A Network Module for the Perseus Software for Computational Proteomics Facilitates Proteome Interaction Graph AnalysisClick to copy article linkArticle link copied!
- Jan Daniel RudolphJan Daniel RudolphComputational Systems Biochemistry, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, GermanyMore by Jan Daniel Rudolph
- Jürgen Cox*Jürgen Cox*E-mail: [email protected]Computational Systems Biochemistry, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, GermanyDepartment of Biological and Medical Psychology, University of Bergen, Jonas Liesvei 91, 5009 Bergen, NorwayMore by Jürgen Cox
Abstract
Proteomics data analysis strongly benefits from not studying single proteins in isolation but taking their multivariate interdependence into account. We introduce PerseusNet, the new Perseus network module for the biological analysis of proteomics data. Proteomics is commonly used to generate networks, e.g., with affinity purification experiments, but networks are also used to explore proteomics data. PerseusNet supports the biomedical researcher for both modes of data analysis with a multitude of activities. For affinity purification, a volcano-plot-based statistical analysis method for network generation is featured which is scalable to large numbers of baits. For posttranslational modifications of proteins, such as phosphorylation, a collection of dedicated network analysis tools helps in elucidating cellular signaling events. Co-expression network analysis of proteomics data adopts established tools from transcriptome co-expression analysis. PerseusNet is extensible through a plugin architecture in a multi-lingual way, integrating analyses in C#, Python, and R, and is freely available at http://www.perseus-framework.org.
Introduction
Experimental Section
Creating Interaction Networks from Pulldown Experiments
Approximately Scale-Free Topology of the STRING Interaction Network
Network Analysis of a Phosphoproteomic Dataset of EGF Stimulation
Co-expression Analysis of a Clinical Proteomics Dataset
PluginInterop Provides a Central Entry Point for All External Plugins
Library Support for Scripting Languages
Implementation of PluginPHOTON
Implementation of PluginCoExpression
Implementation of KSEA in Perseus
Results and Discussion
Workflow-Based Biological Network Analysis
Multilingual Plugin Activities
Affinity Enrichment MS Interactomics
Importing, Curating, and Probing Large-Scale PPI Networks
Network Analysis of PTM Data
Co-expression Clustering and Clinical Data
Software Implementation, Download, and Maintenance
Conclusions
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jproteome.8b00927.
Figure S1, Graphical workflow combining matrix and network activities; Figure S2, Organization of the new Perseus plugin API for networks; Figure S3, Context-specific documentation; and explanations of Tables S1–S3 (PDF)
Table S1, AP-MS pull screen (TXT)
Table S2, phosphoproteomics of EGF stimulation (TXT)
Table S3, clinical proteomics dataset (TXT)
Supplementary Data 1, Perseus network collection data format: example of a network collection describing three small, randomly generated networks (ZIP)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank J. Sebastian Paez and Sung-Huan Yu for contributing to PerseusR, and Caroline Friedel and Tamar Geiger for helpful discussions. This project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement no. 686547 and from the FP7 grant agreement GA ERC-2012-SyG_318987–ToPAG.
References
This article references 49 other publications.
- 1Bar-Yam, Y. General Features of Complex Systems. Knowledge Management, Organizational Intelligence and Learning, and Complexity, Vol. I; Encyclopedia of Life Support Systems, 1997; www.eolss.netGoogle ScholarThere is no corresponding record for this reference.
- 2O’Connor, T.; Wong, H. Y. Emergent Properties. Stanford Encyclopedia of Philosophy, Spring 2012 ed.; Metaphysics Research Lab, Stanford University, 2012; https://plato.stanford.edu/archives/spr2012/entries/properties-emergent/Google ScholarThere is no corresponding record for this reference.
- 3Grandjean, M. A social network analysis of Twitter: Mapping the digital humanities community. Cogent Arts Humanit. 2016, 3, 1171458, DOI: 10.1080/23311983.2016.1171458Google ScholarThere is no corresponding record for this reference.
- 4Goffman, C. And What is Your Erdös Number?. Am. Math. Mon. 1969, 76, 791, DOI: 10.1080/00029890.1969.12000324Google ScholarThere is no corresponding record for this reference.
- 5Sporns, O.; Tononi, G.; Kötter, R. The human connectome: A structural description of the human brain. PLoS Comput. Biol. 2005, 1, e42, DOI: 10.1371/journal.pcbi.0010042Google ScholarThere is no corresponding record for this reference.
- 6Seung, S. I am my connectome. TED: Ideas Worth Spreading , 2010; https://www.ted.com/talks/sebastian_seung.Google ScholarThere is no corresponding record for this reference.
- 7Seung, H. S. Connectome: How the Brain’s Wiring Makes Us Who We Are; Penguin Books Ltd., 2013.Google ScholarThere is no corresponding record for this reference.
- 8Barabási, A.-L.; Oltvai, Z. N. Network biology: understanding the cell’s functional organization. Nat. Rev. Genet. 2004, 5, 101– 113, DOI: 10.1038/nrg1272Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXovV2mtg%253D%253D&md5=2ac9082dfaf88d56da1fdd75b9edddb4Network biology: Understanding the cell's functional organizationBarabasi, Albert-Laszlo; Oltvai, Zoltan N.Nature Reviews Genetics (2004), 5 (2), 101-113CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)A key aim of postgenomic biomedical research is to systematically catalog all mols. and their interactions within a living cell. There is a clear need to understand how these mols. and the interactions between them det. the function of this enormously complex machinery, both in isolation and when surrounded by other cells. Rapid advances in network biol. indicate that cellular networks are governed by universal laws and offer a new conceptual framework that could potentially revolutionize our view of biol. and disease pathologies in the twenty-first century.
- 9Butte, A. J.; Kohane, I. S. Mutual Information Relevance Networks: Functional Genomic Clustering Using Pairwise Entropy Measurements. Biocomputing 2000, 2000, 418– 429, DOI: 10.1142/9789814447331_0040Google ScholarThere is no corresponding record for this reference.
- 10Sinitcyn, P.; Rudolph, J. D.; Cox, J. Computational Methods for Understanding Mass Spectrometry-Based Shotgun Proteomics Data. Annu. Rev. Biomed. Data Sci. 2018, 1, 207– 234, DOI: 10.1146/annurev-biodatasci-080917-013516Google ScholarThere is no corresponding record for this reference.
- 11Tyanova, S. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 2016, 13, 731– 40, DOI: 10.1038/nmeth.3901Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVKntbnN&md5=f8c3e2876e4d724518054bb1a2d1e6eeThe Perseus computational platform for comprehensive analysis of (prote)omics dataTyanova, Stefka; Temu, Tikira; Sinitcyn, Pavel; Carlson, Arthur; Hein, Marco Y.; Geiger, Tamar; Mann, Matthias; Cox, JuergenNature Methods (2016), 13 (9), 731-740CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)A main bottleneck in proteomics is the downstream biol. anal. of highly multivariate quant. protein abundance data generated using mass-spectrometry-based anal. We developed the Perseus software platform (http://www.perseus-framework.org) to support biol. and biomedical researchers in interpreting protein quantification, interaction and post-translational modification data. Perseus contains a comprehensive portfolio of statistical tools for high-dimensional omics data anal. covering normalization, pattern recognition, time-series anal., cross-omics comparisons and multiple-hypothesis testing. A machine learning module supports the classification and validation of patient groups for diagnosis and prognosis, and it also detects predictive protein signatures. Central to Perseus is a user-friendly, interactive workflow environment that provides complete documentation of computational methods used in a publication. All activities in Perseus are realized as plugins, and users can extend the software by programming their own, which can be shared through a plugin store. We anticipate that Perseus's arsenal of algorithms and its intuitive usability will empower interdisciplinary anal. of complex large data sets.
- 12Tyanova, S.; Cox, J. Methods in Molecular Biology Springer, 2018; Vol. 1711, pp 133– 148.Google ScholarThere is no corresponding record for this reference.
