Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Cell ; 186(11): 2361-2379.e25, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37192619

ABSTRACT

Multiple anticancer drugs have been proposed to cause cell death, in part, by increasing the steady-state levels of cellular reactive oxygen species (ROS). However, for most of these drugs, exactly how the resultant ROS function and are sensed is poorly understood. It remains unclear which proteins the ROS modify and their roles in drug sensitivity/resistance. To answer these questions, we examined 11 anticancer drugs with an integrated proteogenomic approach identifying not only many unique targets but also shared ones-including ribosomal components, suggesting common mechanisms by which drugs regulate translation. We focus on CHK1 that we find is a nuclear H2O2 sensor that launches a cellular program to dampen ROS. CHK1 phosphorylates the mitochondrial DNA-binding protein SSBP1 to prevent its mitochondrial localization, which in turn decreases nuclear H2O2. Our results reveal a druggable nucleus-to-mitochondria ROS-sensing pathway-required to resolve nuclear H2O2 accumulation and mediate resistance to platinum-based agents in ovarian cancers.


Subject(s)
Antineoplastic Agents , Reactive Oxygen Species , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Cell Nucleus/metabolism , Humans
2.
bioRxiv ; 2023 Mar 11.
Article in English | MEDLINE | ID: mdl-36945474

ABSTRACT

Multiple chemotherapies are proposed to cause cell death in part by increasing the steady-state levels of cellular reactive oxygen species (ROS). However, for most of these drugs exactly how the resultant ROS function and are sensed is poorly understood. In particular, it's unclear which proteins the ROS modify and their roles in chemotherapy sensitivity/resistance. To answer these questions, we examined 11 chemotherapies with an integrated proteogenomic approach identifying many unique targets for these drugs but also shared ones including ribosomal components, suggesting one mechanism by which chemotherapies regulate translation. We focus on CHK1 which we find is a nuclear H 2 O 2 sensor that promotes an anti-ROS cellular program. CHK1 acts by phosphorylating the mitochondrial-DNA binding protein SSBP1, preventing its mitochondrial localization, which in turn decreases nuclear H 2 O 2 . Our results reveal a druggable nucleus-to-mitochondria ROS sensing pathway required to resolve nuclear H 2 O 2 accumulation, which mediates resistance to platinum-based chemotherapies in ovarian cancers.

3.
Sci Signal ; 15(757): eabm0808, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36282911

ABSTRACT

Multiple coronaviruses have emerged independently in the past 20 years that cause lethal human diseases. Although vaccine development targeting these viruses has been accelerated substantially, there remain patients requiring treatment who cannot be vaccinated or who experience breakthrough infections. Understanding the common host factors necessary for the life cycles of coronaviruses may reveal conserved therapeutic targets. Here, we used the known substrate specificities of mammalian protein kinases to deconvolute the sequence of phosphorylation events mediated by three host protein kinase families (SRPK, GSK-3, and CK1) that coordinately phosphorylate a cluster of serine and threonine residues in the viral N protein, which is required for viral replication. We also showed that loss or inhibition of SRPK1/2, which we propose initiates the N protein phosphorylation cascade, compromised the viral replication cycle. Because these phosphorylation sites are highly conserved across coronaviruses, inhibitors of these protein kinases not only may have therapeutic potential against COVID-19 but also may be broadly useful against coronavirus-mediated diseases.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Humans , SARS-CoV-2/genetics , Phosphorylation , Glycogen Synthase Kinase 3/metabolism , Virus Replication , Nucleocapsid Proteins/metabolism , Nucleocapsid/metabolism , Serine/metabolism , Threonine/metabolism , Mammals/metabolism , Protein Serine-Threonine Kinases
4.
Nat Methods ; 19(11): 1371-1375, 2022 11.
Article in English | MEDLINE | ID: mdl-36280721

ABSTRACT

Mass-spectrometry-based phosphoproteomics has become indispensable for understanding cellular signaling in complex biological systems. Despite the central role of protein phosphorylation, the field still lacks inexpensive, regenerable, and diverse phosphopeptides with ground-truth phosphorylation positions. Here, we present Iterative Synthetically Phosphorylated Isomers (iSPI), a proteome-scale library of human-derived phosphoserine-containing phosphopeptides that is inexpensive, regenerable, and diverse, with precisely known positions of phosphorylation. We demonstrate possible uses of iSPI, including use as a phosphopeptide standard, a tool to evaluate and optimize phosphorylation-site localization algorithms, and a benchmark to compare performance across data analysis pipelines. We also present AScorePro, an updated version of the AScore algorithm specifically optimized for phosphorylation-site localization in higher energy fragmentation spectra, and the FLR viewer, a web tool for phosphorylation-site localization, to enable community use of the iSPI resource. iSPI and its associated data constitute a useful, multi-purpose resource for the phosphoproteomics community.


