized SRC targets. The collection of substrates includes proteins involved in multiple cellular processes including cell-cell adhesion, endocytosis, and signal transduction. Analyses of phosphoproteomic data from breast and lung cancer patient samples identified a subset of the SRC-dependent phosphorylation sites as being strongly correlated with SRC activation, which represent candidate markers of SRC activation downstream of receptor tyrosine kinases in human tumors. In summary, our data reveal quantitative site-specific changes in tyrosine phosphorylation induced by CSF-1R activation in epithelial cells and identify many candidate SRC-dependent substrates phosphorylated downstream of an RTK.
Collapse
Affiliation(s)
- Melodie L. Knowlton
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Laura M. Selfors
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carolyn N. Wrobel
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ting-Lei Gu
- Cell Signaling Technology, Inc., Danvers, Massachusetts, United States of America
| | - Bryan A. Ballif
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Roberto Polakiewicz
- Cell Signaling Technology, Inc., Danvers, Massachusetts, United States of America
| | - Joan S. Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
73
|
Takeda H, Kawamura Y, Miura A, Mori M, Wakamatsu A, Yamamoto JI, Isogai T, Matsumoto M, Nakayama KI, Natsume T, Nomura N, Goshima N. Comparative Analysis of Human Src-Family Kinase Substrate Specificity in Vitro. J Proteome Res 2010; 9:5982-93. [DOI: 10.1021/pr100773t] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hiroyuki Takeda
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Yoshifumi Kawamura
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Aya Miura
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Masatoshi Mori
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Ai Wakamatsu
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Jun-ichi Yamamoto
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Takao Isogai
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Masaki Matsumoto
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Keiichi I. Nakayama
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Tohru Natsume
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Nobuo Nomura
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| | - Naoki Goshima
- Japan Biological Informatics Consortium, TIME24 Building 10F 2-4-32 Aomi, Koto-ku, Tokyo 135-8073, Japan, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and National Institute of Advanced Industrial Science and Technology, 2-4-7 Aomi, Koto-ku, Tokyo 135
| |
Collapse
|
74
|
Feike AC, Rachor K, Gentzel M, Schambony A. Wnt5a/Ror2-induced upregulation of xPAPC requires xShcA. Biochem Biophys Res Commun 2010; 400:500-6. [DOI: 10.1016/j.bbrc.2010.08.074] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 08/17/2010] [Indexed: 10/19/2022]
|
75
|
Cheng Z, Tseng Y, White MF. Insulin signaling meets mitochondria in metabolism. Trends Endocrinol Metab 2010; 21:589-98. [PMID: 20638297 PMCID: PMC3994704 DOI: 10.1016/j.tem.2010.06.005] [Citation(s) in RCA: 331] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/04/2010] [Accepted: 06/04/2010] [Indexed: 12/11/2022]
Abstract
Insulin controls nutrient and metabolic homeostasis via the IRS-PI3K-AKT signaling cascade that targets FOXO1 and mTOR. Mitochondria, as the prime metabolic platform, malfunction during insulin resistance in metabolic diseases. However, the molecular link between insulin resistance and mitochondrial dysfunction remains undefined. Here we review recent studies on insulin action and the mechanistic association with mitochondrial metabolism. These studies suggest that insulin signaling underpins mitochondrial electron transport chain integrity and activity by suppressing FOXO1/HMOX1 and maintaining the NAD(+)/NADH ratio, the mediator of the SIRT1/PGC1α pathway for mitochondrial biogenesis and function. Mitochondria generate moderately reactive oxygen species (ROS) and enhance insulin sensitivity upon redox regulation of protein tyrosine phosphatase and insulin receptor. However, chronic exposure to high ROS levels could alter mitochondrial function and thereby cause insulin resistance.
