Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(23)2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36499482

ABSTRACT

A reduction in FADD levels has been reported in precursor T-cell neoplasms and other tumor types. Such reduction would impact on the ability of tumor cells to undergo apoptosis and has been associated with poor clinical outcomes. However, FADD is also known to participate in non-apoptotic functions, but these mechanisms are not well-understood. Linking FADD expression to the severity of precursor T-cell neoplasms could indicate its use as a prognostic marker and may open new avenues for targeted therapeutic strategies. Using transcriptomic and clinical data from patients with precursor T-cell neoplasms, complemented by in vitro analysis of cellular functions and by high-throughput interactomics, our results allow us to propose a dual role for FADD in precursor T-cell neoplasms, whereby resisting cell death and chemotherapy would be a canonical consequence of FADD deficiency in these tumors, whereas deregulation of the cellular metabolism would be a relevant non-canonical function in patients expressing FADD. These results reveal that evaluation of FADD expression in precursor T-cell neoplasms may aid in the understanding of the biological processes that are affected in the tumor cells. The altered biological processes can be of different natures depending on the availability of FADD influencing its ability to exert its canonical or non-canonical functions. Accordingly, specific therapeutic interventions would be needed in each case.


Subject(s)
Apoptosis , Neoplasms , Humans , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Apoptosis/genetics , Gene Expression Profiling , Cell Death , T-Lymphocytes/metabolism
2.
J Proteome Res ; 21(4): 930-939, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35235327

ABSTRACT

Protein post-translational modifications (PTMs) enable cells to rapidly change in response to biological stimuli. With hundreds of different PTMs, understanding these control mechanisms is complex. To date, efforts have focused on investigating the effect of a single PTM on protein function. Yet, many proteins contain multiple PTMs. Moreover, one PTM can alter the prevalence of another, a phenomenon termed PTM crosstalk. Understanding PTM crosstalk is critical; however, its detection is challenging since PTMs occur substoichiometrically. Here, we develop an enrichment-free, label-free proteomics method that utilizes high-field asymmetric ion mobility spectrometry (FAIMS) to enhance the detection of PTM crosstalk. We show that by searching for multiple combinations of dynamic PTMs on peptide sequences, a 6-fold increase in candidate PTM crosstalk sites is identified compared with that of standard liquid chromatography-tandem mass spectrometry (LC-MS/MS) workflows. Additionally, by cycling through FAIMS compensation voltages within a single LC-FAIMS-MS/MS run, we show that our LC-FAIMS-MS/MS workflow can increase multi-PTM-containing peptide identifications without additional increases in run times. With 159 novel candidate crosstalk sites identified, we envisage LC-FAIMS-MS/MS to play an important role in expanding the repertoire of multi-PTM identifications. Moreover, it is only by detecting PTM crosstalk that we can "see" the full picture of how proteins are regulated.


Subject(s)
Ion Mobility Spectrometry , Tandem Mass Spectrometry , Chromatography, Liquid/methods , Ion Mobility Spectrometry/methods , Protein Processing, Post-Translational , Proteomics/methods , Tandem Mass Spectrometry/methods
3.
Stem Cell Res ; 50: 102133, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33383406

ABSTRACT

Human embryonic stem cells (hESCs) are an invaluable tool in the fields of embryology and regenerative medicine. Activin A and BMP4 are well-characterised growth factors implicated in pluripotency and differentiation. In the current study, hESCs are cultured in a modified version of mTeSR1, where low concentrations of ActivinA substitute for TGFß. This culture system is further used to investigate the changes induced by BMP4 on hESCs by employing a combination of transcriptomic and phosphoproteomic approaches. Results indicate that in a pluripotent state, hESCs maintain WNT signaling under negative regulation by expressing pathway inhibitors. Initial stages of differentiation are characterized by upregulation of WNT pathway ligands, TGFß pathway inhibitors which have been shown in Xenopus to expand the BMP signaling range essential for embryonic patterning, and mesendodermal transcripts. Moreover, BMP4 enhances the phosphorylation of proteins associated with migration and transcriptional regulation. Results further indicate the vital regulatory role of Activin A and BMP4 in crucial fate decisions in hESCs.