- 13Shannon, P. Cytoscape: A software Environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498– 2504, DOI: 10.1101/gr.1239303Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXovFWrtr4%253D&md5=2bcbca9a3bd04717761f0424c0209e43Cytoscape: A software environment for integrated models of biomolecular interaction networksShannon, Paul; Markiel, Andrew; Ozier, Owen; Baliga, Nitin S.; Wang, Jonathan T.; Ramage, Daniel; Amin, Nada; Schwikowski, Benno; Ideker, TreyGenome Research (2003), 13 (11), 2498-2504CODEN: GEREFS; ISSN:1088-9051. (Cold Spring Harbor Laboratory Press)Cytoscape is an open source software project for integrating biomol. interaction networks with high-throughput expression data and other mol. states into a unified conceptual framework. Although applicable to any system of mol. components and interactions, Cytoscape is most powerful when used in conjunction with large databases of protein-protein, protein-DNA, and genetic interactions that are increasingly available for humans and model organisms. Cytoscape's software Core provides basic functionality to layout and query the network; to visually integrate the network with expression profiles, phenotypes, and other mol. states; and to link the network to databases of functional annotations. The Core is extensible through a straightforward plug-in architecture, allowing rapid development of addnl. computational analyses and features. Several case studies of Cytoscape plug-ins are surveyed, including a search for interaction pathways correlating with changes in gene expression, a study of protein complexes involved in cellular recovery to DNA damage, inference of a combined phys./functional interaction network for Halobacterium, and an interface to detailed stochastic/kinetic gene regulatory models.
- 14Kloet, S. L. The dynamic interactome and genomic targets of Polycomb complexes during stem-cell differentiation. Nat. Struct. Mol. Biol. 2016, 23, 682– 690, DOI: 10.1038/nsmb.3248Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsVCku7c%253D&md5=b5b35be17f21b312c593a16bedc27026The dynamic interactome and genomic targets of Polycomb complexes during stem-cell differentiationKloet, Susan L.; Makowski, Matthew M.; Baymaz, H. Irem; van Voorthuijsen, Lisa; Karemaker, Ino D.; Santanach, Alexandra; Jansen, Pascal W. T. C.; Di Croce, Luciano; Vermeulen, MichielNature Structural & Molecular Biology (2016), 23 (7), 682-690CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Although the core subunits of Polycomb group (PcG) complexes are well characterized, little is known about the dynamics of these protein complexes during cellular differentiation. We used quant. interaction proteomics and genome-wide profiling to study PcG proteins in mouse embryonic stem cells (ESCs) and neural progenitor cells (NPCs). We found that the stoichiometry and genome-wide binding of PRC1 and PRC2 were highly dynamic during neural differentiation. Intriguingly, we obsd. a downregulation and loss of PRC2 from chromatin marked with trimethylated histone H3 K27 (H3K27me3) during differentiation, whereas PRC1 was retained at these sites. Addnl., we found PRC1 at enhancer and promoter regions independently of PRC2 binding and H3K27me3. Finally, overexpression of NPC-specific PRC1 interactors in ESCs led to increased Ring1b binding to, and decreased expression of, NPC-enriched Ring1b-target genes. In summary, our integrative analyses uncovered dynamic PcG subcomplexes and their widespread colocalization with active chromatin marks during differentiation.
- 15Smith, C. L.; Blake, J. A.; Kadin, J. A.; Richardson, J. E.; Bult, C. J. Mouse Genome Database (MGD)-2018: Knowledgebase for the laboratory mouse. Nucleic Acids Res. 2018, 46, D836– 42, DOI: 10.1093/nar/gkx1006Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGit77M&md5=083422379b7c3bf55592a79bedb59056Mouse genome database (MGD)-2018: knowledge-base for the laboratory mouseSmith, Cynthia L.; Blake, Judith A.; Kadin, James A.; Richardson, Joel E.; Bult, Carol J.; Anagnostopoulos, A.; Andrews, A.; Baldarelli, R. M.; Beal, J. S.; Bello, S. M.; Blodgett, O.; Butler, N. E.; Christie, K.; Corbani, L. E.; Drabkin, H. J.; Espinoza, R.; Franco, J.; Giannatto, S. L.; Hale, P.; Hill, D. P.; Hutchins, L.; Law, M.; Lewis, J. R.; Mcandrews, M.; Mez, N.; Miers, D.; Motenko, H.; Ni, L.; Onda, H.; Perry, M.; Recla, J. M.; Reed, D. J.; Richards-Smith, B.; Sitnikov, D.; Tomczuk, M.; Wilming, L.; Zhu, Y.Nucleic Acids Research (2018), 46 (D1), D836-D842CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The Mouse Genome Database is the key community mouse database which supports basic, translational and computational research by providing integrated data on the genetics, genomics, and biol. of the lab. mouse. MGD serves as the source for biol. ref. data sets related to mouse genes, gene functions, phenotypes and disease models with an increasing emphasis on the assocn. of these data to human biol. and disease. We report here on recent enhancements to this resource, including improved access to mouse disease model and human phenotype data and enhanced relationships of mouse models to human disease.
- 16Langfelder, P.; Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinf. 2008, 9, 559, DOI: 10.1186/1471-2105-9-559Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M%252FpvFOitw%253D%253D&md5=8e22feffdc9e990fe5a4aeecebfe4a7eWGCNA: an R package for weighted correlation network analysisLangfelder Peter; Horvath SteveBMC bioinformatics (2008), 9 (), 559 ISSN:.BACKGROUND: Correlation networks are increasingly being used in bioinformatics applications. For example, weighted gene co-expression network analysis is a systems biology method for describing the correlation patterns among genes across microarray samples. Weighted correlation network analysis (WGCNA) can be used for finding clusters (modules) of highly correlated genes, for summarizing such clusters using the module eigengene or an intramodular hub gene, for relating modules to one another and to external sample traits (using eigengene network methodology), and for calculating module membership measures. Correlation networks facilitate network based gene screening methods that can be used to identify candidate biomarkers or therapeutic targets. These methods have been successfully applied in various biological contexts, e.g. cancer, mouse genetics, yeast genetics, and analysis of brain imaging data. While parts of the correlation network methodology have been described in separate publications, there is a need to provide a user-friendly, comprehensive, and consistent software implementation and an accompanying tutorial. RESULTS: The WGCNA R software package is a comprehensive collection of R functions for performing various aspects of weighted correlation network analysis. The package includes functions for network construction, module detection, gene selection, calculations of topological properties, data simulation, visualization, and interfacing with external software. Along with the R package we also present R software tutorials. While the methods development was motivated by gene expression data, the underlying data mining approach can be applied to a variety of different settings. CONCLUSION: The WGCNA package provides R functions for weighted correlation network analysis, e.g. co-expression network analysis of gene expression data. The R package along with its source code and additional material are freely available at http://www.genetics.ucla.edu/labs/horvath/CoexpressionNetwork/Rpackages/WGCNA.
- 17Rudolph, J. D.; de Graauw, M.; van de Water, B.; Geiger, T.; Sharan, R. Elucidation of Signaling Pathways from Large-Scale Phosphoproteomic Data Using Protein Interaction Networks. Cell Syst. 2016, 3, 585– 593, DOI: 10.1016/j.cels.2016.11.005Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFamu78%253D&md5=4bc19764bdb6820543b579e86d00df0dElucidation of Signaling Pathways from Large-Scale Phosphoproteomic Data Using Protein Interaction NetworksRudolph, Jan Daniel; de Graauw, Marjo; van de Water, Bob; Geiger, Tamar; Sharan, RodedCell Systems (2016), 3 (6), 585-593.e3CODEN: CSEYA4; ISSN:2405-4712. (Cell Press)Phosphoproteomic expts. typically identify sites within a protein that are differentially phosphorylated between two or more cell states. However, the interpretation of these data is hampered by the lack of methods that can translate site-specific information into global maps of active proteins and signaling networks, esp. as the phosphoproteome is often undersampled. Here, we describe PHOTON, a method for interpreting phosphorylation data within their signaling context, as captured by protein-protein interaction networks, to identify active proteins and pathways and pinpoint functional phosphosites. We apply PHOTON to interpret existing and novel phosphoproteomic datasets related to epidermal growth factor and insulin responses. PHOTON substantially outperforms the widely used cutoff approach, providing highly reproducible predictions that are more in line with current biol. knowledge. Altogether, PHOTON overcomes the fundamental challenge of delineating signaling pathways from large-scale phosphoproteomic data, thereby enabling translation of environmental cues to downstream cellular responses.
- 18Hornbeck, P. V. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 2015, 43, D512– 20, DOI: 10.1093/nar/gku1267Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVymt7rO&md5=df6fdbeb9f2e8cf025f1dea5179220afPhosphoSitePlus, 2014: mutations, PTMs and recalibrationsHornbeck, Peter V.; Zhang, Bin; Murray, Beth; Kornhauser, Jon M.; Latham, Vaughan; Skrzypek, ElzbietaNucleic Acids Research (2015), 43 (D1), D512-D520CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)PhosphoSitePlus (PSP), a knowledge-base dedicated to mammalian post-translational modifications (PTMs), contains over 330000 non-redundant PTMs, including phospho, acetyl, ubiquityl and Me groups. Over 95% of the sites are from mass spectrometry (MS) expts. In order to improve data reliability, early MS data have been reanalyzed, applying a common std. of anal. across over 1000000 spectra. Site assignments with P > 0.05 were filtered out. Two new downloads are available from PSP. The 'Regulatory sites' dataset includes curated information about modification sites that regulate downstream cellular processes, mol. functions and protein-protein interactions. The 'PTMVar' dataset, an intersect of missense mutations and PTMs from PSP, identifies over 25000 PTMVars (PTMs Impacted by Variants) that can rewire signaling pathways. The PTMVar data include missense mutations from UniPROTKB, TCGA and other sources that cause over 2000 diseases or syndromes (MIM) and polymorphisms, or are assocd. with hundreds of cancers. PTMVars include 18548 phosphorlyation sites, 3412 ubiquitylation sites, 2316 acetylation sites, 685 methylation sites and 245 succinylation sites.