Subject(s)
Phosphopeptides , Proteome , Humans , Proteome/metabolism , Phosphopeptides/metabolism , Phosphoserine/metabolism , Proteomics , Mass Spectrometry , Phosphorylation
5.
bioRxiv ; 2020 Dec 16.
Article in English | MEDLINE | ID: mdl-32817937

ABSTRACT

While vaccines are vital for preventing COVID-19 infections, it is critical to develop new therapies to treat patients who become infected. Pharmacological targeting of a host factor required for viral replication can suppress viral spread with a low probability of viral mutation leading to resistance. In particular, host kinases are highly druggable targets and a number of conserved coronavirus proteins, notably the nucleoprotein (N), require phosphorylation for full functionality. In order to understand how targeting kinases could be used to compromise viral replication, we used a combination of phosphoproteomics and bioinformatics as well as genetic and pharmacological kinase inhibition to define the enzymes important for SARS-CoV-2 N protein phosphorylation and viral replication. From these data, we propose a model whereby SRPK1/2 initiates phosphorylation of the N protein, which primes for further phosphorylation by GSK-3a/b and CK1 to achieve extensive phosphorylation of the N protein SR-rich domain. Importantly, we were able to leverage our data to identify an FDA-approved kinase inhibitor, Alectinib, that suppresses N phosphorylation by SRPK1/2 and limits SARS-CoV-2 replication. Together, these data suggest that repurposing or developing novel host-kinase directed therapies may be an efficacious strategy to prevent or treat COVID-19 and other coronavirus-mediated diseases.

7.
Nat Med ; 26(1): 131-142, 2020 01.
Article in English | MEDLINE | ID: mdl-31932797

ABSTRACT

Glia have been implicated in Alzheimer's disease (AD) pathogenesis. Variants of the microglia receptor triggering receptor expressed on myeloid cells 2 (TREM2) increase AD risk, and activation of disease-associated microglia (DAM) is dependent on TREM2 in mouse models of AD. We surveyed gene-expression changes associated with AD pathology and TREM2 in 5XFAD mice and in human AD by single-nucleus RNA sequencing. We confirmed the presence of Trem2-dependent DAM and identified a previously undiscovered Serpina3n+C4b+ reactive oligodendrocyte population in mice. Interestingly, remarkably different glial phenotypes were evident in human AD. Microglia signature was reminiscent of IRF8-driven reactive microglia in peripheral-nerve injury. Oligodendrocyte signatures suggested impaired axonal myelination and metabolic adaptation to neuronal degeneration. Astrocyte profiles indicated weakened metabolic coordination with neurons. Notably, the reactive phenotype of microglia was less evident in TREM2-R47H and TREM2-R62H carriers than in non-carriers, demonstrating a TREM2 requirement in both mouse and human AD, despite the marked species-specific differences.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Transcriptome/genetics , Aged , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/pathology , Brain/metabolism , Brain/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Middle Aged , Nerve Degeneration/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Transcription, Genetic
8.
J Proteome Res ; 16(4): 1506-1514, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28171727

ABSTRACT

A comprehensive view of protein phosphorylation remains an unmet challenge in the field of cell biology. Mass spectrometry-based proteomics is one of the most promising approaches for identifying thousands of phosphorylation events in a single experiment, yet the full breadth of the phosphoproteome has yet to be elucidated. In this article, we examined the complementarity of two methods for phosphopeptide enrichment based on either titanium dioxide (TiO2) enrichment or phosphorylation motif-specific immunoaffinity precipitation (IAP) with four different antibodies. Each method identified nearly 2000 phosphoproteins. However, distinct populations of phosphopeptides were observed. Despite quantifying over 10 000 unique phosphorylation events using TiO2 and over 3900 with IAP, less than 5% of the sites were in common. Agreeing with published literature, the ratio of pS:pT:pY phosphorylation for the TiO2-enriched data set approximated 90:10:<1. In contrast, that ratio for the combined IAP data sets was 51:29:20. These differences not only suggest the complementarity between multiple enrichment methods but also emphasize their collective importance in obtaining a comprehensive view of the phosphoproteome.


