Abstract
The signal transduction pathways initiated by lymphocyte activation play a critical role in regulating host immunity. High-resolution mass spectrometry has accelerated the investigation of these complex and dynamic pathways by enabling the qualitative and quantitative investigation of thousands of proteins and phosphoproteins simultaneously. In addition, the unbiased and wide-scale identification of protein-protein interaction networks and protein kinase substrates in lymphocyte signaling pathways can be achieved by mass spectrometry-based approaches. Critically, the integration of these discovery-driven strategies with single-cell analysis using mass cytometry can facilitate the understanding of complex signaling phenotypes in distinct immunophenotypes.
Introduction
High-resolution mass spectrometry (MS) has emerged as an attractive wide-scale approach to studying intracellular and extracellular signaling events in lymphocytes. Identification and quantitation of thousands of proteins, as well as their modifications and interaction partners can now be achieved with this approach. Furthermore, the development of novel technologies utilizing MS, such as mass cytometry, has opened up a new age of performing highly parallel single-cell analysis. In this review, we will discuss recent applications of MS technology to unraveling complex signaling networks in lymphocytes.
Shotgun proteomics to interrogate lymphocyte signaling
A comprehensive investigation of protein signaling networks requires the ability to evaluate the dynamic composition of molecular components in time and space. Shotgun proteomics using high-resolution MS provides an unbiased, quantitative, and wide-scale, analysis of complex protein mixtures, delivering snapshots of proteome compositions during cellular processes. In a typical workflow, proteins extracted from biological samples are enzymatically digested and analyzed using reversed-phase liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Peptides identified using MS are fragmented and sequenced using tandem mass spectrometry (MS/MS), and resulting spectra are analyzed through database search engines to identify the corresponding peptide sequences and proteins. Finally, statistical validation of the data is performed, often through decoy search strategies in which the MS/MS spectra are competitively matched against databases containing both normal and reversed protein sequences to estimate the false discovery rate (FDR) for sequence assignment.
Shotgun proteomics is a powerful approach for quantitation of relative changes in peptide and protein abundance across different cellular states or treatments (Table 1). Both label-free and label-based strategies can be employed for quantitation and have been reviewed in detail elsewhere [1,2]. In LC-MS/MS experiments, MS data is collected for quantitation by the sequential determination of the m/z values and intensities of all peptides eluted into the instrument. Although selection of peptides for fragmentation and MS/MS generation is stochastic and based on the most abundant signals, MS-based quantitation using both label-free and SILAC strategies allows for quantitation in the absence of MS/MS spectra by retention time alignment and accurate mass analysis of MS spectra using information from replicate samples. Importantly, our group has demonstrated the power of this approach by showing that calculating phosphopeptide replicate selected ion chromatogram peak areas using retention time alignment increases the number of replicate peptides quantified 5-fold [3]. Conversely, quantitative information in isobaric mass tag experiments is generated from MS/MS spectra and thus can suffer from ‘missing data’ that may undermine the application of statistical tests.
Table 1.
Overview of quantitative approaches in MS-based proteomics
| Method | Sample application | Number of comparisons | Quantitation level | Sequencing depth | Quantitative accuracy | Cost | |
|---|---|---|---|---|---|---|---|
| Label-free | Spectral counting | Primary and cultured cells | Many | MS | +++ | + | $ |
| Ion intensity | Primary and cultured cells | Many | MS | +++ | ++ | $ | |
|
| |||||||
| Label-based | SILAC | Cultured cells | 2–3 | MS | ++ | +++ | $$ |
| Isobaric mass tag | Primary and cultured cells | 2–8 | MS/MS | ++ | ++ | $$$ | |
Shotgun proteomic approaches have benefited investigations of critical signaling pathways in lymphocytes as it has enabled the identification and quantitation of thousands of global alterations in the proteomes of lymphocytes across a diverse range of cellular processes. These MS-based proteomic studies have revealed: (a) insights into proteomic changes occurring in response to various cellular treatments [4–8], (b) unique proteomic signatures across the maturation states of lymphocytes [9,10], and (c) proteins with clinical significance in lymphocyte disease states [11–13]. Another powerful utility of quantitative MS-based proteomics is the interrogation of protein networks within isolated subcellular locations in lymphocytes. Already, the membrane proteomes of primary B cells, NK cells, and T cells have been characterized utilizing this technique on plasma membrane enriched samples [14–16]. Studies have also performed proteomic characterizations of enriched secretory lysosomes from cytotoxic T cells and NK cells [17–19].
Emerging ‘omics’ fields from MS-based proteomics
Harnessing the high-throughput power of MS, investigators have begun the characterization of the secretome and immunopeptidome from signaling lymphocytes. Secreted proteins, which include cytokines, interleukins, and growth factors, are critical intercellular messengers in the immune system, mediating both the communication between effector cells and the orchestration of the immune response. Recently, a quantitative, high-resolution MS workflow was described to detect and quantify the time-resolved release of proteins from immune cells, the secretome, upon receptor ligation. From this investigation, the authors detected secreted proteins whose abundance increased by a factor of greater than 10,000, illustrating the significant sensitivity and dynamic range of this label-free quantitative approach [20]. Although this particular workflow was applied to macrophages, it will have great utility in interrogating lymphocyte responses to various stimuli. Another recent study implemented quantitative MS along with next-generation sequencing technology to profile the serum antibody repertoire elicited by vaccination, detailing its molecular composition and characteristics [21]. Utilizing similar approaches will be invaluable to defining the serum antibody repertoire and how its diversity and specificity changes in response to diseases.