4.
Sci Rep ; 10(1): 7950, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409632

ABSTRACT

Fibroblast Growth Factor (FGF) dependent signalling is frequently activated in cancer by a variety of different mechanisms. However, the downstream signal transduction pathways involved are poorly characterised. Here a quantitative differential phosphoproteomics approach, SILAC, is applied to identify FGF-regulated phosphorylation events in two triple- negative breast tumour cell lines, MFM223 and SUM52, that exhibit amplified expression of FGF receptor 2 (FGFR2) and are dependent on continued FGFR2 signalling for cell viability. Comparative Gene Ontology proteome analysis revealed that SUM52 cells were enriched in proteins associated with cell metabolism and MFM223 cells enriched in proteins associated with cell adhesion and migration. FGFR2 inhibition by SU5402 impacts a significant fraction of the observed phosphoproteome of these cells. This study expands the known landscape of FGF signalling and identifies many new targets for functional investigation. FGF signalling pathways are found to be flexible in architecture as both shared, and divergent, responses to inhibition of FGFR2 kinase activity in the canonical RAF/MAPK/ERK/RSK and PI3K/AKT/PDK/mTOR/S6K pathways are identified. Inhibition of phosphorylation-dependent negative-feedback pathways is observed, defining mechanisms of intrinsic resistance to FGFR2 inhibition. These findings have implications for the therapeutic application of FGFR inhibitors as they identify both common and divergent responses in cells harbouring the same genetic lesion and pathways of drug resistance.


Subject(s)
Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Gene Ontology , Humans , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
5.
Sci Adv ; 3(8): e1700898, 2017 08.
Article in English | MEDLINE | ID: mdl-28835924

ABSTRACT

Vomocytosis, or nonlytic extrusion, is a poorly understood process through which macrophages release live pathogens that they have failed to kill back into the extracellular environment. Vomocytosis is conserved across vertebrates and occurs with a diverse range of pathogens, but to date, the host signaling events that underpin expulsion remain entirely unknown. We use a targeted inhibitor screen to identify the MAP kinase ERK5 as a critical suppressor of vomocytosis. Pharmacological inhibition or genetic manipulation of ERK5 activity significantly raises vomocytosis rates in human macrophages, whereas stimulation of the ERK5 signaling pathway inhibits vomocytosis. Lastly, using a zebrafish model of cryptococcal disease, we show that reducing ERK5 activity in vivo stimulates vomocytosis and results in reduced dissemination of infection. ERK5 therefore represents the first host signaling regulator of vomocytosis to be identified and a potential target for the future development of vomocytosis-modulating therapies.


Subject(s)
Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Line , Cytokines/metabolism , Humans , Macrophages/drug effects , Mice , Protein Kinase Inhibitors/pharmacology , Zebrafish
6.
Sci Rep ; 7(1): 3970, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28638064

ABSTRACT

The Platelet Derived Growth Factor (PDGF) family of ligands have well established functions in the induction of cell proliferation and migration during development, tissue homeostasis and interactions between tumours and stroma. However, the mechanisms by which these actions are executed are incompletely understood. Here we report a differential phosphoproteomics study, using a SILAC approach, of PDGF-stimulated mouse embryonic fibroblasts (MEFs). 116 phospho-sites were identified as up-regulated and 45 down-regulated in response to PDGF stimulation. These encompass proteins involved in cell adhesion, cytoskeleton regulation and vesicle-mediated transport, significantly expanding the range of proteins implicated in PDGF signalling pathways. Included in the down-regulated class was the microtubule bundling protein Collapsin Response Mediator Protein 2 (CRMP2). In response to stimulation with PDGF, CRMP2 was dephosphorylated on Thr514, an event known to increase CRMP2 activity. This was reversed in the presence of micromolar concentrations of the protein phosphatase inhibitor okadaic acid, implicating PDGF-induced activation of protein phosphatase 1 (PP1) in CRMP2 regulation. Depletion of CRMP2 resulted in impairment of PDGF-mediated cell migration in an in vitro wound healing assay. These results show that CRMP2 is required for PDGF-directed cell migration in vitro.