Collapse
Affiliation(s)
- Zhiyong Cheng
- Howard Hughes Medical Institute, Division of Endocrinology, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
76
|
Wu HY, Tseng VS, Chen LC, Chang HY, Chuang IC, Tsay YG, Liao PC. Identification of Tyrosine-Phosphorylated Proteins Associated with Lung Cancer Metastasis using Label-Free Quantitative Analyses. J Proteome Res 2010; 9:4102-12. [DOI: 10.1021/pr1006153] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hsin-Yi Wu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| | - Vincent S. Tseng
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| | - Lien-Chin Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| | - Hui-Yin Chang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| | - I-Chi Chuang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| | - Yeou-Guang Tsay
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| | - Pao-Chi Liao
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan, Institute of Information Science, Academia Sinica, Taipei, Taiwan, Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, and Institute of Medical Informatics, National Cheng Kung
| |
Collapse
|
77
|
Kaneko T, Huang H, Zhao B, Li L, Liu H, Voss CK, Wu C, Schiller MR, Li SSC. Loops govern SH2 domain specificity by controlling access to binding pockets. Sci Signal 2010; 3:ra34. [PMID: 20442417 DOI: 10.1126/scisignal.2000796] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cellular functions require specific protein-protein interactions that are often mediated by modular domains that use binding pockets to engage particular sequence motifs in their partners. Yet, how different members of a domain family select for distinct sequence motifs is not fully understood. The human genome encodes 120 Src homology 2 (SH2) domains (in 110 proteins), which mediate protein-protein interactions by binding to proteins with diverse phosphotyrosine (pTyr)-containing sequences. The structure of the SH2 domain of BRDG1 bound to a peptide revealed a binding pocket that was blocked by a loop residue in most other SH2 domains. Analysis of 63 SH2 domain structures suggested that the SH2 domains contain three binding pockets, which exhibit selectivity for the three positions after the pTyr in a peptide, and that SH2 domain loops defined the accessibility and shape of these pockets. Despite sequence variability in the loops, we identified conserved structural features in the loops of SH2 domains responsible for controlling access to these surface pockets. We engineered new loops in an SH2 domain that altered specificity as predicted. Thus, selective blockage of binding subsites or pockets by surface loops provides a molecular basis by which the diverse modes of ligand recognition by the SH2 domain may have evolved and provides a framework for engineering SH2 domains and designing SH2-specific inhibitors.
Collapse
Affiliation(s)
- Tomonori Kaneko
- Department of Biochemistry and the Siebens-Drake Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Akiba H, Sumaoka J, Komiyama M. Binuclear Terbium(III) Complex as a Probe for Tyrosine Phosphorylation. Chemistry 2010; 16:5018-25. [DOI: 10.1002/chem.200903379] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
79
|
Serine dephosphorylation of receptor protein tyrosine phosphatase alpha in mitosis induces Src binding and activation. Mol Cell Biol 2010; 30:2850-61. [PMID: 20385765 DOI: 10.1128/mcb.01202-09] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Receptor protein tyrosine phosphatase alpha (RPTPalpha) is the mitotic activator of the protein tyrosine kinase Src. RPTPalpha serine hyperphosphorylation was proposed to mediate mitotic activation of Src. We raised phosphospecific antibodies to the two main serine phosphorylation sites, and we discovered that RPTPalpha Ser204 was almost completely dephosphorylated in mitotic NIH 3T3 and HeLa cells, whereas Ser180 and Tyr789 phosphorylation were only marginally reduced in mitosis. Concomitantly, Src pTyr527 and pTyr416 were dephosphorylated, resulting in 2.3-fold activation of Src in mitosis. Using inhibitors and knockdown experiments, we demonstrated that dephosphorylation of RPTPalpha pSer204 in mitosis was mediated by PP2A. Mutation of Ser204 to Ala did not activate RPTPalpha, and intrinsic catalytic activity of RPTPalpha was not affected in mitosis. Interestingly, binding of endogenous Src to RPTPalpha was induced in mitosis. GRB2 binding to RPTPalpha, which was proposed to compete with Src binding to RPTPalpha, was only modestly reduced in mitosis, which could not account for enhanced Src binding. Moreover, we demonstrate that Src bound to mutant RPTPalpha-Y789F, lacking the GRB2 binding site, and mutant Src with an impaired Src homology 2 (SH2) domain bound to RPTPalpha, illustrating that Src binding to RPTPalpha is not mediated by a pTyr-SH2 interaction. Mutation of RPTPalpha Ser204 to Asp, mimicking phosphorylation, reduced coimmunoprecipitation with Src, suggesting that phosphorylation of Ser204 prohibits binding to Src. Based on our results, we propose a new model for mitotic activation of Src in which PP2A-mediated dephosphorylation of RPTPalpha pSer204 facilitates Src binding, leading to RPTPalpha-mediated dephosphorylation of Src pTyr527 and pTyr416 and hence modest activation of Src.
Collapse
|
80
|
Abstract
The human proteome is known to contain >500 protein kinases, which regulate almost all facets of cellular biology by the post-translational attachment of a phosphate moiety to serine, threonine, or tyrosine residues within a substrate protein. Most protein kinases remain poorly characterized and, as a result, current studies are directed toward defining their target substrates experimentally to gain a comprehensive view of the signaling proteins and pathways modulated by these kinases. Herein, we describe a rapid and convenient method for elucidating the consensus substrate motif for phosphorylation by a protein kinase using peptide SPOT arrays that are custom-synthesized on a cellulose membrane support. The definition of the target consensus motif provides an important starting point for the identification of physiologically relevant kinase substrates.