Another area of research that is enabled by MS is the comprehensive analysis of both the membrane-bound and plasma soluble human immunopeptidome [22]. The immunopeptidome is the collection of thousands of peptides displayed by MHC molecules. Presentation of MHC molecules with their bound immunopeptidomes facilitates the scrutiny of the health-state of cells by circulating T cells. To perform this analysis, peptides bound to immunoaffinity purified MHC are extracted and analyzed by LC-MS/MS. Already, MS-based investigations have revealed the complexity and plasticity of the immunopeptidome and have highlighted critical information on the internal state of the cell that can be gleaned from a system-levels analysis of its composition [23–27].
MS-based investigations of protein-protein interaction networks
Protein-protein interaction (PPI) networks play a critical role in signal transduction in lymphocytes. For example, in T cell receptor (TCR) signaling, scaffolding molecules such as LAT nucleate the assembly of multiprotein complexes that are critical for amplification and diversification of signals that mediate responses resulting from TCR engagement [28]. The study of PPIs has been greatly facilitated by MS technologies, in particular, affinity purification mass spectrometry (AP-MS), which requires the ectopic expression of epitope tags on target ‘bait’ proteins of interest as affinity capture probes. Using this approach, a broad scope of PPI networks can be obtained with high sensitivity, accuracy, versatility and speed. A collection of approaches to define PPIs using AP-MS has been reviewed elsewhere [29–36]. Many of these approaches have been implemented to interrogate PPI networks in lymphocyte signaling [37–40]. Recently, mice bearing a genetic tag that permit AP-MS of signaling complexes containing ZAP-70, LAT, and SLP-76 was described using label-free quantitation. From this system, a membrane-proximal TCR signaling network was identified along with quantitative insights into the temporal regulation of these complexes [39]. A concern for AP-MS experiments is the potential loss of weak interactors. To address this issue, Li et al. implemented an enzyme-generated proximity labeling strategy combined with AP-MS to investigate the molecular composition of B cell receptor (BCR) clusters. In this approach, B cells were treated with HRP-conjugated IgM followed by tyramide-biotin, resulting in biotinylation of proximal proteins. These biotinylated proteins were captured by immobilized streptavidin and analyzed by quantitative MS using SILAC [40].
MS-based investigations of phosphorylation networks in lymphocyte signaling
Post-translational modifications (PTMs) such as phosphorylation, ubiquitination, acetylation, and glycosylation, are critical regulators of signal transduction, enzymatic activity, and protein interactions in immune function. Phosphorylation is the most extensively studied PTM in wide-scale MS-based studies to date as a result of advances in enrichment protocols, MS instrumentation, quantitation strategies, and software tools. Application of MS-based phosphoproteomics approaches has allowed for the unbiased identification and quantitation of thousands of dynamic phosphorylation signaling events in immunological signaling pathways. As we will discuss below, these studies have uncovered a wealth of knowledge on phosphorylation dynamics downstream of immune receptor stimulation.
Our lab as well as other groups have implemented quantitative MS-based phosphoproteomic strategies to perform multi-dimensional comparisons of tyrosine signaling events from mutant and wild type T cells through a time course of TCR stimulation [41–44]. Using this approach, we can accurately quantitate temporal changes in phosphorylation on a large number of sites in response to lymphocyte stimulation (Figure 1). The availability of various mutant clones, as well as the ease of manipulation and scalability of the Jurkat T cell line has made it an ideal model system to interrogate critical pathways downstream of TCR stimulation. The goal of this work is to place the large number of uncharacterized phosphorylation sites revealed in these studies relative to canonical T cell signaling landmarks, providing biological insights into the role of each phosphorylation site. Using this approach, our lab recently demonstrated that pharmacological inhibition of ERK activation in Jurkat T cells resulted in constitutive decreases on the majority of upstream tyrosine phosphorylation sites across a TCR stimulation time course, revealing a critical role for ERK positive feedback in TCR signaling [41]. Various SLP-76 mutant Jurkat cell lines have also been interrogated by our lab using this approach, revealing new modes of phosphoregulation on various critical TCR signaling proteins, including the Src family kinases Lck and Fyn, by SLP-76 [3,42].
Figure 1. Quantitative phosphoproteomic analysis of canonical TCR signaling proteins.
Label-free heatmaps represent the temporal change in phosphorylation of Jurkat T cells at 0, 3, 5, and 10 minutes of TCR stimulation. Heatmaps were calculated from the averages of five biological replicate experiments. In the label-free heatmaps, black represents peptide abundance equal to the geometric mean for that peptide across all time points. Blue represents peptide abundance less than the mean, whereas yellow corresponds to peptide abundance greater than the mean. A white dot within a label-free heatmap square indicates a statistically significant difference (q value < 0.05) in the fold change in peptide abundance for that time point.
A large number of studies have exploited the power of MS-based phosphoproteomics to interrogate a variety of signaling paradigms in lymphocytes. For example, MS-based phosphoproteomic studies have used the Jurkat T cell model to investigate TCR-induced activation events at high temporal resolutions. These studies have highlighted the diverse and dynamic patterns of tyrosine phosphorylation that occur seconds after TCR engagement [45,46]. Furthermore, a comprehensive comparison of tyrosine signaling events triggered by stimulation of different types of receptors was achieved using a similar strategy [47,48]. A large body of literature has implemented the quantitative proteomics workflow to interrogate signaling profiles of various B cell lymphomas [49,50]. Large-scale quantitative phosphoproteomics methods have also been implemented to gain system-wide insights into the role of serine/threonine phosphorylation during lymphocyte activation [51–55].
