Multiplex matrix network analysis of protein complexes in the human TCR signalosome

A multiplex antibody-mediated capture technique reveals patient-specific protein complexes. Personalized signaling complexes The response of a cell to ligands that activate cell surface receptors depends on the resulting protein-protein interactions that occur. The formation of different protein complexes leads to the activation of different intracellular signaling pathways and different cellular outputs. Thus, knowing which protein-protein interactions occur can be clinically important. To analyze these complexes in patient samples, Smith et al. devised a multiplex, antibody-based method and used it to capture complexes from T cell lysates and identify binding partners by flow cytometry. Mathematical analyses of these data enabled the construction of protein-protein interaction networks and revealed the relative abundances of particular complexes between resting and stimulated cells. Application of this technique to T cells from skin biopsies identified protein complexes that differed in their relative abundance between control donors and patients with an autoimmune skin disease. This type of analysis brings basic research a step closer to personalized therapy and may be useful as a diagnostic tool. Multiprotein complexes transduce cellular signals through extensive interaction networks, but the ability to analyze these networks in cells from small clinical biopsies is limited. To address this, we applied an adaptable multiplex matrix system to physiologically relevant signaling protein complexes isolated from a cell line or from human patient samples. Focusing on the proximal T cell receptor (TCR) signalosome, we assessed 210 pairs of PiSCES (proteins in shared complexes detected by exposed surface epitopes). Upon stimulation of Jurkat cells with superantigen-loaded antigen-presenting cells, this system produced high-dimensional data that enabled visualization of network activity. A comprehensive analysis platform generated PiSCES biosignatures by applying unsupervised hierarchical clustering, principal component analysis, an adaptive nonparametric with empirical cutoff analysis, and weighted correlation network analysis. We generated PiSCES biosignatures from 4-mm skin punch biopsies from control patients or patients with the autoimmune skin disease alopecia areata. This analysis distinguished disease patients from the controls, detected enhanced basal TCR signaling in the autoimmune patients, and identified a potential signaling network signature that may be indicative of disease. Thus, generation of PiSCES biosignatures represents an approach that can provide information about the activity of protein signaling networks in samples including low-abundance primary cells from clinical biopsies.

[1]  A. Schrum Visualization of Multiprotein Complexes by Flow Cytometry , 2009, Current protocols in immunology.

[2]  S. Raychaudhuri,et al.  Immune cell profiling to guide therapeutic decisions in rheumatic diseases , 2015, Nature Reviews Rheumatology.

[3]  Diana Gil,et al.  Robustness and Specificity in Signal Transduction via Physiologic Protein Interaction Networks , 2012, Clinical & experimental pharmacology.

[4]  R. Clark,et al.  A novel method for the isolation of skin resident T cells from normal and diseased human skin. , 2006, The Journal of investigative dermatology.

[5]  Hon Wai Leong,et al.  A survey of computational methods for protein complex prediction from protein interaction networks , 2012, J. Bioinform. Comput. Biol..

[6]  P. Allen,et al.  How the TCR balances sensitivity and specificity for the recognition of self and pathogens , 2012, Nature Immunology.

[7]  T. Pawson,et al.  Efficient T‐cell receptor signaling requires a high‐affinity interaction between the Gads C‐SH3 domain and the SLP‐76 RxxK motif , 2007, The EMBO journal.

[8]  S. L. Wong,et al.  Towards a proteome-scale map of the human protein–protein interaction network , 2005, Nature.

[9]  T. Pawson ReviewSpecificity in Signal Transduction : From Phosphotyrosine-SH 2 Domain Interactions to Complex Cellular Systems , 2004 .

[10]  E. Palmer,et al.  High-Sensitivity Detection and Quantitative Analysis of Native Protein-Protein Interactions and Multiprotein Complexes by Flow Cytometry , 2007, Science's STKE.

[11]  Benjamin A. Shoemaker,et al.  Deciphering Protein–Protein Interactions. Part II. Computational Methods to Predict Protein and Domain Interaction Partners , 2007, PLoS Comput. Biol..

[12]  Tony Pawson,et al.  Specificity in Signal Transduction From Phosphotyrosine-SH2 Domain Interactions to Complex Cellular Systems , 2004, Cell.

[13]  Lennart Martens,et al.  Annotating the human proteome: beyond establishing a parts list. , 2007, Biochimica et biophysica acta.