Collapse
|
81
|
Akiba H, Sumaoka J, Komiyama M. Selective detection of phosphotyrosine in the presence of various phosphate-containing biomolecules with the aid of a terbium(III) complex. Chembiochem 2009; 10:1773-6. [PMID: 19565595 DOI: 10.1002/cbic.200900227] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Hiroki Akiba
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-Ku, Tokyo, 153-8904, Japan
| | | | | |
Collapse
|
82
|
Cross talk between receptor guanylyl cyclase C and c-src tyrosine kinase regulates colon cancer cell cytostasis. Mol Cell Biol 2009; 29:5277-89. [PMID: 19620276 DOI: 10.1128/mcb.00001-09] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Increased activation of c-src seen in colorectal cancer is an indicator of a poor clinical prognosis, suggesting that identification of downstream effectors of c-src may lead to new avenues of therapy. Guanylyl cyclase C (GC-C) is a receptor for the gastrointestinal hormones guanylin and uroguanylin and the bacterial heat-stable enterotoxin. Though activation of GC-C by its ligands elevates intracellular cyclic GMP (cGMP) levels and inhibits cell proliferation, its persistent expression in colorectal carcinomas and occult metastases makes it a marker for malignancy. We show here that GC-C is a substrate for inhibitory phosphorylation by c-src, resulting in reduced ligand-mediated cGMP production. Consequently, active c-src in colonic cells can overcome GC-C-mediated control of the cell cycle. Furthermore, docking of the c-src SH2 domain to phosphorylated GC-C results in colocalization and further activation of c-src. We therefore propose a novel feed-forward mechanism of activation of c-src that is induced by cross talk between a receptor GC and a tyrosine kinase. Our findings have important implications in understanding the molecular mechanisms involved in the progression and treatment of colorectal cancer.
Collapse
|
83
|
Tan CSH, Pasculescu A, Lim WA, Pawson T, Bader GD, Linding R. Positive selection of tyrosine loss in metazoan evolution. Science 2009; 325:1686-8. [PMID: 19589966 DOI: 10.1126/science.1174301] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
John Nash showed that within a complex system, individuals are best off if they make the best decision that they can, taking into account the decisions of the other individuals. Here, we investigate whether similar principles influence the evolution of signaling networks in multicellular animals. Specifically, by analyzing a set of metazoan species we observed a striking negative correlation of genomically encoded tyrosine content with biological complexity (as measured by the number of cell types in each organism). We discuss how this observed tyrosine loss correlates with the expansion of tyrosine kinases in the evolution of the metazoan lineage and how it may relate to the optimization of signaling systems in multicellular animals. We propose that this phenomenon illustrates genome-wide adaptive evolution to accommodate beneficial genetic perturbation.
Collapse
Affiliation(s)
- Chris Soon Heng Tan
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada
| | | | | | | | | | | |
Collapse
|
84
|
Protein tyrosine kinase characterization based on fully automated synthesis of (phospho) peptide arrays in microplates. Methods Mol Biol 2009. [PMID: 19241019 DOI: 10.1007/978-1-60327-834-8_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
In view of the importance of information transfer mediated throughout the cell by recognition, phosphorylation or dephosphorylation of kinases, their adapters, or substrates, this method was developed. The method provides a potent research tool for rapidly generating and testing these substrates as modeled by synthetic peptide arrays. The peptides or phosphorylated peptides are automatically generated on the inner surfaces of microplate wells, covalently linked to a polylysine polymer so that they are in a sterically favorable conformation, immediately available for in situ testing. Products up to 18 amino acids long have shown excellent mass spectral homogeneity. Thus, determinate peptide libraries can be ready for testing in as little as 2 days after the conception of an experiment. The process can be easily automated using robotic liquid handlers and is extremely rapid, sensitive, and economical. Optionally, the method can be upgraded to a higher throughput level using more powerful workstations with greater capacity, such as the Biomek FX, or any similar robotics capable of transfer-from-file logic to guide synthesis cycles.