The application of quantitative phosphoproteomics to interrogate signaling pathways in primary lymphocytes has recently become feasible with the improvement of instrument sensitivity and mass accuracy, allowing for a physiologically relevant understanding of phosphorylation dynamics in signaling events [49,56–59]. However, limited protein abundance along with the substoichiometric nature of tyrosine site modifications, which is estimated to only make up ~0.5% of the total phosphoproteome [60], has made wide-scale analysis of tyrosine phosphorylation dynamics in primary T cell systems difficult. Nevertheless, studies have interrogated tyrosine phosphorylation events in primary T cells, providing critical insights into tyrosine signaling pathways in a physiological system [56,61].
Statistical analysis of proteomic datasets
A critical requirement for quantifying statistically significant observations in both proteomic and phosphoproteomic datasets is the analysis of at least 3 biological replicates. Many published MS-based studies lack sufficient replicate analyses to perform statistical validation of observations. Furthermore, because of the large number of peptides quantified in these experiments, implementation of multiple testing corrections is required. Corrections such as the family-wise error rate (FWER) or the FDR can be utilized to limit the number of false positive results and have been reviewed extensively elsewhere [62,63]. Our group calculates statistical significance by generating a q-value, the FDR-adjusted p-value, for each replicate observation. A software package called “QVALUE” found on the open-source statistical software R can be used to calculate q-values from p-values [64]. Because of the relative ease of calculating p-values, the software provides an accessible platform to perform this type of robust statistical analysis.
New genetic tools to interrogate signaling pathways
Recent advances in genetic engineering have enabled highly specific perturbations of signaling pathways. In the “analog-sensitive kinase” technology introduced by Kevan Shokat’s group, the ATP-binding pocket of a kinase’s catalytic domain is mutated so that it only accommodates a larger analog of a common non-selective kinase inhibitor [65]. The utility of this technology in interrogating signaling pathways has been demonstrated in recent publications from Arthur Weiss’s group. Interested in understanding the function of basal signaling in T cell activation, they generated mice with an AS allele of Csk in a Csk-deficient genetic background. Using this approach, they confirmed the critical role of Csk in restricting Src family kinase activity in resting primary T cells to maintain basal equilibrium [66]. In another study, this technology was employed to generate mice harboring a ZAP-70 AS allele, which enabled modulation of ZAP-70 kinase activity. Using this system, a sharp TCR signaling threshold for commitment to proliferation was revealed [67]. Engineered AS kinases are powerful tools in which the specificities of critical kinases in lymphocyte signaling pathways can be addressed with minimal off-target effects. Future studies employing this particular model will benefit greatly from phosphoproteomic profiling to determine pathway substrates of kinases within immune signaling pathways that are independent of a potential scaffolding role of the kinase.
MS-based investigations of kinase substrates
Although MS-based phosphoproteomics has facilitated global analysis of phosphorylation networks, this approach in itself is unable to definitively match a given phosphorylation event with an upstream kinase. To address this issue, various groups have designed approaches to screen for kinase substrates using purified active kinases to phosphorylate cell lysates in vitro, followed by MS analysis to identify phosphoproteins [68–74]. A major pitfall of these approaches is that they only identify potential kinase substrates in vitro. To bridge the gap between in vitro phosphorylation and physiological phosphorylation events, Xue et al. introduced a proKALIP approach, in which an in vitro kinase assay-based substrate screen was integrated with in vivo kinase dependent phosphorylation profiling [75]. Proteins tyrosine phosphorylated in vitro overlapping with the kinase-dependent phosphoproteome in vivo represented physiological direct substrates. Using this approach, bona fide substrates of the Src protein kinase Syk in B cell signaling were identified [75]. Another approach has recently been developed that addresses the challenges of performing these types of experiments for serine/threonine kinases, which make up roughly 99% of the phosphoproteome and thus present high background in in vitro kinase assays. In siKALIP, γ-18O-phosphoate ATP and moderate dephosphorylation is incorporated into the kinase assay to identify direct kinase substrates in high throughput [76].
Single-cell analysis of lymphocyte signaling
The enormous repertoire and heterogeneity of lymphocytes is well established and plays an important role in generating plastic and diverse responses [77]. Understanding the role of population heterogeneity and the mechanisms by which protein expression levels and PTMs influence cellular signaling and decision-making requires measurements of phenotypic variation at the single-cell level. Seminal work by both Scott Tanner’s and Gary Nolan’s groups have led to the development of mass cytometry, a technology that couples the single-cell resolution of flow cytometry with the measurement resolution of MS. Antibodies used in this approach are conjugated to metal isotopes that can be differentiated by the mass spectrometer. Because the method is largely unhampered by interference from spectral overlap, it allows for the detection of substantially more simultaneous parameters than traditional flow cytometry (reviewed in [78,79]).
Studies utilizing this approach have revealed that immune cell heterogeneity tends to manifest itself on a continuous scale rather than through distinct populations, underscoring the requirement for greater understanding of signaling pathways on a single-cell level [80,81]. Mass cytometry has also been implemented to investigate the mechanisms utilized by T cells to gauge antigen dose and make the decision to become anergic. The findings revealed that the intracellular balance and signal integration between TCR activating and inhibitory signaling molecules serves as a molecular switch gauging antigen dose [82]. A clear advantage of this technique over traditional MS-based proteomics and phosphoproteomics is the ability to investigate small sample sizes, making it invaluable to the study of primary cells. Despite this advantage, mass cytometry suffers from the lack of unbiased measurements, as well as diminished depth of coverage that is attainable using shotgun proteomics. Furthermore, the subset of modifications or proteins that can be studied is limited by the availability of antibodies. Nevertheless, implementation of mass cytometry has enabled the quantitation of signal transduction pathways downstream of distinct immunophenotypes, bridging single-cell analysis with a systems-level view of hematopoiesis and immunology in diverse lymphocyte populations. Studies will benefit from a synergistic coupling of the two technologies, where phosphoproteomics identifies a list of candidate sites that can be followed up on with mass cytometry to achieve cellular understanding of bulk assay data.