Subject(s)
Cell Movement , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation , Mice , Phosphorylation , Platelet-Derived Growth Factor/administration & dosage , Proteomics , Signal Transduction
7.
J Cell Sci ; 129(15): 2962-71, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27352860

ABSTRACT

Focal adhesions are complex multi-molecular structures that link the actin cytoskeleton to the extracellular matrix through integrin adhesion receptors and play a key role in regulation of many cellular functions. LAR (also known as PTPRF) is a receptor protein tyrosine phosphatase that regulates PDGF signalling and localises to focal adhesions. We have observed that loss of LAR phosphatase activity in mouse embryonic fibroblasts results in reduced numbers of focal adhesions and decreased adhesion to fibronectin. To understand how LAR regulates cell adhesion we used phosphoproteomic data, comparing global phosphorylation events in wild-type and LAR phosphatase-deficient cells, to analyse differential kinase activity. Kinase prediction analysis of LAR-regulated phosphosites identified a node of cytoskeleton- and adhesion-related proteins centred on cyclin-dependent kinase-1 (CDK1). We found that loss of LAR activity resulted in reduced activity of CDK1, and that CDK1 activity was required for LAR-mediated focal adhesion complex formation. We also established that LAR regulates CDK1 activity through c-Abl and Akt family proteins. In summary, we have identified a new role for a receptor protein tyrosine phosphatase in regulating CDK1 activity and hence cell adhesion to the extracellular matrix.


Subject(s)
CDC2 Protein Kinase/metabolism , Focal Adhesions/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Cell Adhesion/drug effects , Fibronectins/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Mice , Models, Biological , Phosphorylation/drug effects , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
8.
Mol Cell Proteomics ; 15(6): 1823-36, 2016 06.
Article in English | MEDLINE | ID: mdl-27074791

ABSTRACT

Intracellular signaling pathways are reliant on protein phosphorylation events that are controlled by a balance of kinase and phosphatase activity. Although kinases have been extensively studied, the role of phosphatases in controlling specific cell signaling pathways has been less so. Leukocyte common antigen-related protein (LAR) is a member of the LAR subfamily of receptor-like protein tyrosine phosphatases (RPTPs). LAR is known to regulate the activity of a number of receptor tyrosine kinases, including platelet-derived growth factor receptor (PDGFR). To gain insight into the signaling pathways regulated by LAR, including those that are PDGF-dependent, we have carried out the first systematic analysis of LAR-regulated signal transduction using SILAC-based quantitative proteomic and phosphoproteomic techniques. We haveanalyzed differential phosphorylation between wild-type mouse embryo fibroblasts (MEFs) and MEFs in which the LAR cytoplasmic phosphatase domains had been deleted (LARΔP), and found a significant change in abundance of phosphorylation on 270 phosphosites from 205 proteins because of the absence of the phosphatase domains of LAR. Further investigation of specific LAR-dependent phosphorylation sites and enriched biological processes reveal that LAR phosphatase activity impacts on a variety of cellular processes, most notably regulation of the actin cytoskeleton. Analysis of putative upstream kinases that may play an intermediary role between LAR and the identified LAR-dependent phosphorylation events has revealed a role for LAR in regulating mTOR and JNK signaling.


Subject(s)
MAP Kinase Signaling System , Platelet-Derived Growth Factor/metabolism , Proteomics/methods , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Isotope Labeling , Mass Spectrometry , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Sequence Deletion , Signal Transduction
9.
J Proteome Res ; 14(12): 5077-87, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26503514

ABSTRACT

We have applied liquid chromatography high-field asymmetric waveform ion mobility spectrometry tandem mass spectrometry (LC-FAIMS-MS/MS) and liquid chromatography tandem mass spectrometry (LC-MS/MS) to the investigation of site-specific phosphorylation in fibroblast growth factor (FGF) signaling. We have combined a SILAC approach with chemical inhibition by SU5402 (an FGF receptor tyrosine kinase inhibitor) and dasatinib (a Src family kinase inhibitor). The results show that incorporation of FAIMS within the workflow results in (a) an increase in the relative proportion of phosphothreonine and phosphotyrosine sites identified, (b) an increase in phosphopeptide identifications from precursors with charge states ≥ +3 (with an associated increase in peptide length), and (c) an increase in the identification of multiply phosphorylated peptides. Approximately 20% of the phosphorylation sites identified via the FAIMS workflow had not been reported previously, and over 80% of those were from multiply phosphorylated peptides. Moreover, FAIMS provided access to a distinct set of phosphorylation sites regulated in response to SU5402 and dasatinib. The enhanced identification of multiply phosphorylated peptides was particularly striking in the case of sites regulated by SU5402. In addition to providing a compelling example of the complementarity of FAIMS in phosphoproteomics, the results provide a valuable resource of phosphorylation sites for further investigation of FGF signaling and trafficking.