- 19Yanovich, G. Clinical Proteomics of Breast Cancer Reveals a Novel Layer of Breast Cancer Classification. Cancer Res. 2018, 78, 6001– 6010, DOI: 10.1158/0008-5472.CAN-18-1079Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGhtbzL&md5=0a4b0f1f9fbd03b93e51335d002b7c12Clinical proteomics of breast cancer reveals a novel layer of breast cancer classificationYanovich, Gali; Agmon, Hadar; Harel, Michal; Sonnenblick, Amir; Peretz, Tamar; Geiger, TamarCancer Research (2018), 78 (20), 6001-6010CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Breast cancer classification has been the focus of numerous worldwide efforts, analyzing the mol. basis of breast cancer subtypes and aiming to assoc. them with clin. outcome and to improve the current diagnostic routine. Genomic and transcriptomic profiles of breast cancer have been well established, however the proteomic contribution to these profiles has yet to be elucidated. In this work, we utilized mass spectrometry-based proteomic anal. on more than 130 clin. breast samples to demonstrate intertumor heterogeneity across three breast cancer subtypes and healthy tissue. Unsupervised anal. identified four proteomic clusters, among them, one that represents a novel luminal subtype characterized by increased PI3K signaling. This subtype was further validated using an independent protein-based dataset, but not in two independent transcriptome cohorts. These results demonstrate the importance of deep proteomic anal., which may affect cancer treatment decision making.
- 20Szklarczyk, D. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362– D368, DOI: 10.1093/nar/gkw937Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWhs70%253D&md5=cef71d32dbd02a49839447f168683d39The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessibleSzklarczyk, Damian; Morris, John H.; Cook, Helen; Kuhn, Michael; Wyder, Stefan; Simonovic, Milan; Santos, Alberto; Doncheva, Nadezhda T.; Roth, Alexander; Bork, Peer; Jensen, Lars J.; von Mering, ChristianNucleic Acids Research (2017), 45 (D1), D362-D368CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A system-wide understanding of cellular function requires knowledge of all functional interactions between the expressed proteins. The STRING database aims to collect and integrate this information, by consolidating known and predicted protein-protein assocn. data for a large no. of organisms. The assocns. in STRING include direct (phys.) interactions, as well as indirect (functional) interactions, as long as both are specific and biol. meaningful. Apart from collecting and reassessing available exptl. data on protein-protein interactions, and importing known pathways and protein complexes from curated databases, interaction predictions are derived from the following sources: (i) systematic coexpression anal., (ii) detection of shared selective signals across genomes, (iii) automated text-mining of the scientific literature and (iv) computational transfer of interaction knowledge between organisms based on gene orthol. In the latest version 10.5 of STRING, the biggest changes are concerned with data dissemination: the web front-end has been completely redesigned to reduce dependency on outdated browser technologies, and the database can now also be queried from inside the popular Cytoscape software framework. Further improvements include automated background anal. of user inputs for functional enrichments, and streamlined download options. The STRING resource is available online, at http://string-db.org/.
- 21Chatr-Aryamontri, A. The BioGRID interaction database: 2017 update. Nucleic Acids Res. 2017, 45, D369– D379, DOI: 10.1093/nar/gkw1102Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWhsrk%253D&md5=96ab82537dc1a8e03f383f8ad0a78298The BioGRID interaction database: 2017 updateChatr-aryamontri, Andrew; Oughtred, Rose; Boucher, Lorrie; Rust, Jennifer; Chang, Christie; Kolas, Nadine K.; O'Donnell, Lara; Oster, Sara; Theesfeld, Chandra; Sellam, Adnane; Stark, Chris; Breitkreutz, Bobby-Joe; Dolinski, Kara; Tyers, MikeNucleic Acids Research (2017), 45 (D1), D369-D379CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The Biol. General Repository for Interaction Datasets (BioGRID: https://thebiogrid.org) is an open access database dedicated to the annotation and archival of protein, genetic and chem. interactions for all major model organism species and humans. As of Sept. 2016 (build 3.4.140), the BioGRID contains 1 072 173 genetic and protein interactions, and 38 559 post-translational modifications, as manually annotated from 48 114 publications. This dataset represents interaction records for 66 model organisms and represents a 30% increase compared to the previous 2015 BioGRID update. BioGRID curates the biomedical literature for major model organism species, including humans, with a recent emphasis on central biol. processes and specific human diseases. To facilitate network-based approaches to drug discovery, BioGRID now incorporates 27 501 chem.-protein interactions for human drug targets, as drawn from the DrugBank database. A new dynamic interaction network viewer allows the easy navigation and filtering of all genetic and protein interaction data, as well as for bioactive compds. and their established targets. BioGRID data are directly downloadable without restriction in a variety of standardized formats and are freely distributed through partner model organism databases and meta-databases.
- 22Orchard, S. The MIntAct project - IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Res. 2014, 42, D358– 63, DOI: 10.1093/nar/gkt1115Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXoslKg&md5=f2964108748995acc8bc98b67880c95aThe MIntAct project-IntAct as a common curation platform for 11 molecular interaction databasesOrchard, Sandra; Ammari, Mais; Aranda, Bruno; Breuza, Lionel; Briganti, Leonardo; Broackes-Carter, Fiona; Campbell, Nancy H.; Chavali, Gayatri; Chen, Carol; del-Toro, Noemi; Duesbury, Margaret; Dumousseau, Marine; Galeota, Eugenia; Hinz, Ursula; Iannuccelli, Marta; Jagannathan, Sruthi; Jimenez, Rafael; Khadake, Jyoti; Lagreid, Astrid; Licata, Luana; Lovering, Ruth C.; Meldal, Birgit; Melidoni, Anna N.; Milagros, Mila; Peluso, Daniele; Perfetto, Livia; Porras, Pablo; Raghunath, Arathi; Ricard-Blum, Sylvie; Roechert, Bernd; Stutz, Andre; Tognolli, Michael; van Roey, Kim; Cesareni, Gianni; Hermjakob, HenningNucleic Acids Research (2014), 42 (D1), D358-D363CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)IntAct (freely available at http://www.ebi.ac.uk/intact) is an open-source, open data mol. interaction database populated by data either curated from the literature or from direct data depositions. IntAct has developed a sophisticated web-based curation tool, capable of supporting both IMEx- and MIMIx-level curation. This tool is now utilized by multiple addnl. curation teams, all of whom annotate data directly into the IntAct database. Members of the IntAct team supply appropriate levels of training, perform quality control on entries and take responsibility for long-term data maintenance. Recently, the MINT and IntAct databases decided to merge their sep. efforts to make optimal use of limited developer resources and maximize the curation output. All data manually curated by the MINT curators have been moved into the IntAct database at EMBL-EBI and are merged with the existing IntAct dataset. Both IntAct and MINT are active contributors to the IMEx consortium (http://www.imexconsortium.org).
- 23Ruepp, A. CORUM: The comprehensive resource of mammalian protein complexes-2009. Nucleic Acids Res. 2010, 38, D497– 501, DOI: 10.1093/nar/gkp914Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXktlynug%253D%253D&md5=8803df58fa6353157f8a058f036d9145CORUM: the comprehensive resource of mammalian protein complexes-2009Ruepp, Andreas; Waegele, Brigitte; Lechner, Martin; Brauner, Barbara; Dunger-Kaltenbach, Irmtraud; Fobo, Gisela; Frishman, Goar; Montrone, Corinna; Mewes, H.-WernerNucleic Acids Research (2010), 38 (Database Iss), D497-D501CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)CORUM is a database that provides a manually curated repository of exptl. characterized protein complexes from mammalian organisms, mainly human (64%), mouse (16%) and rat (12%). Protein complexes are key mol. entities that integrate multiple gene products to perform cellular functions. The new CORUM 2.0 release encompasses 2837 protein complexes offering the largest and most comprehensive publicly available dataset of mammalian protein complexes. The CORUM dataset is built from 3198 different genes, representing ∼16% of the protein coding genes in humans. Each protein complex is described by a protein complex name, subunit compn., function as well as the literature ref. that characterizes the resp. protein complex. Recent developments include mapping of functional annotation to Gene Ontol. terms as well as cross-refs. to Entrez Gene identifiers. In addn., a Phylogenetic Conservation' anal. tool was implemented that analyses the potential occurrence of orthologous protein complex subunits in mammals and other selected groups of organisms. This allows one to predict the occurrence of protein complexes in different phylogenetic groups. CORUM is freely accessible at (http://mips.helmholtz-muenchen.de/genre/proj/corum/index.html).