Conclusions
Unbiased and wide-scale analysis of cellular protein compositions is now achievable with MS-based proteomics technologies, enabling comprehensive examinations into the thousands of signaling events that mediate cellular responses. Investigations into the signaling architecture of lymphocytes in particular has benefitted from recent advances in high-resolution MS. There has been impressive growth in our understanding of the role of phosphorylation in mediating critical signaling events. Additionally, the characterization of dynamic protein interaction networks has been facilitated using MS-based approaches and new ‘omics’ fields are being developed based on MS technologies. Despite these advances, the field still suffers from a lack of appropriate replicate and statistical analyses, which has markedly diminished the capacity to glean biological conclusions from these datasets. As the field moves forward, it is important that journals implement more stringent requirements for publication of proteomic and phosphoproteomic data, by requiring sufficient biological replicates to ascertain statistical significance.
Mass spectrometry (MS) enables the unbiased and comprehensive study of lymphocyte signaling
New ‘omics’ fields in immunology are emerging from MS
Biological replicates are crucial to ascertain statistical significance in MS-based studies
Acknowledgments
The authors wish to acknowledge financial support from NIH grant R01 AI083636.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Romanova EV, Dowd SE, Sweedler JV. Quantitation of endogenous peptides using mass spectrometry based methods. Curr Opin Chem Biol. 2013;17:801–808. doi: 10.1016/j.cbpa.2013.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Schulze WX, Usadel B. Quantitation in mass-spectrometry-based proteomics. Annu Rev Plant Biol. 2010;61:491–516. doi: 10.1146/annurev-arplant-042809-112132. [DOI] [PubMed] [Google Scholar]
- 3.Ji Q, Ding Y, Salomon AR. SLP-76 N-terminal tyrosine residues regulate a dynamic signaling equilibrium involving feedback of proximal TCR signaling. Mol Cell Proteomics. 2014 doi: 10.1074/mcp.M114.037861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lichtenfels R, Rappl G, Hombach AA, Recktenwald CV, Dressler SP, Abken H, Seliger B. A proteomic view at T cell costimulation. PLoS One. 2012;7:e32994. doi: 10.1371/journal.pone.0032994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ma D, Cao W, Kapur A, Felder M, Scarlett CO, Patankar MS, Li L. Differential expression of proteins in naive and IL-2 stimulated primary human NK cells identified by global proteomic analysis. J Proteomics. 2013;91:151–163. doi: 10.1016/j.jprot.2013.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Diandong H, Kefeng S, Weixin F, Zaifu L. Proteomic analysis of NK92-MI cells activated by neuropeptide substance P. Neuropeptides. 2013;47:157–162. doi: 10.1016/j.npep.2013.01.002. [DOI] [PubMed] [Google Scholar]
- 7.Chen YJ, Yu CC, Chen ST, Chen TY, Liao HF. Functional regulation and proteomic characterization of human natural killer cells through recombinant human granulocyte-colony stimulating factor treatment. Proteomics Clinical Applications. 2009;3:563–573. [Google Scholar]
- 8.Huang X, Shen Y, Liu M, Bi C, Jiang C, Iqbal J, McKeithan TW, Chan WC, Ding SJ, Fu K. Quantitative proteomics reveals that miR-155 regulates the PI3K-AKT pathway in diffuse large B-cell lymphoma. Am J Pathol. 2012;181:26–33. doi: 10.1016/j.ajpath.2012.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Salonen J, Ronnholm G, Kalkkinen N, Vihinen M. Proteomic changes during B cell maturation: 2D-DIGE approach. PLoS One. 2013;8:e77894. doi: 10.1371/journal.pone.0077894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Scheiter M, Lau U, van Ham M, Bulitta B, Grobe L, Garritsen H, Klawonn F, Konig S, Jansch L. Proteome analysis of distinct developmental stages of human natural killer (NK) cells. Mol Cell Proteomics. 2013;12:1099–1114. doi: 10.1074/mcp.M112.024596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kashuba E, Eagle GL, Bailey J, Evans P, Welham KJ, Allsup D, Cawkwell L. Proteomic analysis of B-cell receptor signaling in chronic lymphocytic leukaemia reveals a possible role for kininogen. J Proteomics. 2013;91:478–485. doi: 10.1016/j.jprot.2013.08.002. [DOI] [PubMed] [Google Scholar]
- 12.Stranneheim H, Orre LM, Lehtio J, Flygare J. A comparison between protein profiles of B cell subpopulations and mantle cell lymphoma cells. Proteome Sci. 2009;7:43. doi: 10.1186/1477-5956-7-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Deeb SJ, D’Souza RC, Cox J, Schmidt-Supprian M, Mann M. Super-SILAC allows classification of diffuse large B-cell lymphoma subtypes by their protein expression profiles. Mol Cell Proteomics. 2012;11:77–89. doi: 10.1074/mcp.M111.015362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Boyd RS, Jukes-Jones R, Walewska R, Brown D, Dyer MJ, Cain K. Protein profiling of plasma membranes defines aberrant signaling pathways in mantle cell lymphoma. Mol Cell Proteomics. 2009;8:1501–1515. doi: 10.1074/mcp.M800515-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.de Wet B, Zech T, Salek M, Acuto O, Harder T. Proteomic characterization of plasma membrane-proximal T cell activation responses. J Biol Chem. 2011;286:4072–4080. doi: 10.1074/jbc.M110.165415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ghosh D, Lippert D, Krokhin O, Cortens JP, Wilkins JA. Defining the membrane proteome of NK cells. J Mass Spectrom. 2010;45:1–25. doi: 10.1002/jms.1696. [DOI] [PubMed] [Google Scholar]