Collapse
|
85
|
Okamura T, Singh S, Buolamwini J, Haystead T, Friedman H, Bigner D, Ali-Osman F. Tyrosine phosphorylation of the human glutathione S-transferase P1 by epidermal growth factor receptor. J Biol Chem 2009; 284:16979-16989. [PMID: 19254954 DOI: 10.1074/jbc.m808153200] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) gene amplification, mutations, and/or aberrant activation are frequent abnormalities in malignant gliomas and other human cancers and have been associated with an aggressive clinical course and a poor therapeutic outcome. Elevated glutathione S-transferase P1 (GSTP1), a major drug-metabolizing and stress response signaling protein, is also associated with drug resistance and poor clinical outcome in gliomas and other cancers. Here, we provide evidence that GSTP1 is a downstream EGFR target and that EGFR binds to and phosphorylates tyrosine residues in the GSTP1 protein in vitro and in vivo. Mass spectrometry and mutagenesis analyses in a cell-free system and in gliomas cells identified Tyr-7 and Tyr-198 as major EGFR-specific phospho-acceptor residues in the GSTP1 protein. The phosphorylation increased GSTP1 enzymatic activity significantly, and computer-based modeling showed a corresponding increase in electronegativity of the GSTP1 active site. In human glioma and breast cancer cells, epidermal growth factor stimulation rapidly increased GSTP1 tyrosine phosphorylation and decreased cisplatin sensitivity. Lapatinib, a clinically active EGFR inhibitor, significantly reversed the epidermal growth factor-induced cisplatin resistance. These data define phosphorylation and activation of GSTP1 by EGFR as a novel, heretofore unrecognized component of the EGFR signaling network and a novel mechanism of tumor drug resistance, particularly in tumors with elevated GSTP1 and/or activated EGFR.
Collapse
Affiliation(s)
| | - Simendra Singh
- From the Departments of Surgery, Durham, North Carolina 27710
| | - John Buolamwini
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163
| | | | - Henry Friedman
- From the Departments of Surgery, Durham, North Carolina 27710
| | - Darell Bigner
- Pathology, Duke Comprehensive Cancer Center, and the Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina 27710
| | | |
Collapse
|
86
|
Lew ED, Furdui CM, Anderson KS, Schlessinger J. The precise sequence of FGF receptor autophosphorylation is kinetically driven and is disrupted by oncogenic mutations. Sci Signal 2009; 2:ra6. [PMID: 19224897 DOI: 10.1126/scisignal.2000021] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Autophosphorylation of the tyrosine kinase domain of fibroblast growth factor receptor 1 (FGFR1) is mediated by a sequential and precisely ordered three-stage autophosphorylation reaction. First-stage autophosphorylation of an activation loop tyrosine leads to 50- to 100-fold stimulation of kinase activity and is followed by second-stage phosphorylation of three additional tyrosine residues, which are binding sites for signaling molecules. Finally, third-stage phosphorylation of a second activation loop tyrosine leads to an additional 10-fold stimulation of FGFR1 catalytic activity. In this report, we show that sequential autophosphorylation of five tyrosines in the FGFR1 kinase domain is under kinetic control, mediated by both the amino acid sequence surrounding the tyrosines and their locations within the kinase structure, and, moreover, that phosphoryl transfer is the rate-limiting step. Furthermore, the strict order of autophosphorylation is disrupted by a glioblastoma-derived, oncogenic FGFR1 point mutation in the kinase domain. We propose that disrupted stepwise activation of tyrosine autophosphorylation caused by oncogenic and other activating FGFR mutations may lead to aberrant activation of and assembly of signaling molecules by the activated receptor.
Collapse
Affiliation(s)
- Erin D Lew
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
87
|
Han X, Yamanouchi G, Mori T, Kang JH, Niidome T, Katayama Y. Monitoring protein kinase activity in cell lysates using a high-density peptide microarray. ACTA ACUST UNITED AC 2009; 14:256-62. [PMID: 19211777 DOI: 10.1177/1087057108329348] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Monitoring and targeting protein kinases is widely accepted as a promising approach for disease diagnosis and drug discovery. For this purpose, the authors have developed an original type of peptide array as a high-throughput screening assay for quantitatively evaluating kinase activity. A volume of 2 nL of peptide solution was spotted onto a formyl group-modified glass slide by using an arrayer, which was designed for use with protein chip technology. The phosphorylation was recognized by fluorescence-label antibody and detected with an automatic microarray scanner widely used in DNA chip technology. The system needs low sample volume, provides a high-density peptide array, and supplies high reproducibility. It provided enough sensitivity for inhibitor screening, even though a relatively low concentration of purified kinase was employed. The assay also proved useful for the detection of intracellular kinase activity as well as for the measurement of the fluctuations of intracellular protein kinase activity with drug stimulation. Thus, this peptide array would be applicable for kinase-targeted diagnosis, cell-based drug screening, and signal pathway investigation.