- 24Gingras, A. C.; Gstaiger, M.; Raught, B.; Aebersold, R. Analysis of protein complexes using mass spectrometry. Nat. Rev. Mol. Cell Biol. 2007, 8, 645– 654, DOI: 10.1038/nrm2208Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXotVahtro%253D&md5=0f6219bd2475259e258ea56238a8c6dcAnalysis of protein complexes using mass spectrometryGingras, Anne-Claude; Gstaiger, Matthias; Raught, Brian; Aebersold, RuediNature Reviews Molecular Cell Biology (2007), 8 (8), 645-654CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. The versatile combination of affinity purifn. and mass spectrometry (AP-MS) has recently been applied to the detailed characterization of many protein complexes and large protein-interaction networks. The combination of AP-MS with other techniques, such as biochem. fractionation, intact mass measurement and chem. crosslinking, can help to decipher the supramol. organization of protein complexes. AP-MS can also be combined with quant. proteomics approaches to better understand the dynamics of protein-complex assembly.
- 25Dunham, W. H.; Mullin, M.; Gingras, A. C. Affinity-purification coupled to mass spectrometry: Basic principles and strategies. Proteomics 2012, 12, 1576, DOI: 10.1002/pmic.201100523Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xoslehur0%253D&md5=602ff89e9f3496a7934f4f52a3abc1e5Affinity-purification coupled to mass spectrometry: Basic principles and strategiesDunham, Wade H.; Mullin, Michael; Gingras, Anne-ClaudeProteomics (2012), 12 (10), 1576-1590CODEN: PROTC7; ISSN:1615-9853. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Identifying the interactions established by a protein of interest can be a crit. step in understanding its function. This is esp. true when an unknown protein of interest is demonstrated to phys. interact with proteins of known function. While many techniques have been developed to characterize protein-protein interactions, one strategy that has gained considerable momentum over the past decade for identification and quantification of protein-protein interactions, is affinity-purifn. followed by mass spectrometry (AP-MS). Here, we briefly review the basic principles used in affinity-purifn. coupled to mass spectrometry, with an emphasis on tools (both biochem. and computational), which enable the discovery and reporting of high quality protein-protein interactions.
- 26Hein, M. Y. A Human Interactome in Three Quantitative Dimensions Organized by Stoichiometries and Abundances. Cell 2015, 163, 712– 723, DOI: 10.1016/j.cell.2015.09.053Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslWrsLnK&md5=599f46801c2006cbc7b26d6ff8a00d6dA Human Interactome in Three Quantitative Dimensions Organized by Stoichiometries and AbundancesHein, Marco Y.; Hubner, Nina C.; Poser, Ina; Cox, Juergen; Nagaraj, Nagarjuna; Toyoda, Yusuke; Gak, Igor A.; Weisswange, Ina; Mansfeld, Joerg; Buchholz, Frank; Hyman, Anthony A.; Mann, MatthiasCell (Cambridge, MA, United States) (2015), 163 (3), 712-723CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The organization of a cell emerges from the interactions in protein networks. The interactome is critically dependent on the strengths of interactions and the cellular abundances of the connected proteins, both of which span orders of magnitude. However, these aspects have not yet been analyzed globally. Here, the authors have generated a library of HeLa cell lines expressing 1125 GFP-tagged proteins under near-endogenous control, which the authors used as input for a next-generation interaction survey. Using quant. proteomics, the authors detect specific interactions, est. interaction stoichiometries, and measure cellular abundances of interacting proteins. These three quant. dimensions reveal that the protein network is dominated by weak, substoichiometric interactions that play a pivotal role in defining network topol. The minority of stable complexes can be identified by their unique stoichiometry signature. This study provides a rich interaction dataset connecting thousands of proteins and introduces a framework for quant. network anal.
- 27Huttlin, E. L. The BioPlex Network: A Systematic Exploration of the Human Interactome. Cell 2015, 162, 425– 440, DOI: 10.1016/j.cell.2015.06.043Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1KgtL3I&md5=79b8d96037646f6679baab3b966b9d47The BioPlex Network: A Systematic Exploration of the Human InteractomeHuttlin, Edward L.; Ting, Lily; Bruckner, Raphael J.; Gebreab, Fana; Gygi, Melanie P.; Szpyt, John; Tam, Stanley; Zarraga, Gabriela; Colby, Greg; Baltier, Kurt; Dong, Rui; Guarani, Virginia; Vaites, Laura Pontano; Ordureau, Alban; Rad, Ramin; Erickson, Brian K.; Wuhr, Martin; Chick, Joel; Zhai, Bo; Kolippakkam, Deepak; Mintseris, Julian; Obar, Robert A.; Harris, Tim; Artavanis-Tsakonas, Spyros; Sowa, Mathew E.; De Camilli, Pietro; Paulo, Joao A.; Harper, J. Wade; Gygi, Steven P.Cell (Cambridge, MA, United States) (2015), 162 (2), 425-440CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Protein interactions form a network whose structure drives cellular function and whose organization informs biol. inquiry. Using high-throughput affinity-purifn. mass spectrometry, the authors identify interacting partners for 2594 human proteins in HEK293T cells. The resulting network (BioPlex) contains 23,744 interactions among 7668 proteins with 86% previously undocumented. BioPlex accurately depicts known complexes, attaining 80%-100% coverage for most CORUM complexes. The network readily subdivides into communities that correspond to complexes or clusters of functionally related proteins. More generally, network architecture reflects cellular localization, biol. process, and mol. function, enabling functional characterization of thousands of proteins. Network structure also reveals assocns. among thousands of protein domains, suggesting a basis for examg. structurally related proteins. Finally, BioPlex, in combination with other approaches, can be used to reveal interactions of biol. or clin. significance. For example, mutations in the membrane protein VAPB implicated in familial amyotrophic lateral sclerosis perturb a defined community of interactors.
- 28Hubner, N. C. Quantitative proteomics combined with BAC TransgeneOmics reveals in vivo protein interactions. J. Cell Biol. 2010, 189, 739– 754, DOI: 10.1083/jcb.200911091Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmslKktrg%253D&md5=4cf589eedd456fb8a9b141ef0d5bc4f4Quantitative proteomics combined with BAC TransgeneOmics reveals in vivo protein interactionsHubner, Nina C.; Bird, Alexander W.; Cox, Juergen; Splettstoesser, Bianca; Bandilla, Peter; Poser, Ina; Hyman, Anthony; Mann, MatthiasJournal of Cell Biology (2010), 189 (4), 739-754CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)Protein interactions are involved in all cellular processes. Their efficient and reliable characterization is therefore essential for understanding biol. mechanisms. In this study, we show that combining bacterial artificial chromosome (BAC) TransgeneOmics with quant. interaction proteomics, which we call quant. BAC-green fluorescent protein interactomics (QUBIC), allows specific and highly sensitive detection of interactions using rapid, generic, and quant. procedures with minimal material. We applied this approach to identify known and novel components of well-studied complexes such as the anaphase-promoting complex. Furthermore, we demonstrate second generation interaction proteomics by incorporating directed mutational transgene modification and drug perturbation into QUBIC. These methods identified domain/isoform-specific interactors of pericentrin- and phosphorylation-specific interactors of TACC3, which are necessary for its recruitment to mitotic spindles. The scalability, simplicity, cost effectiveness, and sensitivity of this method provide a basis for its general use in small-scale expts. and in mapping the human protein interactome.
- 29Noble, W. S. How does multiple testing correction work?. Nat. Biotechnol. 2009, 27, 1135– 1137, DOI: 10.1038/nbt1209-1135Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFemtr3N&md5=7376f0b000468adffe6e6c681ef84c95How does multiple testing correction work?Noble, William S.Nature Biotechnology (2009), 27 (12), 1135-1137CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. When prioritizing hits from a high-throughput expt., it is important to correct for random events that falsely appear significant. How is this done and what methods should be used. Techniques for doing this are discussed here.