Subject(s)
Phosphopeptides/biosynthesis , Phosphoproteins/biosynthesis , Proteomics , Cell Line, Tumor , Humans , Mass Spectrometry , Phosphopeptides/genetics , Phosphoproteins/genetics , Phosphorylation/genetics , Titanium/chemistry
9.
J Proteome Res ; 16(2): 1069-1076, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27978624

ABSTRACT

While developing a multiplexed phosphotyrosine peptide quantification assay, an unexpected observation was made: significant neutral loss from phosphotyrosine (pY) containing peptides. Using a 2000-member peptide library, we sought to systematically investigate this observation by comparing unlabeled peptides with the two highest-plex isobaric tags (iTRAQ8 and TMT10) across CID, HCD, and ETD fragmentation using high resolution high mass accuracy Orbitrap instrumentation. We found pY peptide neutral loss behavior was consistent with reduced proton mobility, and does not occur during ETD. The site of protonation at the peptide N-terminus changes from a primary to a tertiary amine as a result of TMT labeling which would increase the gas phase basicity and reduce proton mobility at this site. This change in fragmentation behavior has implications during instrument method development and interpretation of MS/MS spectra, and therefore ensuing follow-up studies. We show how sites not localized to tyrosine by search and site localization algorithms can be confidently reassigned to tyrosine using neutral loss and phosphotyrosine immonium ions. We believe these findings will be of general interest to those studying pY signal transduction using isobaric tags.


Subject(s)
Peptides/chemistry , Phosphotyrosine/chemistry , Spectrometry, Mass, Electrospray Ionization/standards , Peptides/analysis , Phosphotyrosine/analysis , Reagent Kits, Diagnostic , Staining and Labeling/methods , Tyrosine/chemistry
10.
Cell Syst ; 3(4): 395-403.e4, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27667366

ABSTRACT

System-wide quantitative analysis of ubiquitylomes has proven to be a valuable tool for elucidating targets and mechanisms of the ubiquitin-driven signaling systems, as well as gaining insights into neurodegenerative diseases and cancer. Current mass spectrometry methods for ubiquitylome detection require large amounts of starting material and rely on stochastic data collection to increase replicate analyses. We describe a method compatible with cell line and tissue samples for large-scale quantification of 5,000-9,000 ubiquitylation forms across ten samples simultaneously. Using this method, we reveal site-specific ubiquitylation in mammalian brain and liver tissues, as well as in cancer cells undergoing proteasome inhibition. To demonstrate the power of the approach for signal-dependent ubiquitylation, we examined protein and ubiquitylation dynamics for mitochondria undergoing PARKIN- and PINK1-dependent mitophagy. This analysis revealed the largest collection of PARKIN- and PINK1-dependent ubiquitylation targets to date in a single experiment, and it also revealed a subset of proteins recruited to the mitochondria during mitophagy.


Subject(s)
Ubiquitination , Animals , Autophagy , Mass Spectrometry , Mitochondria , Mitophagy , Parkinson Disease , Protein Kinases , Ubiquitin , Ubiquitin-Protein Ligases
11.
Proteomics ; 15(2-3): 462-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25195567