- 17.Schmidt H, Gelhaus C, Nebendahl M, Lettau M, Lucius R, Leippe M, Kabelitz D, Janssen O. Effector granules in human T lymphocytes: the luminal proteome of secretory lysosomes from human T cells. Cell Commun Signal. 2011;9:4. doi: 10.1186/1478-811X-9-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Schmidt H, Gelhaus C, Nebendahl M, Lettau M, Lucius R, Leippe M, Kabelitz D, Janssen O. Effector granules in human T lymphocytes: proteomic evidence for two distinct species of cytotoxic effector vesicles. J Proteome Res. 2011;10:1603–1620. doi: 10.1021/pr100967v. [DOI] [PubMed] [Google Scholar]
- 19.Schmidt H, Gelhaus C, Nebendahl M, Lettau M, Watzl C, Kabelitz D, Leippe M, Janssen O. 2-D DIGE analyses of enriched secretory lysosomes reveal heterogeneous profiles of functionally relevant proteins in leukemic and activated human NK cells. Proteomics. 2008;8:2911–2925. doi: 10.1002/pmic.200800170. [DOI] [PubMed] [Google Scholar]
- 20*.Meissner F, Scheltema RA, Mollenkopf HJ, Mann M. Direct proteomic quantification of the secretome of activated immune cells. Science. 2013;340:475–478. doi: 10.1126/science.1232578. This study describes a quantitative, high-resolution MS workflow to detect and quantify proteins that are released from immune cells upon receptor ligation. [DOI] [PubMed] [Google Scholar]
- 21.Boutz DR, Horton AP, Wine Y, Lavinder JJ, Georgiou G, Marcotte EM. Proteomic identification of monoclonal antibodies from serum. Anal Chem. 2014;86:4758–4766. doi: 10.1021/ac4037679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Admon A, Bassani-Sternberg M. The Human Immunopeptidome Project, a suggestion for yet another postgenome next big thing. Mol Cell Proteomics. 2011;10:O111 011833. doi: 10.1074/mcp.O111.011833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Mester G, Hoffmann V, Stevanovic S. Insights into MHC class I antigen processing gained from large-scale analysis of class I ligands. Cell Mol Life Sci. 2011;68:1521–1532. doi: 10.1007/s00018-011-0659-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Caron E, Vincent K, Fortier MH, Laverdure JP, Bramoulle A, Hardy MP, Voisin G, Roux PP, Lemieux S, Thibault P, et al. The MHC I immunopeptidome conveys to the cell surface an integrative view of cellular regulation. Mol Syst Biol. 2011;7:533. doi: 10.1038/msb.2011.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Espinosa G, Collado JA, Scholz E, Mestre-Ferrer A, Kuse N, Takiguchi M, Carrascal M, Canals F, Pujol-Borrell R, Jaraquemada D, et al. Peptides presented by HLA class I molecules in the human thymus. J Proteomics. 2013;94:23–36. doi: 10.1016/j.jprot.2013.08.023. [DOI] [PubMed] [Google Scholar]
- 26*.Granados DP, Sriranganadane D, Daouda T, Zieger A, Laumont CM, Caron-Lizotte O, Boucher G, Hardy MP, Gendron P, Cote C, et al. Impact of genomic polymorphisms on the repertoire of human MHC class I-associated peptides. Nat Commun. 2014;5:3600. doi: 10.1038/ncomms4600. This study describes a novel approach that combines next-generation sequencing and MS that enables high-throughput discovery of polymorphic MHC class I-associated peptides (MIPs) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kumari S, Walchli S, Fallang LE, Yang W, Lund-Johansen F, Schumacher TN, Olweus J. Alloreactive cytotoxic T cells provide means to decipher the immunopeptidome and reveal a plethora of tumor-associated self-epitopes. Proc Natl Acad Sci USA. 2014;111:403–408. doi: 10.1073/pnas.1306549111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Malissen B, Gregoire C, Malissen M, Roncagalli R. Integrative biology of T cell activation. Nat Immunol. 2014;15:790–797. doi: 10.1038/ni.2959. [DOI] [PubMed] [Google Scholar]
- 29**.Kaake RM, Wang X, Burke A, Yu C, Kandur W, Yang Y, Novtisky EJ, Second T, Duan J, Kao A, et al. A new in vivo cross-linking mass spectrometry platform to define protein-protein interactions in living cells. Mol Cell Proteomics. 2014 doi: 10.1074/mcp.M114.042630. This study describes a new in vivo strategy that couples a novel membrane permeable, enrichable, and MS-cleavable cross-linker with MS to identify protein interaction interfaces. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Budayeva HG, Cristea IM. A mass spectrometry view of stable and transient protein interactions. Adv Exp Med Biol. 2014;806:263–282. doi: 10.1007/978-3-319-06068-2_11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Walzthoeni T, Leitner A, Stengel F, Aebersold R. Mass spectrometry supported determination of protein complex structure. Curr Opin Struct Biol. 2013;23:252–260. doi: 10.1016/j.sbi.2013.02.008. [DOI] [PubMed] [Google Scholar]