- 30Tusher, V. G.; Tibshirani, R.; Chu, G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 5116– 5121, DOI: 10.1073/pnas.091062498Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXjt1Ons7w%253D&md5=819240102af66293c77046878e4ca3c3Significance analysis of microarrays applied to the ionizing radiation responseTusher, Virginia Goss; Tibshirani, Robert; Chu, GilbertProceedings of the National Academy of Sciences of the United States of America (2001), 98 (9), 5116-5121CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Microarrays can measure the expression of thousands of genes to identify changes in expression between different biol. states. Methods are needed to det. the significance of these changes while accounting for the enormous no. of genes. We describe a method, Significance Anal. of Microarrays (SAM), that assigns a score to each gene on the basis of change in gene expression relative to the std. deviation of repeated measurements. For genes with scores greater than an adjustable threshold, SAM uses permutations of the repeated measurements to est. the percentage of genes identified by chance, the false discovery rate (FDR). When the transcriptional response of human cells to ionizing radiation was measured by microarrays, SAM identified 34 genes that changed at least 1.5-fold with an estd. FDR of 12%, compared with FDRs of 60 and 84% by using conventional methods of anal. Of the 34 genes, 19 were involved in cell cycle regulation and 3 in apoptosis. Surprisingly, four nucleotide excision repair genes were induced, suggesting that this repair pathway for UV-damaged DNA might play a previously unrecognized role in repairing DNA damaged by ionizing radiation.
- 31Franz, M. Cytoscape.js: A graph theory library for visualisation and analysis. Bioinformatics 2015, 32, 309– 311, DOI: 10.1093/bioinformatics/btv557Google ScholarThere is no corresponding record for this reference.
- 32Dogrusoz, U.; Giral, E.; Cetintas, A.; Civril, A.; Demir, E. A layout algorithm for undirected compound graphs. Inf. Sci. (N. Y.) 2009, 179, 980– 994, DOI: 10.1016/j.ins.2008.11.017Google ScholarThere is no corresponding record for this reference.
- 33Pedamallu, C. S.; Ozdamar, L. A Review on protein-protein interaction network databases. In Springer Proceedings in Mathematics and Statistics; Springer, 2014; Vol. 73, pp 511– 519.Google ScholarThere is no corresponding record for this reference.
- 34Alanis-Lobato, G.; Andrade-Navarro, M. A.; Schaefer, M. H. HIPPIE v2.0: Enhancing meaningfulness and reliability of protein-protein interaction networks. Nucleic Acids Res. 2017, 45, D408– 14, DOI: 10.1093/nar/gkw985Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWht74%253D&md5=400725d91e68e757a112622623fb5e63HIPPIE v2.0: enhancing meaningfulness and reliability of protein-protein interaction networksAlanis-Lobato, Gregorio; Andrade-Navarro, Miguel A.; Schaefer, Martin H.Nucleic Acids Research (2017), 45 (D1), D408-D414CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The increasing no. of exptl. detected interactions between proteins makes it difficult for researchers to ext. the interactions relevant for specific biol. processes or diseases. This makes it necessary to accompany the large-scale detection of protein-protein interactions (PPIs) with strategies and tools to generate meaningful PPI subnetworks. To this end, we generated the Human Integrated Protein-Protein Interaction ref. or HIPPIE (http://cbdm.uni-mainz.de/hippie/). HIPPIE is a one-stop resource for the generation and interpretation of PPI networks relevant to a specific research question. We provide means to generate highly reliable, context-specific PPI networks and to make sense out of them. We just released the second major update of HIPPIE, implementing various new features. HIPPIE grew substantially over the last years and now contains more than 270 000 confidence scored and annotated PPIs. We integrated different types of exptl. information for the confidence scoring and the construction of context-specific networks. We implemented basic graph algorithms that highlight important proteins and interactions. HIPPIE's graphical interface implements several ways for wet lab and computational scientists alike to access the PPI data.
- 35Gene Ontology Consortium, C. Gene Ontology Consortium: going forward. Nucleic Acids Res. 2015, 43, D1049– 56, DOI: 10.1093/nar/gku1179Google ScholarThere is no corresponding record for this reference.
- 36Clauset, A.; Shalizi, C. R.; Newman, M. E. J. Power-law distributions in empirical data. SIAM Rev. 2009, 51, 661, DOI: 10.1137/070710111Google ScholarThere is no corresponding record for this reference.
- 37Albert, R. Scale-free networks in cell biology. J. Cell Sci. 2005, 118, 4947, DOI: 10.1242/jcs.02714Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1ynsbjI&md5=966ea6581f396e2515be7c229748fbf9Scale-free networks in cell biologyAlbert, RekaJournal of Cell Science (2005), 118 (21), 4947-4957CODEN: JNCSAI; ISSN:0021-9533. (Company of Biologists Ltd.)A review. A cell's behavior is a consequence of the complex interactions between its numerous constituents, such as DNA, RNA, proteins and small mols. Cells use signaling pathways and regulatory mechanisms to coordinate multiple processes, allowing them to respond to and adapt to an ever-changing environment. The large no. of components, the degree of interconnectivity and the complex control of cellular networks are becoming evident in the integrated genomic and proteomic analyses that are emerging. It is increasingly recognized that the understanding of properties that arise from whole-cell function require integrated, theor. descriptions of the relationships between different cellular components. Recent theor. advances allow us to describe cellular network structure with graph concepts and have revealed organizational features shared with numerous non-biol. networks. We now have the opportunity to describe quant. a network of hundreds or thousands of interacting components. Moreover, the obsd. topologies of cellular networks give us clues about their evolution and how their organization influences their function and dynamic responses.
- 38Riley, N. M.; Coon, J. J. Phosphoproteomics in the Age of Rapid and Deep Proteome Profiling. Anal. Chem. 2016, 88, 74– 94, DOI: 10.1021/acs.analchem.5b04123Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslygur%252FE&md5=5b91915e258ce17207c3d433adae955bPhosphoproteomics in the Age of Rapid and Deep Proteome ProfilingRiley, Nicholas M.; Coon, Joshua J.Analytical Chemistry (Washington, DC, United States) (2016), 88 (1), 74-94CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A review. Protein phosphorylation is a post-translational modification (PTM) that orchestrates a diverse array of cellular processes. Because this modification serves as a rapid and reversible means to modulate protein activity and transduce signals, the regulation of phosphorylation is a central mechanism in cell health and disease. The addn. and removal of phosphoryl modifications via kinases and phosphates, resp., makes the landscape of phosphorylation particularly dynamic. Understanding the complex networks and functions coordinated by phosphorylation requires knowledge of specific amino acid modifications with both spatial and temporal resoln. - a task that remains a challenging anal. endeavor. Mass spectrometry (MS) has emerged as the premier tool for global PTM anal., boasting high sensitivity, considerable throughput, and the capacity to localize modifications to a single residue. Indeed, MS-centric phosphoproteomics has become a std. approach for investigating protein phosphorylation in labs. worldwide. Anal. Chem. last reviewed the contribution of MS and related technologies to phosphoproteomics in 2011. Since that time, MS methodol. has developed at an impressive pace. While routine proteomic expts. can now analyze thousands of proteins in just a few hours, rather than days or weeks, characterizing the global phosphoproteome is significantly more challenging than measuring non-modified proteins. The relative low abundance of phosphorylated peptides and the need for residue-specific information require special considerations in sample handling, data acquisition, and post-acquisition processing that constrain reproducibility, quant. efficacy, throughput, and depth in phosphoproteomic workflows. Advances in MS-based approaches have remarkably improved our abilities to investigate the many roles of protein phosphorylation across a diverse set of biol. contexts, but many tech. obstacles still exist. Poor run-to-run overlap, challenges in confident phosphosite assignment, and complications inherent to various quant. strategies limit biol. insight, despite ever-increasing nos. of detected phosphopeptides. Focusing on work from the past two years (2013-2015), this review examines major developments in MS technol. that have enabled the characterization of tens of thousands of phosphopeptides in a given expt., and considers the contribution of this anal. power to translational research. We also discuss how future innovation can address tech. challenges of today's methods, and we offer our perspective on how phosphoproteomics will continue to mature.
- 39Casado, P. Kinase-substrate enrichment analysis provides insights into the heterogeneity of signaling pathway activation in leukemia cells. Sci. Signaling 2013, 6, rs6– rs6, DOI: 10.1126/scisignal.2003573Google ScholarThere is no corresponding record for this reference.