ABSTRACT

Multiplexed isobaric tag based quantitative proteomics and phosphoproteomics strategies can comprehensively analyze drug treatments effects on biological systems. Given the role of mitogen-activated protein/extracellular signal-regulated kinase (MEK) signaling in cancer and mitogen-activated protein kinase (MAPK)-dependent diseases, we sought to determine if this pathway could be inhibited safely by examining the downstream molecular consequences. We used a series of tandem mass tag 10-plex experiments to analyze the effect of two MEK inhibitors (GSK1120212 and PD0325901) on three tissues (kidney, liver, and pancreas) from nine mice. We quantified ∼ 6000 proteins in each tissue, but significant protein-level alterations were minimal with inhibitor treatment. Of particular interest was kidney tissue, as edema is an adverse effect of these inhibitors. From kidney tissue, we enriched phosphopeptides using titanium dioxide (TiO2 ) and quantified 10 562 phosphorylation events. Further analysis by phosphotyrosine peptide immunoprecipitation quantified an additional 592 phosphorylation events. Phosphorylation motif analysis revealed that the inhibitors decreased phosphorylation levels of proline-x-serine-proline (PxSP) and serine-proline (SP) sites, consistent with extracellular-signal-regulated kinase (ERK) inhibition. The MEK inhibitors had the greatest decrease on the phosphorylation of two proteins, Barttin and Slc12a3, which have roles in ion transport and fluid balance. Further studies will provide insight into the effect of these MEK inhibitors with respect to edema and other adverse events in mouse models and human patients.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphopeptides/analysis , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , Amino Acid Sequence , Animals , Diphenylamine/pharmacology , Humans , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/chemistry , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Pancreas/drug effects , Pancreas/metabolism , Phosphopeptides/metabolism , Phosphorylation/drug effects , Proteome/chemistry , Proteomics , Tandem Mass Spectrometry
12.
Cancer Cell ; 27(1): 97-108, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25544637

ABSTRACT

BYL719, which selectively inhibits the alpha isoform of the phosphatidylinositol 3-kinase (PI3K) catalytic subunit (p110a), is currently in clinical trials for the treatment of solid tumors, especially luminal breast cancers with PIK3CA mutations and/or HER2 amplification. This study reveals that, even among these sensitive cancers, the initial efficacy of p110α inhibition is mitigated by rapid re-accumulation of the PI3K product PIP3 produced by the p110ß isoform. Importantly, the reactivation of PI3K mediated by p110ß does not invariably restore AKT phosphorylation, demonstrating the limitations of using phospho-AKT as a surrogate to measure PI3K activation. Consistently, we show that the addition of the p110ß inhibitor to BYL719 prevents the PIP3 rebound and induces greater antitumor efficacy in HER2-amplified and PIK3CA mutant cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Class I Phosphatidylinositol 3-Kinases/metabolism , Pyrimidinones/pharmacology , Thiazoles/pharmacology , ortho-Aminobenzoates/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Transplantation , Receptor, ErbB-2/genetics , Signal Transduction/drug effects
13.
Mol Cell ; 56(3): 360-375, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25284222

ABSTRACT

Phosphorylation is often used to promote protein ubiquitylation, yet we rarely understand quantitatively how ligase activation and ubiquitin (UB) chain assembly are integrated with phosphoregulation. Here we employ quantitative proteomics and live-cell imaging to dissect individual steps in the PINK1 kinase-PARKIN UB ligase mitochondrial control pathway disrupted in Parkinson's disease. PINK1 plays a dual role by phosphorylating PARKIN on its UB-like domain and poly-UB chains on mitochondria. PARKIN activation by PINK1 produces canonical and noncanonical UB chains on mitochondria, and PARKIN-dependent chain assembly is required for accumulation of poly-phospho-UB (poly-p-UB) on mitochondria. In vitro, PINK1 directly activates PARKIN's ability to assemble canonical and noncanonical UB chains and promotes association of PARKIN with both p-UB and poly-p-UB. Our data reveal a feedforward mechanism that explains how PINK1 phosphorylation of both PARKIN and poly-UB chains synthesized by PARKIN drives a program of PARKIN recruitment and mitochondrial ubiquitylation in response to mitochondrial damage.


Subject(s)
Mitochondria/enzymology , Polyubiquitin/biosynthesis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Feedback, Physiological , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Mutation, Missense , Parkinson Disease/enzymology , Phosphorylation , Protein Kinases/metabolism , Protein Multimerization , Protein Transport , Proteomics , Ubiquitin-Protein Ligases/genetics
14.
Mol Cell Proteomics ; 13(1): 372-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24129315