- 32.Marcilla M, Albar JP. Quantitative proteomics: A strategic ally to map protein interaction networks. IUBMB Life. 2013;65:9–16. doi: 10.1002/iub.1081. [DOI] [PubMed] [Google Scholar]
- 33.Armean IM, Lilley KS, Trotter MW. Popular computational methods to assess multiprotein complexes derived from label-free affinity purification and mass spectrometry (AP-MS) experiments. Mol Cell Proteomics. 2013;12:1–13. doi: 10.1074/mcp.R112.019554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Choi H. Computational detection of protein complexes in AP-MS experiments. Proteomics. 2012;12:1663–1668. doi: 10.1002/pmic.201100508. [DOI] [PubMed] [Google Scholar]
- 35.Gingras AC, Raught B. Beyond hairballs: The use of quantitative mass spectrometry data to understand protein-protein interactions. FEBS Lett. 2012;586:2723–2731. doi: 10.1016/j.febslet.2012.03.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Dunham WH, Mullin M, Gingras AC. Affinity-purification coupled to mass spectrometry: basic principles and strategies. Proteomics. 2012;12:1576–1590. doi: 10.1002/pmic.201100523. [DOI] [PubMed] [Google Scholar]
- 37.Orr SJ, Boutz DR, Wang R, Chronis C, Lea NC, Thayaparan T, Hamilton E, Milewicz H, Blanc E, Mufti GJ, et al. Proteomic and protein interaction network analysis of human T lymphocytes during cell-cycle entry. Mol Syst Biol. 2012;8:573. doi: 10.1038/msb.2012.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fierro-Monti I, Echeverria P, Racle J, Hernandez C, Picard D, Quadroni M. Dynamic impacts of the inhibition of the molecular chaperone Hsp90 on the T-cell proteome have implications for anti-cancer therapy. PLoS One. 2013;8:e80425. doi: 10.1371/journal.pone.0080425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39**.Roncagalli R, Hauri S, Fiore F, Liang Y, Chen Z, Sansoni A, Kanduri K, Joly R, Malzac A, Lahdesmaki H, et al. Quantitative proteomics analysis of signalosome dynamics in primary T cells identifies the surface receptor CD6 as a Lat adaptor-independent TCR signaling hub. Nat Immunol. 2014;15:384–392. doi: 10.1038/ni.2843. This study describes a series of mice bearing a genetic tag that permits AP-MS analysis of endogenous signaling complexes containing ZAP-70, LAT, and SLP-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li XW, Rees JS, Xue P, Zhang H, Hamaia SW, Sanderson B, Funk PE, Farndale RW, Lilley KS, Perrett S, et al. New insights into the DT40 B cell receptor cluster using a proteomic proximity labeling assay. J Biol Chem. 2014;289:14434–14447. doi: 10.1074/jbc.M113.529578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Helou YA, Nguyen V, Beik SP, Salomon AR. ERK positive feedback regulates a widespread network of tyrosine phosphorylation sites across canonical T cell signaling and actin cytoskeletal proteins in Jurkat T cells. PLoS One. 2013;8:e69641. doi: 10.1371/journal.pone.0069641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Cao L, Ding Y, Hung N, Yu K, Ritz A, Raphael BJ, Salomon AR. Quantitative phosphoproteomics reveals SLP-76 dependent regulation of PAG and Src family kinases in T cells. PLoS One. 2012;7:e46725. doi: 10.1371/journal.pone.0046725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nguyen V, Cao L, Lin JT, Hung N, Ritz A, Yu K, Jianu R, Ulin SP, Raphael BJ, Laidlaw DH, et al. A new approach for quantitative phosphoproteomic dissection of signaling pathways applied to T cell receptor activation. Mol Cell Proteomics. 2009;8:2418–2431. doi: 10.1074/mcp.M800307-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Salek M, McGowan S, Trudgian DC, Dushek O, de Wet B, Efstathiou G, Acuto O. Quantitative phosphoproteome analysis unveils LAT as a modulator of CD3zeta and ZAP-70 tyrosine phosphorylation. PLoS One. 2013;8:e77423. doi: 10.1371/journal.pone.0077423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Brockmeyer C, Paster W, Pepper D, Tan CP, Trudgian DC, McGowan S, Fu G, Gascoigne NRJ, Acuto O, Salek M. T cell receptor (TCR)-induced tyrosine phosphorylation dynamics identifies THEMIS as a new TCR signalosome component. J Biol Chem. 2011;286:7535–7547. doi: 10.1074/jbc.M110.201236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Chylek LA, Akimov V, Dengjel J, Rigbolt KT, Hu B, Hlavacek WS, Blagoev B. Phosphorylation site dynamics of early T-cell receptor signaling. PLoS One. 2014;9:e104240. doi: 10.1371/journal.pone.0104240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Matsumoto M, Oyamada K, Takahashi H, Sato T, Hatakeyama S, Nakayama KI. Largescale proteomic analysis of tyrosine-phosphorylation induced by T-cell receptor or B-cell receptor activation reveals new signaling pathways. Proteomics. 2009;9:3549–3563. doi: 10.1002/pmic.200900011. [DOI] [PubMed] [Google Scholar]
- 48.Kim JE, White FM. Quantitative analysis of phosphotyrosine signaling networks triggered by CD3 and CD28 costimulation in Jurkat cells. J Immunol. 2006;176:2833–2843. doi: 10.4049/jimmunol.176.5.2833. [DOI] [PubMed] [Google Scholar]