Collapse
Affiliation(s)
- Xiaoming Han
- Graduate School of System Life Sciences, Department of Applied Chemistry, Faculty of Engineering, Center of Future Chemistry, Kyushu University, Japan
| | | | | | | | | | | |
Collapse
|
88
|
Li SSC, Wu C. Using peptide array to identify binding motifs and interaction networks for modular domains. Methods Mol Biol 2009; 570:67-76. [PMID: 19649589 DOI: 10.1007/978-1-60327-394-7_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Specific protein-protein interactions underlie all essential biological processes and form the basis of cellular signal transduction. The recognition of a short, linear peptide sequence in one protein by a modular domain in another represents a common theme of macromolecular recognition in cells, and the importance of this mode of protein-protein interaction is highlighted by the large number of peptide-binding domains encoded by the human genome. This phenomenon also provides a unique opportunity to identify protein-protein binding events using peptide arrays and complementary biochemical assays. Accordingly, high-density peptide array has emerged as a useful tool by which to map domain-mediated protein-protein interaction networks at the proteome level. Using the Src-homology 2 (SH2) and 3 (SH3) domains as examples, we describe the application of oriented peptide array libraries in uncovering specific motifs recognized by an SH2 domain and the use of high-density peptide arrays in identifying interaction networks mediated by the SH3 domain. Methods reviewed here could also be applied to other modular domains, including catalytic domains, that recognize linear peptide sequences.
Collapse
Affiliation(s)
- Shawn S-C Li
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | |
Collapse
|
89
|
Ritz A, Shakhnarovich G, Salomon AR, Raphael BJ. Discovery of phosphorylation motif mixtures in phosphoproteomics data. ACTA ACUST UNITED AC 2008; 25:14-21. [PMID: 18996944 DOI: 10.1093/bioinformatics/btn569] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MOTIVATION Modification of proteins via phosphorylation is a primary mechanism for signal transduction in cells. Phosphorylation sites on proteins are determined in part through particular patterns, or motifs, present in the amino acid sequence. RESULTS We describe an algorithm that simultaneously discovers multiple motifs in a set of peptides that were phosphorylated by several different kinases. Such sets of peptides are routinely produced in proteomics experiments.Our motif-finding algorithm uses the principle of minimum description length to determine a mixture of sequence motifs that distinguish a foreground set of phosphopeptides from a background set of unphosphorylated peptides. We show that our algorithm outperforms existing motif-finding algorithms on synthetic datasets consisting of mixtures of known phosphorylation sites. We also derive a motif specificity score that quantifies whether or not the phosphoproteins containing an instance of a motif have a significant number of known interactions. Application of our motif-finding algorithm to recently published human and mouse proteomic studies recovers several known phosphorylation motifs and reveals a number of novel motifs that are enriched for interactions with a particular kinase or phosphatase. Our tools provide a new approach for uncovering the sequence specificities of uncharacterized kinases or phosphatases.
Collapse
Affiliation(s)
- Anna Ritz
- Department of Computer Science, Brown University, Toyota Technological Institute at Chicago, Chicago, IL, USA.
| | | | | | | |
Collapse
|
90
|
De Keersmaecker K, Versele M, Cools J, Superti-Furga G, Hantschel O. Intrinsic differences between the catalytic properties of the oncogenic NUP214-ABL1 and BCR-ABL1 fusion protein kinases. Leukemia 2008; 22:2208-16. [PMID: 18784740 DOI: 10.1038/leu.2008.242] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The NUP214-ABL1 fusion kinase has recently been identified in 6% of patients with T-cell acute lymphoblastic leukemia. In contrast to the more common oncogenic ABL1 fusion BCR-ABL1, NUP214-ABL1 localizes to the nuclear pore complexes and has attenuated transforming properties in hematopoietic cells and in mouse bone marrow transplant models. We have performed a thorough biochemical comparative analysis of NUP214-ABL1 and BCR-ABL1 and show that, despite their common tyrosine kinase domain, the two fusion proteins differ in many critical catalytic properties. NUP214-ABL1 has lower in vitro tyrosine kinase activity, which is in agreement with the absence of phosphorylation on its activation loop. NUP214-ABL1 was more sensitive to imatinib (Glivec) than BCR-ABL1 in vitro and in cells, indicating a different activation state and conformation of the two ABL1 fusion kinases. Using a peptide array, we identified differences in the spectrum and efficiency of substrate peptide phosphorylation and a differential involvement of Src kinases in downstream signaling. These results clearly indicate that different fusion partners of the same kinase can determine not only localization, but also critical functional properties of the enzyme such as inhibitor sensitivity and substrate preference, with subsequent differences in downstream signaling effectors and likely consequences in disease pathogenesis.