ABSTRACT

Protein methylation is a common posttranslational modification that mostly occurs on arginine and lysine residues. Arginine methylation has been reported to regulate RNA processing, gene transcription, DNA damage repair, protein translocation, and signal transduction. Lysine methylation is best known to regulate histone function and is involved in epigenetic regulation of gene transcription. To better study protein methylation, we have developed highly specific antibodies against monomethyl arginine; asymmetric dimethyl arginine; and monomethyl, dimethyl, and trimethyl lysine motifs. These antibodies were used to perform immunoaffinity purification of methyl peptides followed by LC-MS/MS analysis to identify and quantify arginine and lysine methylation sites in several model studies. Overall, we identified over 1000 arginine methylation sites in human cell line and mouse tissues, and ∼160 lysine methylation sites in human cell line HCT116. The number of methylation sites identified in this study exceeds those found in the literature to date. Detailed analysis of arginine-methylated proteins observed in mouse brain compared with those found in mouse embryo shows a tissue-specific distribution of arginine methylation, and extends the types of proteins that are known to be arginine methylated to include many new protein types. Many arginine-methylated proteins that we identified from the brain, including receptors, ion channels, transporters, and vesicle proteins, are involved in synaptic transmission, whereas the most abundant methylated proteins identified from mouse embryo are transcriptional regulators and RNA processing proteins.


Subject(s)
Arginine/metabolism , Brain/metabolism , Lysine/metabolism , Protein Processing, Post-Translational , Amino Acid Motifs/genetics , Animals , Arginine/genetics , Chromatography, Liquid , HCT116 Cells , Humans , Lysine/genetics , Methylation , Mice , Tandem Mass Spectrometry
15.
Clin Cancer Res ; 19(13): 3577-90, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23633453

ABSTRACT

PURPOSE: Ovarian cancer has the highest mortality rate of all female reproductive malignancies. Drug resistance is a major cause of treatment failure and novel therapeutic strategies are urgently needed. MLN4924 is a NEDDylation inhibitor currently under investigation in multiple phase I studies. We investigated its anticancer activity in cisplatin-sensitive and -resistant ovarian cancer models. EXPERIMENTAL DESIGN: Cellular sensitivity to MLN4924/cisplatin was determined by measuring viability, clonogenic survival, and apoptosis. The effects of drug treatment on global protein expression, DNA damage, and reactive oxygen species generation were determined. RNA interference established natural born killer/bcl-2-interacting killer (NBK/BIK) as a regulator of therapeutic sensitivity. The in vivo effects of MLN4924/cisplatin on tumor burden and key pharmacodynamics were assessed in cisplatin-sensitive and -resistant xenograft models. RESULTS: MLN4924 possessed significant activity against both cisplatin-sensitive and -resistant ovarian cancer cells and provoked the stabilization of key NEDD8 substrates and regulators of cellular redox status. Notably, MLN4924 significantly augmented the activity of cisplatin against cisplatin-resistant cells, suggesting that aberrant NEDDylation may contribute to drug resistance. MLN4924 and cisplatin cooperated to induce DNA damage, oxidative stress, and increased expression of the BH3-only protein NBK/BIK. Targeted NBK/BIK knockdown diminished the proapoptotic effects of the MLN4924/cisplatin combination. Administration of MLN4924 to mice bearing ovarian tumor xenografts significantly increased the efficacy of cisplatin against both cisplatin-sensitive and -resistant tumors. CONCLUSIONS: Our collective data provide a rationale for the clinical investigation of NEDD8-activating enzyme (NAE) inhibition as a novel strategy to augment cisplatin efficacy in patients with ovarian cancer and other malignancies.


Subject(s)
Cisplatin/pharmacology , Cyclopentanes/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Survival/drug effects , DNA Damage/genetics , Female , Humans , Membrane Proteins/metabolism , Mice , Mitochondrial Proteins , NF-kappa B/antagonists & inhibitors , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proteome , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
17.
Nat Biotechnol ; 30(5): 447-52, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446692

ABSTRACT

We describe a proteomics approach that identifies antigen-specific antibody sequences directly from circulating polyclonal antibodies in the serum of an immunized animal. The approach involves affinity purification of antibodies with high specific activity and then analyzing digested antibody fractions by nano-flow liquid chromatography coupled to tandem mass spectrometry. High-confidence peptide spectral matches of antibody variable regions are obtained by searching a reference database created by next-generation DNA sequencing of the B-cell immunoglobulin repertoire of the immunized animal. Finally, heavy and light chain sequences are paired and expressed as recombinant monoclonal antibodies. Using this technology, we isolated monoclonal antibodies for five antigens from the sera of immunized rabbits and mice. The antigen-specific activities of the monoclonal antibodies recapitulate or surpass those of the original affinity-purified polyclonal antibodies. This technology may aid the discovery and development of vaccines and antibody therapeutics, and help us gain a deeper understanding of the humoral response.