- 40Hernandez-Armenta, C.; Ochoa, D.; Gonçalves, E.; Saez-Rodriguez, J.; Beltrao, P. Benchmarking substrate-based kinase activity inference using phosphoproteomic data. Bioinformatics 2017, 33, 1845– 1851, DOI: 10.1093/bioinformatics/btx082Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFagsbfI&md5=dafb0673d76db1772c48306366ba37c0Benchmarking substrate-based kinase activity inference using phosphoproteomic dataHernandez-Armenta, Claudia; Ochoa, David; Goncalves, Emanuel; Saez-Rodriguez, Julio; Beltrao, PedroBioinformatics (2017), 33 (12), 1845-1851CODEN: BOINFP; ISSN:1460-2059. (Oxford University Press)Motivation: Phosphoproteomic expts. are increasingly used to study the changes in signaling occurring across different conditions. It has been proposed that changes in phosphorylation of kinase target sites can be used to infer when a kinase activity is under regulation. However, these approaches have not yet been benchmarked due to a lack of appropriate benchmarking strategies. Results: We used curated phosphoproteomic expts. and a gold std. dataset contg. a total of 184 kinase-condition pairs where regulation is expected to occur to benchmark and compare different kinase activity inference strategies: Z-test, Kolmogorov Smirnov test, Wilcoxon rank sum test, gene set enrichment anal. (GSEA), and a multiple linear regression model. We also tested weighted variants of the Z-test and GSEA that include information on kinase sequence specificity as proxy for affinity. Finally, we tested how the no. of known substrates and the type of evidence (in vivo, in vitro or in silico) supporting these influence the predictions. Conclusions: Most models performed well with the Z-test and the GSEA performing best as detd. by the area under the ROC curve (Mean AUC 1/4 0.722). Weighting kinase targets by the kinase target sequence preference improves the results marginally. However, the no. of known substrates and the evidence supporting the interactions has a strong effect on the predictions.
- 41Herranz, N. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 2015, 17, 1205, DOI: 10.1038/ncb3225Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlCksbzL&md5=18eaa8c3ea3ee236774ef90e489535bcmTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotypeHerranz, Nicolas; Gallage, Suchira; Mellone, Massimiliano; Wuestefeld, Torsten; Klotz, Sabrina; Hanley, Christopher J.; Raguz, Selina; Acosta, Juan Carlos; Innes, Andrew J.; Banito, Ana; Georgilis, Athena; Montoya, Alex; Wolter, Katharina; Dharmalingam, Gopuraja; Faull, Peter; Carroll, Thomas; Martinez-Barbera, Juan Pedro; Cutillas, Pedro; Reisinger, Florian; Heikenwalder, Mathias; Miller, Richard A.; Withers, Dominic; Zender, Lars; Thomas, Gareth J.; Gil, JesusNature Cell Biology (2015), 17 (9), 1205-1217CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)Senescent cells secrete a combination of factors collectively known as the senescence-assocd. secretory phenotype (SASP). The SASP reinforces senescence and activates an immune surveillance response, but it can also show pro-tumorigenic properties and contribute to age-related pathologies. In a drug screen to find new SASP regulators, we uncovered the mTOR inhibitor rapamycin as a potent SASP suppressor. Here we report a mechanism by which mTOR controls the SASP by differentially regulating the translation of the MK2 (also known as MAPKAPK2) kinase through 4EBP1. In turn, MAPKAPK2 phosphorylates the RNA-binding protein ZFP36L1 during senescence, inhibiting its ability to degrade the transcripts of numerous SASP components. Consequently, mTOR inhibition or constitutive activation of ZFP36L1 impairs the non-cell-autonomous effects of senescent cells in both tumor-suppressive and tumor-promoting contexts. Altogether, our results place regulation of the SASP as a key mechanism by which mTOR could influence cancer, age-related diseases and immune responses.
- 42Wilkes, E. H.; Terfve, C.; Gribben, J. G.; Saez-Rodriguez, J.; Cutillas, P. R. Empirical inference of circuitry and plasticity in a kinase signaling network. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 7719, DOI: 10.1073/pnas.1423344112Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXpslyjsLw%253D&md5=e144af280676314d91f41d0b06f79a3dEmpirical inference of circuitry and plasticity in a kinase signaling networkWilkes, Edmund H.; Terfve, Camille; Gribben, John G.; Saez-Rodriguez, Julio; Cutillas, Pedro RodriguezProceedings of the National Academy of Sciences of the United States of America (2015), 112 (25), 7719-7724CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Our understanding of physiol. and disease is hampered by the difficulty of measuring the circuitry and plasticity of signaling networks that regulate cell biol., and how these relate to phenotypes. Here, using mass spectrometry-based phosphoproteomics, we systematically characterized the topol. of a network comprising the PI3K/Akt/mTOR and MEK/ERK signaling axes and confirmed its biol. relevance by assessing its dynamics upon EGF and IGF1 stimulation. Measuring the activity of this network in models of acquired drug resistance revealed that cells chronically treated with PI3K or mTORC1/2 inhibitors differed in the way their networks were remodeled. Unexpectedly, we also obsd. a degree of heterogeneity in the network state between cells resistant to the same inhibitor, indicating that even identical and carefully controlled exptl. conditions can give rise to the evolution of distinct kinase network statuses. These data suggest that the initial conditions of the system do not necessarily det. the mechanism by which cancer cells become resistant to PI3K/mTOR targeted therapies. The patterns of signaling network activity obsd. in the resistant cells mirrored the patterns of response to several drug combination treatments, suggesting that the activity of the defined signaling network truly reflected the evolved phenotypic diversity.
- 43Linding, R. NetworKIN: A resource for exploring cellular phosphorylation networks. Nucleic Acids Res. 2007, 36, D695– 99, DOI: 10.1093/nar/gkm902Google ScholarThere is no corresponding record for this reference.
- 44Wiredja, D. D.; Koyutürk, M.; Chance, M. R. The KSEA App: a web-based tool for kinase activity inference from quantitative phosphoproteomics. Bioinformatics 2017, 33, 3489, DOI: 10.1093/bioinformatics/btx415Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFWmsbnO&md5=7500765789d1dd6304968f51b89958bcThe KSEA app: A web-based tool for kinase activity inference from quantitative phosphoproteomicsWiredja, Danica D.; Koyuturk, Mehmet; Chance, Mark R.Bioinformatics (2017), 33 (21), 3489-3491CODEN: BOINFP; ISSN:1367-4811. (Oxford University Press)Summary: Computational characterization of differential kinase activity from phosphoproteomics datasets is crit. for correctly inferring cellular circuitry and how signaling cascades are altered in drug treatment and/or disease. Kinase-Substrate Enrichment Anal. (KSEA) offers a powerful approach to estg. changes in a kinase's activity based on the collective phosphorylation changes of its identified substrates. However, KSEA has been limited to programmers who are able to implement the algorithms. Thus, to make it accessible to the larger scientific community, we present a web-based application of this method: the KSEA App. Overall, we expect that this tool will offer a quick and user-friendly way of generating kinase activity ests. from high-throughput phosphoproteomics datasets.
- 45Zhang, B.; Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 2005, 4, 17, DOI: 10.2202/1544-6115.1128Google ScholarThere is no corresponding record for this reference.
- 46Cox, J.; Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 2008, 26, 1367– 1372, DOI: 10.1038/nbt.1511Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVWjtLzJ&md5=675d31ca84e3a7e4fb9bdd601d8075eaMaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantificationCox, Juergen; Mann, MatthiasNature Biotechnology (2008), 26 (12), 1367-1372CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Efficient anal. of very large amts. of raw data for peptide identification and protein quantification is a principal challenge in mass spectrometry (MS)-based proteomics. Here we describe MaxQuant, an integrated suite of algorithms specifically developed for high-resoln., quant. MS data. Using correlation anal. and graph theory, MaxQuant detects peaks, isotope clusters and stable amino acid isotope-labeled (SILAC) peptide pairs as three-dimensional objects in m/z, elution time and signal intensity space. By integrating multiple mass measurements and correcting for linear and nonlinear mass offsets, we achieve mass accuracy in the p.p.b. range, a sixfold increase over std. techniques. We increase the proportion of identified fragmentation spectra to 73% for SILAC peptide pairs via unambiguous assignment of isotope and missed-cleavage state and individual mass precision. MaxQuant automatically quantifies several hundred thousand peptides per SILAC-proteome expt. and allows statistically robust identification and quantification of >4000 proteins in mammalian cell lysates.
- 47Sinitcyn, P. MaxQuant goes Linux. Nat. Methods 2018, 15, 401, DOI: 10.1038/s41592-018-0018-yGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFOmtL%252FI&md5=d1aaed94810a18c96d0da5de8a9714d4MaxQuant goes LinuxSinitcyn, Pavel; Tiwary, Shivani; Rudolph, Jan; Gutenbrunner, Petra; Wichmann, Christoph; Yilmaz, Sule; Hamzeiy, Hamid; Salinas, Favio; Cox, JuergenNature Methods (2018), 15 (6), 401CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)There is no expanded citation for this reference.
