High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform

Mass spectrometry has transformed the field of cell signaling by enabling global studies of dynamic protein phosphorylation (‘phosphoproteomics’). Recent developments are enabling increasingly sophisticated phosphoproteomics studies, but practical challenges remain. The EasyPhos workflow addresses these and is sufficiently streamlined to enable the analysis of hundreds of phosphoproteomes at a depth of >10,000 quantified phosphorylation sites. Here we present a detailed and updated workflow that further ensures high performance in sample-limited conditions while also reducing sample preparation time. By eliminating protein precipitation steps and performing the entire protocol, including digestion, in a single 96-well plate, we now greatly minimize opportunities for sample loss and variability. This results in very high reproducibility and a small sample size requirement (≤200 μg of protein starting material). After cell culture or tissue collection, the protocol takes 1 d, whereas mass spectrometry measurements require ~1 h per sample. Applied to glioblastoma cells acutely treated with epidermal growth factor (EGF), EasyPhos quantified 20,132 distinct phosphopeptides from 200 μg of protein in less than 1 d of measurement time, revealing thousands of EGF-regulated phosphorylation events.This protocol describes the EasyPhos platform, a high-throughput and user-friendly workflow for reliable identification of phosphopeptides from small amounts (<200 μg) of sample.

[1]  S. Lemieux,et al.  Phosphoproteome dynamics reveal novel ERK1/2 MAP kinase substrates with broad spectrum of functions , 2013, Molecular systems biology.

[2]  Sean J. Humphrey,et al.  Protein Phosphorylation: A Major Switch Mechanism for Metabolic Regulation , 2015, Trends in Endocrinology & Metabolism.

[3]  M. Mann,et al.  Global and site-specific quantitative phosphoproteomics: principles and applications. , 2009, Annual review of pharmacology and toxicology.

[4]  Masaru Tomita,et al.  Microscale phosphoproteome analysis of 10,000 cells from human cancer cell lines. , 2011, Analytical chemistry.

[5]  J. Shabanowitz,et al.  Phosphoproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae , 2002, Nature Biotechnology.

[6]  M. Larsen,et al.  Highly selective enrichment of phosphorylated peptides using titanium dioxide , 2006, Nature Protocols.

[7]  David E. James,et al.  Dynamic Adipocyte Phosphoproteome Reveals that Akt Directly Regulates mTORC2 , 2013, Cell metabolism.

[8]  Sean J. Humphrey,et al.  Phosphorylation Is a Central Mechanism for Circadian Control of Metabolism and Physiology. , 2017, Cell metabolism.

[9]  Liang-Yu Shih,et al.  An improved trypsin digestion method minimizes digestion-induced modifications on proteins. , 2009, Analytical biochemistry.

[10]  A. Heck,et al.  Single-step Enrichment by Ti4+-IMAC and Label-free Quantitation Enables In-depth Monitoring of Phosphorylation Dynamics with High Reproducibility and Temporal Resolution * , 2014, Molecular & Cellular Proteomics.

[11]  M. Mann,et al.  Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips , 2007, Nature Protocols.

[12]  Matthias Mann,et al.  Phosphoproteomics reveals that Parkinson's disease kinase LRRK2 regulates a subset of Rab GTPases , 2016, eLife.

[13]  G A Anderson,et al.  An ion funnel interface for improved ion focusing and sensitivity using electrospray ionization mass spectrometry. , 1998, Analytical chemistry.

[14]  M. Mann,et al.  MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification , 2008, Nature Biotechnology.

[15]  M. Tomita,et al.  Phosphopeptide Enrichment by Aliphatic Hydroxy Acid-modified Metal Oxide Chromatography for Nano-LC-MS/MS in Proteomics Applications*S , 2007, Molecular & Cellular Proteomics.

[16]  Frederick E. Petry,et al.  Principles and Applications , 1997 .

[17]  P. Roepstorff,et al.  Highly Selective Enrichment of Phosphorylated Peptides from Peptide Mixtures Using Titanium Dioxide Microcolumns* , 2005, Molecular & Cellular Proteomics.

[18]  J. Olsen,et al.  Large-Scale Phosphoproteomics Reveals Shp-2 Phosphatase-Dependent Regulators of Pdgf Receptor Signaling. , 2018, Cell reports.

[19]  Martin R Larsen,et al.  TiO(2)-based phosphoproteomic analysis of the plasma membrane and the effects of phosphatase inhibitor treatment. , 2008, Journal of proteome research.

[20]  Sean J Humphrey,et al.  High-throughput phosphoproteomics reveals in vivo insulin signaling dynamics , 2015, Nature Biotechnology.

[21]  M. Mann,et al.  Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells , 2014, Nature Methods.

[22]  B. Blagoev,et al.  Quantitative phosphoproteomics to characterize signaling networks. , 2012, Seminars in cell & developmental biology.

[23]  Masaru Tomita,et al.  Phase transfer surfactant-aided trypsin digestion for membrane proteome analysis. , 2008, Journal of proteome research.