Collapse
Affiliation(s)
- K De Keersmaecker
- Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | | | | | | | | |
Collapse
|
91
|
Luković E, González-Vera JA, Imperiali B. Recognition-domain focused chemosensors: versatile and efficient reporters of protein kinase activity. J Am Chem Soc 2008; 130:12821-7. [PMID: 18759402 DOI: 10.1021/ja8046188] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Catalyzed by kinases, serine/threonine and tyrosine phosphorylation is a vital mechanism of intracellular regulation. Thus, assays that easily monitor kinase activity are critical in both academic and pharmaceutical settings. We previously developed sulfonamido-oxine (Sox)-based fluorescent peptides following a beta-turn focused (BTF) design for the continuous assay of kinase activity in vitro and in cell lysates. Upon phosphorylation of the Sox-containing peptide, the chromophore binds Mg (2+) and undergoes chelation-enhanced fluorescence (CHEF). Although the design was applied successfully to the development of several kinase sensors, an intrinsic limitation was that only residues C- or N-terminal to the phosphorylated residue could be used to derive specificity for the target kinase. To address this limitation, a new, recognition-domain focused (RDF) strategy was developed that also relies on CHEF. In this approach, the requirement for the constrained beta-turn motif is obviated by alkylation of a cysteine residue with a Sox-based derivative to afford an amino acid termed C-Sox. The RDF design allows inclusion of extended binding determinants to maximize recognition by the cognate kinase, which has now permitted the construction of chemosensors for a variety of representative Ser/Thr (PKC alpha, PKC betaIota, PKC delta, Pim2, Akt1, MK2, and PKA) as well as receptor (IRK) and nonreceptor (Src, Abl) Tyr kinases with greatly enhanced selectivity. The new sensors have up to 28-fold improved catalytic efficiency and up to 66-fold lower K M when compared to the corresponding BTF probes. The improved generality of the strategy is exemplified with the synthesis and analysis of Sox-based probes for PKC betaIota and PKC delta, which were previously unattainable using the BTF approach.
Collapse
Affiliation(s)
- Elvedin Luković
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
92
|
Harrison S, Das K, Karim F, Maclean D, Mendel D. Non-ATP-competitive kinase inhibitors – enhancing selectivity through new inhibition strategies. Expert Opin Drug Discov 2008; 3:761-74. [DOI: 10.1517/17460441.3.7.761] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
93
|
Gomes P, Saito T, Del Corsso C, Alioua A, Eghbali M, Toro L, Stefani E. Identification of a functional interaction between Kv4.3 channels and c-Src tyrosine kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1884-92. [PMID: 18620005 DOI: 10.1016/j.bbamcr.2008.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2008] [Revised: 06/02/2008] [Accepted: 06/02/2008] [Indexed: 12/31/2022]
Abstract
Voltage-gated K(+) (Kv) channels are key determinants of cardiac and neuronal excitability. A substantial body of evidence has accumulated in support of a role for Src family tyrosine kinases in the regulation of Kv channels. In this study, we examined the possibility that c-Src tyrosine kinase participates in the modulation of the transient voltage-dependent K(+) channel Kv4.3. Supporting a mechanistic link between Kv4.3 and c-Src, confocal microscopy analysis of HEK293 cells stably transfected with Kv4.3 showed high degree of co-localization of the two proteins at the plasma membrane. Our results further demonstrate an association between Kv4.3 and c-Src by co-immunoprecipitation and GST pull-down assays, this interaction being mediated by the SH2 and SH3 domains of c-Src. Furthermore, we show that Kv4.3 is tyrosine phosphorylated under basal conditions. The functional relevance of the observed interaction between Kv4.3 and c-Src was established in patch-clamp experiments, where application of the Src inhibitor PP2 caused a decrease in Kv4.3 peak current amplitude, but not the inactive structural analogue PP3. Conversely, intracellular application of recombinant c-Src kinase or the protein tyrosine phosphatase inhibitor bpV(phen) increased Kv4.3 peak current amplitude. In conclusion, our findings provide evidence that c-Src-induced Kv4.3 channel activation involves their association in a macromolecular complex and suggest a role for c-Src-Kv4.3 pathway in regulating cardiac and neuronal excitability.