Subject(s)
Antibodies, Monoclonal/chemistry , Blood Proteins/chemistry , Proteomics/methods , Animals , Antibodies/chemistry , Antibody Specificity/genetics , B-Lymphocytes/cytology , Chromatography, Liquid/methods , Immunoglobulin G/chemistry , Immunoglobulin Variable Region/chemistry , Mice , Rabbits , Receptors, Progesterone/chemistry , Serum/immunology , Tandem Mass Spectrometry/methods
18.
Cell ; 143(7): 1174-89, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21183079

ABSTRACT

Although most tissues in an organism are genetically identical, the biochemistry of each is optimized to fulfill its unique physiological roles, with important consequences for human health and disease. Each tissue's unique physiology requires tightly regulated gene and protein expression coordinated by specialized, phosphorylation-dependent intracellular signaling. To better understand the role of phosphorylation in maintenance of physiological differences among tissues, we performed proteomic and phosphoproteomic characterizations of nine mouse tissues. We identified 12,039 proteins, including 6296 phosphoproteins harboring nearly 36,000 phosphorylation sites. Comparing protein abundances and phosphorylation levels revealed specialized, interconnected phosphorylation networks within each tissue while suggesting that many proteins are regulated by phosphorylation independently of their expression. Our data suggest that the "typical" phosphoprotein is widely expressed yet displays variable, often tissue-specific phosphorylation that tunes protein activity to the specific needs of each tissue. We offer this dataset as an online resource for the biological research community.


Subject(s)
Gene Expression Profiling , Mice/genetics , Organ Specificity , Phosphorylation , Proteins/metabolism , Animals , Mice/metabolism , Protein Kinases/genetics , Proteomics
19.
J Proteome Res ; 9(6): 3103-7, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20377248

ABSTRACT

Intramolecular transfer of phosphate during collision-induced dissociation (CID) in ion-trap mass spectrometers has recently been described. Because phosphorylation events are assigned to discrete serine, threonine, and tyrosine residues based on the presence of site-determining ions in MS/MS spectra, phosphate transfer may invalidate or confound site localization in published large-scale phosphorylation data sets. Here, we present evidence for the occurrence of this phenomenon using synthetic phosphopeptide libraries, specifically for doubly charged species. We found, however, that the extent of the transfer reaction was insufficient to cause localization of phosphorylation sites to incorrect residues. We further compared CID to electron-transfer dissociation (ETD) for site localization using synthetic libraries and a large-scale yeast phosphoproteome experiment. The agreement in site localization was >99.5 and 93%, respectively, suggesting that ETD-based site localization is no more reliable than CID. We conclude that intramolecular phosphate transfer does not affect the reliability of current or past phosphorylation data sets.


Subject(s)
Phosphoproteins/chemistry , Proteome/chemistry , Proteomics/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Gases/chemistry , Molecular Sequence Data , Phosphorylation , Saccharomyces cerevisiae Proteins/chemistry
20.
Proteomics ; 8(21): 4444-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18972524

ABSTRACT

Phosphopeptide identification and site determination are major challenges in biomedical MS. Both are affected by frequent and often overwhelming losses of phosphoric acid in ion trap CID fragmentation spectra. These losses are thought to translate into reduced intensities of sequence informative ions and a general decline in the quality of MS/MS spectra. To address this issue, several methods have been proposed, which rely on extended fragmentation schemes including collecting MS3 scans from neutral loss-containing ions and multi-stage activation to further fragment these same ions. Here, we have evaluated the utility of these methods in the context of a large-scale phosphopeptide analysis strategy with current instrumentation capable of accurate precursor mass determination. Remarkably, we found that MS3-based schemes did not increase the overall number of confidently identified peptides and had only limited value in site localization. We conclude that the collection of MS3 or pseudo-MS3 scans in large-scale proteomics studies is not worthwhile when high-mass accuracy instrumentation is used.


Subject(s)
Phosphoproteins/analysis , Saccharomyces cerevisiae Proteins/analysis , Mass Spectrometry/methods , Phosphopeptides/analysis , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...