Subject(s)
Chromatography, Liquid/methods , Fibroblast Growth Factors/metabolism , Mass Spectrometry/methods , Phosphopeptides/analysis , Phosphopeptides/metabolism , Amino Acid Sequence , Dasatinib/pharmacology , Humans , Molecular Sequence Data , Phosphorylation , Phosphotyrosine/analysis , Phosphotyrosine/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Pyrroles/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Tandem Mass Spectrometry/methods , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
10.
J Cell Sci ; 127(Pt 24): 5303-16, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25359883

ABSTRACT

Eps8 is an actin regulatory scaffold protein whose expression is increased in squamous cell carcinoma (SCC) cells. It forms a complex with both focal adhesion kinase (FAK, also known as PTK2) and Src in SCC cells derived from skin carcinomas induced by administration of the chemical DMBA followed by TPA (the DMBA/TPA model). Here, we describe two new roles for Eps8. Firstly, it controls the spatial distribution of active Src in a FAK-dependent manner. Specifically, Eps8 participates in, and regulates, a biochemical complex with Src and drives trafficking of Src to autophagic structures that SCC cells use to cope with high levels of active Src when FAK is absent. Secondly, when FAK is expressed in SCC cells, thereby meaning active Src becomes tethered at focal adhesion complexes, Eps8 is also recruited to focal adhesions and is required for FAK-dependent polarization and invasion. Therefore, Eps8 is a crucial mediator of Src- and FAK-regulated processes; it participates in specific biochemical complexes and promotes actin re-arrangements that determine the spatial localization of Src, and modulates the functions of Src and FAK during invasive migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , 3T3 Cells , Actins/metabolism , Amino Acid Sequence , Animals , Autophagy , Cell Line, Tumor , Cell Movement , Cell Polarity , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesions/metabolism , Gene Knockdown Techniques , Humans , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Peptides/chemistry , Peptides/metabolism , Phagosomes/metabolism , Phenotype , Protein Binding , Protein Transport , Up-Regulation
11.
J Cell Sci ; 127(Pt 5): 994-1006, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24413169

ABSTRACT

Growth factor signalling regulates multiple cellular functions and its misregulation has been linked to the development and progression of cancer. Ack1 (activated Cdc42-associated kinase 1, also known as TNK2) is a non-receptor tyrosine kinase that has been implicated in trafficking and degradation of epidermal growth factor receptor (EGFR), yet its precise functions remain elusive. In this report, we investigate the role of Ack1 in EGFR trafficking and show that Ack1 partially colocalises to Atg16L-positive structures upon stimulation with EGF. These structures are proposed to be the isolation membranes that arise during formation of autophagosomes. In addition, we find that Ack1 colocalises and interacts with sequestosome 1 (p62/SQSTM1), a receptor for selective autophagy, through a ubiquitin-associated domain, and this interaction decreases upon treatment with EGF, thus suggesting that Ack1 moves away from p62/SQSTM1 compartments. Furthermore, Ack1 interacts and colocalises with NBR1, another autophagic receptor, and this colocalisation is enhanced in the presence of ectopically expressed p62/SQSTM1. Finally, knockdown of Ack1 results in accelerated localisation of EGFR to lysosomes upon treatment with EGF. Structure-function analyses of a panel of Ack1 deletion mutants revealed key mechanistic aspects of these relationships. The Mig6-homology domain and clathrin-binding domain both contribute to colocalisation with EGFR, whereas the UBA domain is essential for colocalisation with p62/SQSTM1, but not NBR1. Taken together, our studies demonstrate a novel role for Ack1 in diverting activated EGFR into a non-canonical degradative pathway, marked by association with p62/SQSTM1, NBR1 and Atg16L.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , ErbB Receptors/metabolism , Protein-Tyrosine Kinases/physiology , Proteins/metabolism , Autophagy , Epidermal Growth Factor/physiology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Phagosomes , Protein Interaction Domains and Motifs , Protein Transport , Receptors, Fibroblast Growth Factor/metabolism , Sequestosome-1 Protein , Tumor Suppressor Proteins/metabolism , Ubiquitin/metabolism
12.
PLoS One ; 8(4): e61513, 2013.
Article in English | MEDLINE | ID: mdl-23626693