Collapse
Affiliation(s)
- Pedro Gomes
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095-1778, USA.
| | | | | | | | | | | | | |
Collapse
|
94
|
de Souza AJ, Oak JS, Jordanhazy R, DeKruyff RH, Fruman DA, Kane LP. T cell Ig and mucin domain-1-mediated T cell activation requires recruitment and activation of phosphoinositide 3-kinase. THE JOURNAL OF IMMUNOLOGY 2008; 180:6518-26. [PMID: 18453570 DOI: 10.4049/jimmunol.180.10.6518] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Ligation of the transmembrane protein T cell Ig and mucin domain (Tim)-1 can costimulate T cell activation. Agonistic Abs to Tim-1 are also capable of inducing T cell activation without additional stimuli. However, little is known about the biochemical mechanisms underlying T cell stimulation or costimulation through Tim-1. We show that a tyrosine in Tim-1 becomes phosphorylated in a lck-dependent manner, whereupon it can directly recruit p85 adaptor subunits of PI3K. This results in PI3K activation, which is required for Tim-1 function. We also provide genetic evidence that p85 expression is required for optimal Tim-1 function. Thus, we describe a pathway from Tim-1 tyrosine phosphorylation to the PI3K signaling pathway, which appears to be a major effector of Tim-1-mediated T cell activation.
Collapse
Affiliation(s)
- Anjali J de Souza
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
95
|
Alberta JA, Segal RA. Generation and utilization of phosphorylation state-specific antibodies to investigate signaling pathways. ACTA ACUST UNITED AC 2008; Chapter 3:Unit 3.14. [PMID: 18428463 DOI: 10.1002/0471142301.ns0314s04] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Phosphorylation state-specific antibodies can be of great use, for example, in studying individual steps within a given signal transduction pathway. This unit presents a general approach to the generation and purification of phosphorylation state-specific antibodies. In addition to their ability to detect phosphorylation at a particular key site, these antibodies are often more sensitive for biochemical studies. Besides their application in immunoblotting procedures, activation state-specific antibodies can be used as immunohistochemical reagents. Thus, critical changes in phosphorylation can be monitored as described on an individual cell basis or in fixed tissue sections. Such antibodies can be used to address fundamental questions about signal transduction pathways during physiologic events that cannot be resolved by more conventional methodologies.
Collapse
Affiliation(s)
- J A Alberta
- Harvard Medical School and Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | |
Collapse
|
96
|
Physiological role for phosphatidic acid in the translocation of the novel protein kinase C Apl II in Aplysia neurons. Mol Cell Biol 2008; 28:4719-33. [PMID: 18505819 DOI: 10.1128/mcb.00178-08] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In Aplysia californica, the serotonin-mediated translocation of protein kinase C (PKC) Apl II to neuronal membranes is important for synaptic plasticity. The orthologue of PKC Apl II, PKCepsilon, has been reported to require phosphatidic acid (PA) in conjunction with diacylglycerol (DAG) for translocation. We find that PKC Apl II can be synergistically translocated to membranes by the combination of DAG and PA. We identify a mutation in the C1b domain (arginine 273 to histidine; PKC Apl II-R273H) that removes the effects of exogenous PA. In Aplysia neurons, the inhibition of endogenous PA production by 1-butanol inhibited the physiological translocation of PKC Apl II by serotonin in the cell body and at the synapse but not the translocation of PKC Apl II-R273H. The translocation of PKC Apl II-R273H in the absence of PA was explained by two additional effects of this mutation: (i) the mutation removed C2 domain-mediated inhibition, and (ii) the mutation decreased the concentration of DAG required for PKC Apl II translocation. We present a model in which, under physiological conditions, PA is important to activate the novel PKC Apl II both by synergizing with DAG and removing C2 domain-mediated inhibition.
Collapse
|
97
|
Li L, Wu C, Huang H, Zhang K, Gan J, Li SSC. Prediction of phosphotyrosine signaling networks using a scoring matrix-assisted ligand identification approach. Nucleic Acids Res 2008; 36:3263-73. [PMID: 18424801 PMCID: PMC2425477 DOI: 10.1093/nar/gkn161] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Systematic identification of binding partners for modular domains such as Src homology 2 (SH2) is important for understanding the biological function of the corresponding SH2 proteins. We have developed a worldwide web-accessible computer program dubbed SMALI for scoring matrix-assisted ligand identification for SH2 domains and other signaling modules. The current version of SMALI harbors 76 unique scoring matrices for SH2 domains derived from screening oriented peptide array libraries. These scoring matrices are used to search a protein database for short peptides preferred by an SH2 domain. An experimentally determined cut-off value is used to normalize an SMALI score, therefore allowing for direct comparison in peptide-binding potential for different SH2 domains. SMALI employs distinct scoring matrices from Scansite, a popular motif-scanning program. Moreover, SMALI contains built-in filters for phosphoproteins, Gene Ontology (GO) correlation and colocalization of subject and query proteins. Compared to Scansite, SMALI exhibited improved accuracy in identifying binding peptides for SH2 domains. Applying SMALI to a group of SH2 domains identified hundreds of interactions that overlap significantly with known networks mediated by the corresponding SH2 proteins, suggesting SMALI is a useful tool for facile identification of signaling networks mediated by modular domains that recognize short linear peptide motifs.