- 48Kristensen, A. R.; Foster, L. J. Protein correlation profiling-SILAC to study protein-protein interactions. Methods Mol. Biol. 2014, 1188, 263, DOI: 10.1007/978-1-4939-1142-4_18Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cbmtVOktA%253D%253D&md5=f8ed51d259f10f8fbfe305629a531e85Protein correlation profiling-SILAC to study protein-protein interactionsKristensen Anders R; Foster Leonard JMethods in molecular biology (Clifton, N.J.) (2014), 1188 (), 263-70 ISSN:.An interactome describes the global organization of protein interactions within a cell and is typically generated using affinity purification-mass spectrometry (AP-MS), yeast two-hybrid screening, or protein-fragment complementation assays (Gavin et al. Nature 440: 631-636, 2006; Krogan et al. Nature 440: 637-643, 2006; Uetz et al. Nature 403: 623-627, 2000; Tarassov et al. Science 320: 1465-1470, 2008). These techniques have been widely used to depict the interactome as we know it today but current models of interactomes do not contain stoichiometric or temporal information. In this chapter we describe size-exclusion chromatography (SEC) combined with protein correlation profiling-stable isotope labeling by amino acids in cell culture (PCP-SILAC) to generate dynamic chromatographs for thousands of proteins (Kristensen et al. Nat Methods 9: 907-909, 2012). Using the precise co-elution of two proteins as evidence that they interact, it is possible to identify similar numbers of protein interactions without overexpression or creating fusion proteins as other high-throughput techniques require. In addition, triplex SILAC allows us to quantify protein stoichiometry and temporal changes to the interactome following perturbation. Finally, SEC-PCP-SILAC is very time efficient since it generates two orders of magnitude fewer samples for LC-MS analysis and avoids the tedious tagging and purification steps, making it possible for everyone with a single mass spectrometer to study the interactome.
- 49Liu, F.; Lössl, P.; Scheltema, R.; Viner, R.; Heck, A. J. R. Optimized fragmentation schemes and data analysis strategies for proteome-wide cross-link identification. Nat. Commun. 2017, 8, 15473, DOI: 10.1038/ncomms15473Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXot1Wlurw%253D&md5=ca5c671ac595773e1aecefa279536e80Optimized fragmentation schemes and data analysis strategies for proteome-wide cross-link identificationLiu, Fan; Loessl, Philip; Scheltema, Richard; Viner, Rosa; Heck, Albert J. R.Nature Communications (2017), 8 (), 15473CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)We describe optimized fragmentation schemes and data anal. strategies substantially enhancing the depth and accuracy in identifying protein cross-links using non-restricted whole proteome databases. These include a novel hybrid data acquisition strategy to sequence cross-links at both MS2 and MS3 level and a new algorithmic design XlinkX v2.0 for data anal. As proof-of-concept we investigated proteome-wide protein interactions in E. coli and HeLa cell lysates, resp., identifying 1,158 and 3,301 unique cross-links at ~ 1% false discovery rate. These protein interaction repositories provide meaningful structural information on many endogenous macromol. assemblies, as we showcase on several protein complexes involved in translation, protein folding and carbohydrate metab.
Cited By
This article is cited by 49 publications.
- Tehmina Bharucha, Bevin Gangadharan, Abhinav Kumar, Ashleigh C. Myall, Nazli Ayhan, Boris Pastorino, Anisone Chanthongthip, Manivanh Vongsouvath, Mayfong Mayxay, Onanong Sengvilaipaseuth, Ooyanong Phonemixay, Sayaphet Rattanavong, Darragh P. O’Brien, Iolanda Vendrell, Roman Fischer, Benedikt Kessler, Lance Turtle, Xavier de Lamballerie, Audrey Dubot-Pérès, Paul N. Newton, Nicole Zitzmann, SEAe Consortium. Deep Proteomics Network and Machine Learning Analysis of Human Cerebrospinal Fluid in Japanese Encephalitis Virus Infection. Journal of Proteome Research 2023, 22
(6)
, 1614-1629. https://doi.org/10.1021/acs.jproteome.2c00563
- Miroslava Strmiskova, Jason D. Josephson, Caroline Toudic, John Paul Pezacki. Optimized Bioorthogonal Non-canonical Amino Acid Tagging to Identify Serotype-Specific Biomarkers in Verotoxigenic Escherichia coli. ACS Infectious Diseases 2023, 9
(4)
, 856-863. https://doi.org/10.1021/acsinfecdis.2c00548
- Kevin W. Cormier, Brett Larsen, Anne-Claude Gingras, James R. Woodgett. Interactomes of Glycogen Synthase Kinase-3 Isoforms. Journal of Proteome Research 2023, 22
(3)
, 977-989. https://doi.org/10.1021/acs.jproteome.2c00825
- Joseph Bloom, Aaron Triantafyllidis, Anna Quaglieri, Paula Burton Ngov, Giuseppe Infusini, Andrew Webb. Mass Dynamics 1.0: A Streamlined, Web-Based Environment for Analyzing, Sharing, and Integrating Label-Free Data. Journal of Proteome Research 2021, 20
(11)
, 5180-5188. https://doi.org/10.1021/acs.jproteome.1c00683
- Weijun Gui, Siqi Shen, Zhihao Zhuang. Photocaged Cell-Permeable Ubiquitin Probe for Temporal Profiling of Deubiquitinating Enzymes. Journal of the American Chemical Society 2020, 142
(46)
, 19493-19501. https://doi.org/10.1021/jacs.9b12426
- Tristan Cardon, Bilgehan Ozcan, Soulaimane Aboulouard, Firas Kobeissy, Marie Duhamel, Franck Rodet, Isabelle Fournier, Michel Salzet. Epigenetic Studies Revealed a Ghost Proteome in PC1/3 KD Macrophages under Antitumoral Resistance Induced by IL-10. ACS Omega 2020, 5
(43)
, 27774-27782. https://doi.org/10.1021/acsomega.0c02530
- Sung-Huan Yu, Pelagia Kyriakidou, Jürgen Cox. Isobaric Matching between Runs and Novel PSM-Level Normalization in MaxQuant Strongly Improve Reporter Ion-Based Quantification. Journal of Proteome Research 2020, 19
(10)
, 3945-3954. https://doi.org/10.1021/acs.jproteome.0c00209
- S. Ghorbanalipoor, T. Hommel, T. Kolbe, T. Fröhlich, B. Wagner, C. Posch, M. Dahlhoff. The loss of keratin 77 in murine skin is functionally compensated by keratin 1. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2025, 1872
(2)
, 119881. https://doi.org/10.1016/j.bbamcr.2024.119881
- Mengyao Wu, Huihui Tao, Tiantian Xu, Xuejia Zheng, Chunmei Wen, Guoying Wang, Yali Peng, Yong Dai. Spatial proteomics: unveiling the multidimensional landscape of protein localization in human diseases. Proteome Science 2024, 22
(1)
https://doi.org/10.1186/s12953-024-00231-2
- Xiaosha Wang, Layla Frühn, Panpan Li, Xin Shi, Nini Wang, Yuan Feng, Julia Prinz, Hanhan Liu, Verena Prokosch. Comparative proteomic analysis of regenerative mechanisms in mouse retina to identify markers for neuro-regeneration in glaucoma. Scientific Reports 2024, 14
(1)
https://doi.org/10.1038/s41598-024-72378-z
- Rico Ledwith, Tobias Stobernack, Antje Bergert, Aileen Bahl, Mario Pink, Andrea Haase, Verónica I. Dumit. Towards characterization of cell culture conditions for reliable proteomic analysis: in vitro studies on A549, differentiated THP-1, and NR8383 cell lines. Archives of Toxicology 2024, 98
(12)
, 4021-4031. https://doi.org/10.1007/s00204-024-03858-4
- Zhi-Feng Pan, Ling Du, Feng Liu. Protocol for analysis of plasma proteomes from patients with hepatocellular carcinoma receiving combination therapy. STAR Protocols 2024, 5
(4)
, 103308. https://doi.org/10.1016/j.xpro.2024.103308