[24]  R. Doolittle,et al.  A simple method for displaying the hydropathic character of a protein. , 1982, Journal of molecular biology.

[25]  Jesper V Olsen,et al.  Performance Evaluation of the Q Exactive HF-X for Shotgun Proteomics. , 2018, Journal of proteome research.

[26]  M. Mann,et al.  Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. , 2014, Cell reports.

[27]  S. Mohammed,et al.  Robust phosphoproteome enrichment using monodisperse microsphere–based immobilized titanium (IV) ion affinity chromatography , 2013, Nature Protocols.

[28]  Matthias Mann,et al.  The Q Exactive HF, a Benchtop Mass Spectrometer with a Pre-filter, High-performance Quadrupole and an Ultra-high-field Orbitrap Analyzer* , 2014, Molecular & Cellular Proteomics.

[29]  J. Porath,et al.  Isolation of phosphoproteins by immobilized metal (Fe3+) affinity chromatography. , 1986, Analytical biochemistry.

[30]  Beisi Xu,et al.  Retinal Cell Type DNA Methylation and Histone Modifications Predict Reprogramming Efficiency and Retinogenesis in 3D Organoid Cultures , 2018, Cell reports.

[31]  Marcus Svensson,et al.  Heat stabilization of the tissue proteome: a new technology for improved proteomics. , 2009, Journal of proteome research.

[32]  Juan Antonio Vizcaíno,et al.  The ProteomeXchange consortium in 2017: supporting the cultural change in proteomics public data deposition , 2016, Nucleic Acids Res..

[33]  J. Olsen,et al.  Off-line high-pH reversed-phase fractionation for in-depth phosphoproteomics. , 2014, Journal of proteome research.

[34]  Shang-Yu Huang,et al.  Nano-titanium dioxide composites for the enrichment of phosphopeptides. , 2006, Journal of chromatography. A.

[35]  Andrew R. Jones,et al.  ProteomeXchange provides globally co-ordinated proteomics data submission and dissemination , 2014, Nature Biotechnology.

[36]  Matthias Mann,et al.  In vivo brain GPCR signaling elucidated by phosphoproteomics , 2018, Science.

[37]  Joost W Gouw,et al.  Highly robust, automated, and sensitive online TiO2-based phosphoproteomics applied to study endogenous phosphorylation in Drosophila melanogaster. , 2008, Journal of proteome research.

[38]  Sean J. Humphrey,et al.  Glucose-regulated and drug-perturbed phosphoproteome reveals molecular mechanisms controlling insulin secretion , 2016, Nature Communications.

[39]  J. Coon,et al.  Phosphoproteomics in the Age of Rapid and Deep Proteome Profiling , 2015, Analytical chemistry.

[40]  K. Lage,et al.  Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues , 2012, Nature Communications.

[41]  M. Mann,et al.  Global, In Vivo, and Site-Specific Phosphorylation Dynamics in Signaling Networks , 2006, Cell.

[42]  S. Lemeer,et al.  The phosphoproteomics data explosion. , 2009, Current opinion in chemical biology.

[43]  Y. Ishihama,et al.  Specificity of immobilized metal affinity-based IMAC/C18 tip enrichment of phosphopeptides for protein phosphorylation analysis. , 2005, Analytical chemistry.

[44]  S. Michnick,et al.  A cell-signaling network temporally resolves specific versus promiscuous phosphorylation. , 2015, Cell reports.

[45]  Jesper V Olsen,et al.  Rapid and deep proteomes by faster sequencing on a benchtop quadrupole ultra-high-field Orbitrap mass spectrometer. , 2014, Journal of proteome research.

[46]  M. Mann,et al.  Quantitative phosphoproteome analysis of a mouse liver cell line reveals specificity of phosphatase inhibitors , 2008, Proteomics.

[47]  Ruedi Aebersold,et al.  Mass spectrometry‐driven phosphoproteomics: patterning the systems biology mosaic , 2014, Wiley interdisciplinary reviews. Developmental biology.

[48]  M. Mann,et al.  Status of Large-scale Analysis of Post-translational Modifications by Mass Spectrometry* , 2013, Molecular & Cellular Proteomics.

[49]  Jüergen Cox,et al.  The MaxQuant computational platform for mass spectrometry-based shotgun proteomics , 2016, Nature Protocols.

[50]  Steven P Gygi,et al.  The SCX/IMAC enrichment approach for global phosphorylation analysis by mass spectrometry , 2008, Nature Protocols.

[51]  Ronald J. Moore,et al.  Reversed‐phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells , 2011, Proteomics.

[52]  Marco Y. Hein,et al.  The Perseus computational platform for comprehensive analysis of (prote)omics data , 2016, Nature Methods.

[53]  M. Posewitz,et al.  Immobilized gallium(III) affinity chromatography of phosphopeptides. , 1999, Analytical chemistry.