- 49.Rolland D, Basrur V, Conlon K, Wolfe T, Fermin D, Nesvizhskii AI, Lim MS, Elenitoba-Johnson KS. Global phosphoproteomic profiling reveals distinct signatures in B-cell non-Hodgkin lymphomas. Am J Pathol. 2014;184:1331–1342. doi: 10.1016/j.ajpath.2014.01.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.McDonnell SR, Hwang SR, Rolland D, Murga-Zamalloa C, Basrur V, Conlon KP, Fermin D, Wolfe T, Raskind A, Ruan C, et al. Integrated phosphoproteomic and metabolomic profiling reveals NPM-ALK-mediated phosphorylation of PKM2 and metabolic reprogramming in anaplastic large cell lymphoma. Blood. 2013;122:958–968. doi: 10.1182/blood-2013-01-482026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mayya V, Lundgren DH, Hwang SI, Rezaul K, Wu L, Eng JK, Rodionov V, Han DK. Quantitative phosphoproteomic analysis of T cell receptor signaling reveals system-wide modulation of protein-protein interactions. Sci Signal. 2009;2:ra46. doi: 10.1126/scisignal.2000007. [DOI] [PubMed] [Google Scholar]
- 52.Jayasundera KB, Iliuk AB, Nguyen A, Higgins R, Geahlen RL, Tao WA. Global phosphoproteomics of activated B cells using complementary metal ion functionalized soluble nanopolymers. Anal Chem. 2014;86:6363–6371. doi: 10.1021/ac500599r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pighi C, Gu TL, Dalai I, Barbi S, Parolini C, Bertolaso A, Pedron S, Parisi A, Ren J, Cecconi D, et al. Phospho-proteomic analysis of mantle cell lymphoma cells suggests a pro-survival role of B-cell receptor signaling. Cell Oncol. 2011;34:141–153. doi: 10.1007/s13402-011-0019-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gioia R, Leroy C, Drullion C, Lagarde V, Etienne G, Dulucq S, Lippert E, Roche S, Mahon FX, Pasquet JM. Quantitative phosphoproteomics revealed interplay between Syk and Lyn in the resistance to nilotinib in chronic myeloid leukemia cells. Blood. 2011;118:2211–2221. doi: 10.1182/blood-2010-10-313692. [DOI] [PubMed] [Google Scholar]
- 55.Rubbi L, Titz B, Brown L, Galvan E, Komisopoulou E, Chen SS, Low T, Tahmasian M, Skaggs B, Muschen M, et al. Global phosphoproteomics reveals crosstalk between Bcr-Abl and negative feedback mechanisms controlling Src signaling. Sci Signal. 2011;4:ra18. doi: 10.1126/scisignal.2001314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ruperez P, Gago-Martinez A, Burlingame AL, Oses-Prieto JA. Quantitative phosphoproteomic analysis reveals a role for serine and threonine kinases in the cytoskeletal reorganization in early T cell receptor activation in human primary T cells. Mol Cell Proteomics. 2012;11:171–186. doi: 10.1074/mcp.M112.017863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Navarro MN, Goebel J, Feijoo-Carnero C, Morrice N, Cantrell DA. Phosphoproteomic analysis reveals an intrinsic pathway for the regulation of histone deacetylase 7 that controls the function of cytotoxic T lymphocytes. Nat Immunol. 2011;12:352–361. doi: 10.1038/ni.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Tsai CM, Wu HY, Su TH, Kuo CW, Huang HW, Chung CH, Chen CS, Khoo KH, Chen YJ, Lin KI. Phosphoproteomic analyses reveal that galectin-1 augments the dynamics of B-cell receptor signaling. J Proteomics. 2014;103:241–253. doi: 10.1016/j.jprot.2014.03.031. [DOI] [PubMed] [Google Scholar]
- 59.Caruthers NJ, Stemmer PM, Shin N, Dombkowski A, Caruso JA, Gill R, Rosenspire A. Mercury alters B-cell protein phosphorylation profiles. J Proteome Res. 2014;13:496–505. doi: 10.1021/pr400657k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hunter T, Sefton BM. Transforming gene product of Rous sarcoma virus phosphorylates tyrosine. Proc Natl Acad Sci USA. 1980;77:1311–1315. doi: 10.1073/pnas.77.3.1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Iwai LK, Benoist C, Mathis D, White FM. Quantitative phosphoproteomic analysis of T cell receptor signaling in diabetes prone and resistant mice. J Proteome Res. 2010;9:3135–3145. doi: 10.1021/pr100035b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Diz AP, Carvajal-Rodriguez A, Skibinski DO. Multiple hypothesis testing in proteomics: a strategy for experimental work. Mol Cell Proteomics. 2011;10:M110 004374. doi: 10.1074/mcp.M110.004374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chadeau-Hyam M, Campanella G, Jombart T, Bottolo L, Portengen L, Vineis P, Liquet B, Vermeulen RC. Deciphering the complex: methodological overview of statistical models to derive OMICS-based biomarkers. Environ Mol Mutagen. 2013;54:542–557. doi: 10.1002/em.21797. [DOI] [PubMed] [Google Scholar]
- 64.Storey JD. The positive false discovery rate: A Bayesian interpretation and the q-value. Annals of Statistics. 2003;31:2013–2035. [Google Scholar]