ABSTRACT

Eps8 is involved in both cell signalling and receptor trafficking. It is a known phosphorylation substrate for two proteins involved in the fibroblast growth factor receptor (FGFR) signalling pathway: the receptor itself and Src. Here we report a differential proteomic analysis of Eps8 aimed to identify specific FGFR and Src family kinase dependent phosphosites and co-associated phosphodependent binding partners. This study reveals a total of 22 Eps8 pTyr and pSer/Thr phosphorylation sites, including those that are dependent on Src family and FGFR kinase activity. Peptide affinity purification of proteins that bind to a selection of the pTyr phosphosites has identified a range of novel Eps8 binding partners including members of the intracellular vesicle trafficking machinery (clathrin and AP-2), proteins which have been shown to regulate activated receptor trafficking (NBR1 and Vav2), and proteins involved in receptor signalling (IRS4 and Shp2). Collectively this study significantly extends the understanding of Eps8 post-translational modification by regulated phosphorylation, identifies novel Eps8 binding partners implicated in receptor trafficking and signalling, and confirms the functions of Eps8 at the nexus of receptor signalling and vesicular trafficking.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Protein Interaction Maps , Protein Processing, Post-Translational , Receptors, Fibroblast Growth Factor/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , Cytoskeletal Proteins , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Oligopeptides/analysis , Phosphoproteins/genetics , Phosphorylation , Phosphotyrosine/chemistry , Phosphotyrosine/metabolism , Protein Binding , Protein Interaction Mapping , Protein Transport , Proteomics , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction , src-Family Kinases/genetics
13.
J Cell Sci ; 126(Pt 2): 613-24, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23203811

ABSTRACT

Fibroblast growth factor receptors (FGFRs) mediate a wide spectrum of cellular responses that are crucial for development and wound healing. However, aberrant FGFR activity leads to cancer. Activated growth factor receptors undergo stimulated endocytosis, but can continue to signal along the endocytic pathway. Endocytic trafficking controls the duration and intensity of signalling, and growth factor receptor signalling can lead to modifications of trafficking pathways. We have developed live-cell imaging methods for studying FGFR dynamics to investigate mechanisms that coordinate the interplay between receptor trafficking and signal transduction. Activated FGFR enters the cell following recruitment to pre-formed clathrin-coated pits (CCPs). However, FGFR activation stimulates clathrin-mediated endocytosis; FGF treatment increases the number of CCPs, including those undergoing endocytosis, and this effect is mediated by Src and its phosphorylation target Eps8. Eps8 interacts with the clathrin-mediated endocytosis machinery and depletion of Eps8 inhibits FGFR trafficking and immediate Erk signalling. Once internalized, FGFR passes through peripheral early endosomes en route to recycling and degredative compartments, through an Src- and Eps8-dependent mechanism. Thus Eps8 functions as a key coordinator in the interplay between FGFR signalling and trafficking. This work provides the first detailed mechanistic analysis of growth factor receptor clustering at the cell surface through signal transduction and endocytic trafficking. As we have characterised the Src target Eps8 as a key regulator of FGFR signalling and trafficking, and identified the early endocytic system as the site of Eps8-mediated effects, this work provides novel mechanistic insight into the reciprocal regulation of growth factor receptor signalling and trafficking.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Receptors, Fibroblast Growth Factor/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Growth Processes/physiology , Clathrin/metabolism , Dynamins/metabolism , Endocytosis/physiology , Endosomes/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Phosphorylation , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction , Transfection , src-Family Kinases/genetics
14.
J Proteome Res ; 9(5): 2317-28, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20225815