[14]  A. Weiss,et al.  Jurkat T cells and development of the T-cell receptor signalling paradigm , 2004, Nature Reviews Immunology.

[15]  B. Kholodenko,et al.  Computational Approaches for Analyzing Information Flow in Biological Networks , 2012, Science Signaling.

[16]  Barbara Hausmann,et al.  A motif in the αβ T-cell receptor controls positive selection by modulating ERK activity , 2000, Nature.

[17]  K. Takase,et al.  [T cell activation]. , 1995, Ryumachi. [Rheumatism].

[18]  J Douglas Armstrong,et al.  Reconstructing protein complexes: From proteomics to systems biology , 2006, Proteomics.

[19]  Jianmin Wu,et al.  PINA v2.0: mining interactome modules , 2011, Nucleic Acids Res..

[20]  M. Pittelkow,et al.  Signalling protein complexes isolated from primary human skin‐resident T cells can be analysed by Multiplex IP‐FCM , 2014, Experimental dermatology.

[21]  A. Weiss,et al.  Signal transduction by the TCR for antigen. , 2000, Current opinion in immunology.

[22]  Jason A. Papin,et al.  Reconstruction of cellular signalling networks and analysis of their properties , 2005, Nature Reviews Molecular Cell Biology.

[23]  Tony Pawson,et al.  Modular evolution of phosphorylation-based signalling systems , 2012, Philosophical Transactions of the Royal Society B: Biological Sciences.

[24]  Michiko Kato,et al.  Systems for the detection and analysis of protein–protein interactions , 2006, Applied Microbiology and Biotechnology.

[25]  F. Avilés,et al.  Detecting and interfering protein interactions: towards the control of biochemical pathways. , 2009, Current medicinal chemistry.

[26]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[27]  Steve Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

[28]  Xiufen Zou,et al.  A theoretical framework for specificity in cell signaling , 2005, Molecular systems biology.

[29]  P. Linsley,et al.  Binding of the B cell activation antigen B7 to CD28 costimulates T cell proliferation and interleukin 2 mRNA accumulation , 1991, The Journal of experimental medicine.

[30]  Diana Gil,et al.  Multiplex IP-FCM (immunoprecipitation-flow cytometry): Principles and guidelines for assessing physiologic protein-protein interactions in multiprotein complexes. , 2012, Methods.

[31]  Ursula Pieper,et al.  Protein complex compositions predicted by structural similarity , 2006, Nucleic acids research.

[32]  Jonathan M. Green,et al.  An enigmatic tail of CD28 signaling. , 2010, Cold Spring Harbor perspectives in biology.

[33]  Rainer Breitling,et al.  What is Systems Biology? , 2010, Front. Physiology.

[34]  M. Hayden,et al.  Ultrasensitive measurement of huntingtin protein in cerebrospinal fluid demonstrates increase with Huntington disease stage and decrease following brain huntingtin suppression , 2015, Scientific Reports.

[35]  Tony Pawson,et al.  Dynamic control of signaling by modular adaptor proteins. , 2007, Current opinion in cell biology.

[36]  Peter Langfelder,et al.  Fast R Functions for Robust Correlations and Hierarchical Clustering. , 2012, Journal of statistical software.

[37]  H. Lähdesmäki,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 , 2014, Nature Immunology.

[38]  William J. R. Longabaugh,et al.  Combing the hairball with BioFabric: a new approach for visualization of large networks , 2012, BMC Bioinformatics.

[39]  Annette Lee,et al.  Genome-wide association study in alopecia areata implicates both innate and adaptive immunity , 2010, Nature.

[40]  D. Vignali,et al.  Organization of proximal signal initiation at the TCR:CD3 complex , 2009, Immunological reviews.

[41]  M. Tyers,et al.  From genomics to proteomics , 2003, Nature.

[42]  A. Barabasi,et al.  Uncovering disease-disease relationships through the incomplete interactome , 2015, Science.

[43]  Ronald N Germain,et al.  Modeling T Cell Antigen Discrimination Based on Feedback Control of Digital ERK Responses , 2005, PLoS biology.

[44]  Mark M. Davis,et al.  Significance analysis of xMap cytokine bead arrays , 2012, Proceedings of the National Academy of Sciences.