Collapse
Affiliation(s)
- Lei Li
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | | | | | |
Collapse
|
98
|
Warner N, Wybenga-Groot LE, Pawson T. Analysis of EphA4 receptor tyrosine kinase substrate specificity using peptide-based arrays. FEBS J 2008; 275:2561-73. [PMID: 18422655 DOI: 10.1111/j.1742-4658.2008.06405.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Eph receptor tyrosine kinases regulate many important biological processes. In the present study, we explored the substrate specificity of the EphA4 receptor tyrosine kinase using peptide arrays. We define a consensus substrate motif for EphA4 and go on to identify and test a number of potential EphA4 substrates and map their putative site(s) of phosphorylation. Cotransfection studies validate two of the predicted substrates: Nck2 and Dok1. Our findings identify several potential EphA4 substrates and demonstrate the general utility of using peptide arrays to rapidly identify and map protein kinase phosphorylation sites.
Collapse
Affiliation(s)
- Neil Warner
- Program in Systems Biology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | | | | |
Collapse
|
99
|
Kaushansky A, Gordus A, Chang B, Rush J, MacBeath G. A quantitative study of the recruitment potential of all intracellular tyrosine residues on EGFR, FGFR1 and IGF1R. MOLECULAR BIOSYSTEMS 2008; 4:643-53. [PMID: 18493663 DOI: 10.1039/b801018h] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Receptor tyrosine kinases transmit and process extracellular cues by recruiting intracellular signaling proteins to sites of tyrosine phosphorylation. Using protein microarrays comprising virtually every human SH2 and PTB domain, we generated quantitative protein interaction maps for three well-studied receptors--EGFR, FGFR1 and IGF1R--using phosphopeptides derived from every intracellular tyrosine residue on each receptor, regardless of whether or not they are phosphorylated in vivo. We found that, in general, peptides derived from physiological sites of tyrosine phosphorylation bind to substantially more SH2 or PTB domains than do peptides derived from nonphysiological sites, supporting the idea that kinases and interaction domains co-evolve and suggesting that new sites arise predominantly through selection favoring advantageous interactions, rather than through selection disfavoring unwanted interactions. We also found substantial qualitative overlap in the recruitment profiles of these three receptors, suggesting that their different biological effects arise, at least in part, from quantitative differences in their affinities for the proteins they recruit.
Collapse
Affiliation(s)
- Alexis Kaushansky
- Program in Molecular and Cellular Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | | | | | | | | |
Collapse
|
100
|
McMurray JS. Structural basis for the binding of high affinity phosphopeptides to Stat3. Biopolymers 2008; 90:69-79. [PMID: 18058821 DOI: 10.1002/bip.20901] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Signal transducer and activator of transcription 3 (Stat3) is constitutively active in a number of cancers where it participates in aberrant transcription of prosurvival, cell cycling, and angiogenesis genes. Since Stat3 initiates its signaling activity through binding of its SH2 domain to phosphotyrosine residues on cell surface receptors, inhibitors targeting this region of the protein are potential chemotherapeutic agents. To date, no NMR or X-ray crystallographic structures of high-affinity phosphopeptides complexed with the Stat3 SH2 domain are available to aid in the development of peptidomimetic antagonists. Examination of the crystal structures of several STAT proteins and the complex of Stat1 with Ac-pTyr-Asp-Lys-Pro-His-NH(2) led to a hypothesis that the specificity determinant for Stat3, glutamine at position pY+3 in pTyr-Xxx-Xxx-Gln sequences, resides in a unique pocket on the protein surface at the juncture of the third strand of the central beta-sheet and a unique, STAT specific alpha-helix. Docking of Ac-pTyr-Leu-Pro-Gln-NHBn to the SH2 domain of Stat3 using molecular modeling showed that the Gln binds tightly in this pocket and participates in a network of hydrogen bonds. Novel interactions between the peptide main chain and the protein were also discovered. Phosphopeptide structure-affinity studies using unnatural amino acids and glutamine derivatives provide evidence for the peptide-protein interactions revealed by the model and lend support to the binding hypothesis.
Collapse
Affiliation(s)
- John S McMurray
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|