ABSTRACT

Activation of signal transduction by the receptor tyrosine kinase, fibroblast growth factor receptor (FGFR), results in a cascade of protein-protein interactions that rely on the occurrence of specific tyrosine phosphorylation events. One such protein recruited to the activated receptor complex is the nonreceptor tyrosine kinase, Src, which is involved in both initiation and termination of further signaling events. To gain a further understanding of the tyrosine phosphorylation events that occur during FGF signaling, with a specific focus on those that are dependent on Src family kinase (SFK) activity, we have applied SILAC combined with chemical inhibition of SFK activity to search for phosphorylation events that are dependent on SFK activity in FGF stimulated cells. In addition, we used a more targeted approach to carry out high coverage phosphopeptide mapping of one Src substrate protein, the multifunctional adaptor Dok1, and to identify SFK-dependent Dok1 binding partners. From these analyses we identify 80 SFK-dependent phosphorylation events on 40 proteins. We further identify 18 SFK-dependent Dok1 interactions and 9 SFK-dependent Dok1 phosphorylation sites, 6 of which had not previously been known to be SFK-dependent.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Phosphoproteins/metabolism , Proteome/metabolism , Proteomics/methods , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 2/chemistry , Humans , Immunoprecipitation , Isotope Labeling , Mice , Molecular Sequence Data , NIH 3T3 Cells , Peptides/chemistry , Peptides/metabolism , Phosphoproteins/chemistry , Phosphorylation , Protein Binding , Proteome/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Signal Transduction , src-Family Kinases/chemistry
15.
J Proteome Res ; 8(12): 5475-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19821632

ABSTRACT

Large data sets of electron capture dissociation (ECD) mass spectra from proteomic experiments are rich in information; however, extracting that information in an optimal manner is not straightforward. Protein database search engines currently available are designed for low resolution CID data, from which Fourier transform ion cyclotron resonance (FT-ICR) ECD data differs significantly. ECD mass spectra contain both z-prime and z-dot fragment ions (and c-prime and c-dot); ECD mass spectra contain abundant peaks derived from neutral losses from charge-reduced precursor ions; FT-ICR ECD spectra are acquired with a larger precursor m/z isolation window than their low-resolution CID counterparts. Here, we consider three distinct stages of postacquisition analysis: (1) processing of ECD mass spectra prior to the database search; (2) the database search step itself and (3) postsearch processing of results. We demonstrate that each of these steps has an effect on the number of peptides identified, with the postsearch processing of results having the largest effect. We compare two commonly used search engines: Mascot and OMSSA. Using an ECD data set of modest size (3341 mass spectra) from a complex sample (mouse whole cell lysate), we demonstrate that search results can be improved from 630 identifications (19% identification success rate) to 1643 identifications (49% identification success rate). We focus in particular on improving identification rates for doubly charged precursors, which are typically low for ECD fragmentation. We compare our presearch processing algorithm with a similar algorithm recently developed for electron transfer dissociation (ETD) data.


Subject(s)
Databases, Protein , Mass Spectrometry/methods , Proteomics/methods , Animals , Cells/chemistry , Mass Spectrometry/instrumentation , Mass Spectrometry/standards , Mice , Search Engine/standards
16.
J Proteome Res ; 8(4): 1965-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19275241

ABSTRACT

Recently, software has become available to automate localization of phosphorylation sites from CID data and to assign associated confidence scores. We present an algorithm, SLoMo (Site Localization of Modifications), which extends this capability to ETD/ECD mass spectra. Furthermore, SLoMo caters for both high and low resolution data and allows for site-localization of any UniMod post-translational modification. SLoMo accepts input data from a variety of formats (e.g., Sequest, OMSSA). We validate SLoMo with high and low resolution ETD, ECD, and CID data.


Subject(s)
Algorithms , Protein Processing, Post-Translational/physiology , Proteome/metabolism , Sequence Analysis, Protein
17.
Mol Cell Proteomics ; 8(5): 904-12, 2009 May.
Article in English | MEDLINE | ID: mdl-19131326

ABSTRACT

We used on-line electron capture dissociation (ECD) for the large scale identification and localization of sites of phosphorylation. Each FT-ICR ECD event was paired with a linear ion trap collision-induced dissociation (CID) event, allowing a direct comparison of the relative merits of ECD and CID for phosphopeptide identification and site localization. Linear ion trap CID was shown to be most efficient for phosphopeptide identification, whereas FT-ICR ECD was superior for localization of sites of phosphorylation. The combination of confident CID and ECD identification and confident CID and ECD localization is particularly valuable in cases where a phosphopeptide is identified just once within a phosphoproteomics experiment.


Subject(s)
Mass Spectrometry/methods , Proteins/metabolism , Amino Acid Sequence , Animals , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphopeptides/analysis , Phosphopeptides/chemistry , Phosphorylation , Proteins/chemistry , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...