- 65*.Lopez MS, Choy JW, Peters U, Sos ML, Morgan DO, Shokat KM. Staurosporine-derived inhibitors broaden the scope of analog-sensitive kinase technology. J Am Chem Soc. 2013;135:18153–18159. doi: 10.1021/ja408704u. This study describes the structure-based design of synthetically tractable, potent, and selective AS-kinase inhibitors based on staurosporine. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tan YX, Manz BN, Freedman TS, Zhang C, Shokat KM, Weiss A. Inhibition of the kinase Csk in thymocytes reveals a requirement for actin remodeling in the initiation of full TCR signaling. Nat Immunol. 2014;15:186–194. doi: 10.1038/ni.2772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67*.Au-Yeung BB, Zikherman J, Mueller JL, Ashouri JF, Matloubian M, Cheng DA, Chen Y, Shokat KM, Weiss A. A sharp T-cell antigen receptor signaling threshold for T-cell proliferation. Proc Natl Acad Sci USA. 2014;111:E3679–3688. doi: 10.1073/pnas.1413726111. This study utilizes the combination of a ZAP-70 AS chemical-genetic system and the Nur77-GFP reporter to track and modulate TCR signal strength integrated over time. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Morandell S, Grosstessner-Hain K, Roitinger E, Hudecz O, Lindhorst T, Teis D, Wrulich OA, Mazanek M, Taus T, Ueberall F, et al. QIKS--Quantitative identification of kinase substrates. Proteomics. 2010;10:2015–2025. doi: 10.1002/pmic.200900749. [DOI] [PubMed] [Google Scholar]
- 69.Kettenbach AN, Schweppe DK, Faherty BK, Pechenick D, Pletnev AA, Gerber SA. Quantitative phosphoproteomics identifies substrates and functional modules of Aurora and Polo-like kinase activities in mitotic cells. Sci Signal. 2011;4:rs5. doi: 10.1126/scisignal.2001497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Xue L, Wang WH, Iliuk A, Hu L, Galan JA, Yu S, Hans M, Geahlen RL, Tao WA. Sensitive kinase assay linked with phosphoproteomics for identifying direct kinase substrates. Proc Natl Acad Sci USA. 2012;109:5615–5620. doi: 10.1073/pnas.1119418109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chou MF, Prisic S, Lubner JM, Church GM, Husson RN, Schwartz D. Using bacteria to determine protein kinase specificity and predict target substrates. PLoS One. 2012;7:e52747. doi: 10.1371/journal.pone.0052747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kettenbach AN, Wang T, Faherty BK, Madden DR, Knapp S, Bailey-Kellogg C, Gerber SA. Rapid determination of multiple linear kinase substrate motifs by mass spectrometry. Chem Biol. 2012;19:608–618. doi: 10.1016/j.chembiol.2012.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Knight JD, Tian R, Lee RE, Wang F, Beauvais A, Zou H, Megeney LA, Gingras AC, Pawson T, Figeys D, et al. A novel whole-cell lysate kinase assay identifies substrates of the p38 MAPK in differentiating myoblasts. Skelet Muscle. 2012;2:5. doi: 10.1186/2044-5040-2-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Garre S, Senevirathne C, Pflum MK. A comparative study of ATP analogs for phosphorylation-dependent kinase-substrate crosslinking. Bioorg Med Chem. 2014;22:1620–1625. doi: 10.1016/j.bmc.2014.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75*.Xue L, Geahlen RL, Tao WA. Identification of direct tyrosine kinase substrates based on protein kinase assay-linked phosphoproteomics. Mol Cell Proteomics. 2013;12:2969–2980. doi: 10.1074/mcp.O113.027722. This study describes a proteomic strategy that integrates in vitro kinase assay-based substrate screening and endogenous kinase dependent phosphorylation profiling that can be used to identify kinase specificity and direct substrates in high throughput. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76**.Xue L, Wang P, Cao P, Zhu JK, Tao WA. Identification of ERK1 Direct Substrates using Stable Isotope Labeled Kinase Assay-Linked Phosphoproteomics. Mol Cell Proteomics. 2014 doi: 10.1074/mcp.O114.038588. This study describes a proteomic strategy to identify the specificity and direct substrates of kinases by coupling phosphoproteomics with a sensitive stable isotope labeled kinase reaction. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Satija R, Shalek AK. Heterogeneity in immune responses: from populations to single cells. Trends Immunol. 2014;35:219–229. doi: 10.1016/j.it.2014.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bjornson ZB, Nolan GP, Fantl WJ. Single-cell mass cytometry for analysis of immune system functional states. Curr Opin Immunol. 2013;25:484–494. doi: 10.1016/j.coi.2013.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chattopadhyay PK, Gierahn TM, Roederer M, Love JC. Single-cell technologies for monitoring immune systems. Nat Immunol. 2014;15:128–135. doi: 10.1038/ni.2796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Bendall SC, Simonds EF, Qiu P, Amir el AD, Krutzik PO, Finck R, Bruggner RV, Melamed R, Trejo A, Ornatsky OI, et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science. 2011;332:687–696. doi: 10.1126/science.1198704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Amir el AD, Davis KL, Tadmor MD, Simonds EF, Levine JH, Bendall SC, Shenfeld DK, Krishnaswamy S, Nolan GP, Pe’er D. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat Biotechnol. 2013;31:545–552. doi: 10.1038/nbt.2594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82**.Wolchinsky R, Hod-Marco M, Oved K, Shen-Orr SS, Bendall SC, Nolan GP, Reiter Y. Antigen-dependent integration of opposing proximal TCR-signaling cascades determines the functional fate of T lymphocytes. J Immunol. 2014;192:2109–2119. doi: 10.4049/jimmunol.1301142. This study utilizes mass cytometry to study the molecular mechanisms by which CTLs distinguish between low-to-optimal and supraoptimal levels of pMHC-I complexes. [DOI] [PubMed] [Google Scholar]