- Faheem Seedat, Neva Kandzija, Michael J. Ellis, Shuhan Jiang, Asselzhan Sarbalina, James Bancroft, Edward Drydale, Svenja S. Hester, Roman Fischer, Alisha N. Wade, M. Irina Stefana, John A. Todd, Manu Vatish. Placental small extracellular vesicles from normal pregnancy and gestational diabetes increase insulin gene transcription and content in β cells. Clinical Science 2024, 138
(22)
, 1481-1502. https://doi.org/10.1042/CS20241782
- Delia Oosthuizen, Tariq A. Ganief, Kenneth E. Bernstein, Edward D. Sturrock. Proteomic Analysis of Human Macrophages Overexpressing Angiotensin-Converting Enzyme. International Journal of Molecular Sciences 2024, 25
(13)
, 7055. https://doi.org/10.3390/ijms25137055
- Maya Belghazi, Cécile Iborra, Ophélie Toutendji, Manon Lasserre, Dominique Debanne, Jean-Marc Goaillard, Béatrice Marquèze-Pouey. High-Resolution Proteomics Unravel a Native Functional Complex of Cav1.3, SK3, and Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels in Midbrain Dopaminergic Neurons. Cells 2024, 13
(11)
, 944. https://doi.org/10.3390/cells13110944
- André C. Michaelis, Andreas-David Brunner, Maximilian Zwiebel, Florian Meier, Maximilian T. Strauss, Isabell Bludau, Matthias Mann. The social and structural architecture of the yeast protein interactome. Nature 2023, 624
(7990)
, 192-200. https://doi.org/10.1038/s41586-023-06739-5
- Hee-Yeon Kim, Janbolat Ashim, Song Park, Wansoo Kim, Sangho Ji, Seoung-Woo Lee, Yi-Rang Jung, Sang Won Jeong, Se-Guen Lee, Hyun-Chul Kim, Young-Jae Lee, Mi Kyung Kwon, Jun-Seong Hwang, Jung Min Shin, Sung-Jun Lee, Wookyung Yu, Jin-Kyu Park, Seong-Kyoon Choi. A preliminary study about the potential risks of the UV-weathered microplastic: The proteome-level changes in the brain in response to polystyrene derived weathered microplastics. Environmental Research 2023, 233 , 116411. https://doi.org/10.1016/j.envres.2023.116411
- Birgitte Villadsen, Camilla Thygesen, Manuela Grebing, Stefan J. Kempf, Marie B. Sandberg, Pia Jensen, Stefanie H. Kolstrup, Helle H. Nielsen, Martin R. Larsen, Bente Finsen. Ceruloplasmin‐deficient mice show changes in
PTM
profiles of proteins involved in messenger
RNA
processing and neuronal projections and synaptic processes. Journal of Neurochemistry 2023, 165
(1)
, 76-94. https://doi.org/10.1111/jnc.15754
- Paula Carrillo-Rodriguez, Frode Selheim, Maria Hernandez-Valladares. Mass Spectrometry-Based Proteomics Workflows in Cancer Research: The Relevance of Choosing the Right Steps. Cancers 2023, 15
(2)
, 555. https://doi.org/10.3390/cancers15020555
- Mahmut Emir, Ahmet Caglar Ozketen, Ayse Andac Ozketen, Arzu Çelik Oğuz, Mei Huang, Aziz Karakaya, Christof Rampitsch, Aslihan Gunel. Increased levels of cell wall degrading enzymes and peptidases are associated with aggressiveness in a virulent isolate of Pyrenophora teres f. maculata. Journal of Plant Physiology 2022, 279 , 153839. https://doi.org/10.1016/j.jplph.2022.153839
- Ahmed B. Montaser, Janita Kuiri, Teemu Natunen, Pavel Hruška, David Potěšil, Seppo Auriola, Mikko Hiltunen, Tetsuya Terasaki, Marko Lehtonen, Aaro Jalkanen, Kristiina M. Huttunen. Enhanced drug delivery by a prodrug approach effectively relieves neuroinflammation in mice. Life Sciences 2022, 310 , 121088. https://doi.org/10.1016/j.lfs.2022.121088
- Adam Aleksander Karpiński, Julio Cesar Torres Elguera, Anne Sanner, Witold Konopka, Leszek Kaczmarek, Dominic Winter, Anna Konopka, Ewa Bulska. Study on Tissue Homogenization Buffer Composition for Brain Mass Spectrometry-Based Proteomics. Biomedicines 2022, 10
(10)
, 2466. https://doi.org/10.3390/biomedicines10102466
- Laken Kruger, Guihua Yue, Vijaya Saradhi Mettu, Alison Paquette, Sheela Sathyanarayana, Bhagwat Prasad. Differential proteomics analysis of JEG-3 and JAR placental cell models and the effect of androgen treatment. The Journal of Steroid Biochemistry and Molecular Biology 2022, 222 , 106138. https://doi.org/10.1016/j.jsbmb.2022.106138
- Minna M. Konert, Anna Wysocka, Peter Koník, Roman Sobotka. High-light-inducible proteins HliA and HliB: pigment binding and protein–protein interactions. Photosynthesis Research 2022, 152
(3)
, 317-332. https://doi.org/10.1007/s11120-022-00904-z
- Miao-Hsia Lin, Pei-Shan Wu, Tzu-Hsuan Wong, I-Ying Lin, Johnathan Lin, Jürgen Cox, Sung-Huan Yu. Benchmarking differential expression, imputation and quantification methods for proteomics data. Briefings in Bioinformatics 2022, 23
(3)
https://doi.org/10.1093/bib/bbac138
- Julia Patricia Schessner, Eugenia Voytik, Isabell Bludau. A practical guide to interpreting and generating bottom‐up proteomics data visualizations. PROTEOMICS 2022, 22
(8)
https://doi.org/10.1002/pmic.202100103
- Hamid Hamzeiy, Daniela Ferretti, Maria S. Robles, Jürgen Cox. Perseus plugin “Metis” for metabolic-pathway-centered quantitative multi-omics data analysis for static and time-series experimental designs. Cell Reports Methods 2022, 2
(4)
, 100198. https://doi.org/10.1016/j.crmeth.2022.100198
- Andrew T. Rajczewski, Pratik D. Jagtap, Timothy J. Griffin. An overview of technologies for MS-based proteomics-centric multi-omics. Expert Review of Proteomics 2022, 19
(3)
, 165-181. https://doi.org/10.1080/14789450.2022.2070476
- Balasubramanian Cibichakravarthy, Juan A. Oses-Prieto, Michael Ben-Yosef, Alma L. Burlingame, Timothy L. Karr, Yuval Gottlieb, . Comparative Proteomics of
Coxiella
like Endosymbionts (CLEs) in the Symbiotic Organs of Rhipicephalus sanguineus Ticks. Microbiology Spectrum 2022, 10
(1)
https://doi.org/10.1128/spectrum.01673-21
- Rodrigo Mohallem, Uma K. Aryal. Quantitative Proteomics and Phosphoproteomics Reveal TNF-α-Mediated Protein Functions in Hepatocytes. Molecules 2021, 26
(18)
, 5472. https://doi.org/10.3390/molecules26185472
- Fumiko Matsuzaki, Shinsuke Uda, Yukiyo Yamauchi, Masaki Matsumoto, Tomoyoshi Soga, Kazumitsu Maehara, Yasuyuki Ohkawa, Keiichi I. Nakayama, Shinya Kuroda, Hiroyuki Kubota. An extensive and dynamic trans-omic network illustrating prominent regulatory mechanisms in response to insulin in the liver. Cell Reports 2021, 36
(8)
, 109569. https://doi.org/10.1016/j.celrep.2021.109569
- Alexa Derksen, Hung-Yu Shih, Diane Forget, Lama Darbelli, Luan T. Tran, Christian Poitras, Kether Guerrero, Sundaresan Tharun, Fowzan S. Alkuraya, Wesam I. Kurdi, Cam-Tu Emilie Nguyen, Anne-Marie Laberge, Yue Si, Marie-Soleil Gauthier, Joshua L. Bonkowsky, Benoit Coulombe, Geneviève Bernard. Variants in LSM7 impair LSM complexes assembly, neurodevelopment in zebrafish and may be associated with an ultra-rare neurological disease. Human Genetics and Genomics Advances 2021, 2
(3)
, 100034. https://doi.org/10.1016/j.xhgg.2021.100034
- Rahimah Hassan, Nurulhasanah Othman, Sharif M. Mansor, Christian P. Müller, Zurina Hassan. Proteomic analysis reveals brain Rab35 as a potential biomarker of mitragynine withdrawal in rats. Brain Research Bulletin 2021, 172 , 139-150. https://doi.org/10.1016/j.brainresbull.2021.04.018
- Lir Beck, Michal Harel, Shun Yu, Ettai Markovits, Ben Boursi, Gal Markel, Tamar Geiger. Clinical Proteomics of Metastatic Melanoma Reveals Profiles of Organ Specificity and Treatment Resistance. Clinical Cancer Research 2021, 27
(7)
, 2074-2086. https://doi.org/10.1158/1078-0432.CCR-20-3752
- Gali Yanovich-Arad, Paula Ofek, Eilam Yeini, Mariya Mardamshina, Artem Danilevsky, Noam Shomron, Rachel Grossman, Ronit Satchi-Fainaro, Tamar Geiger. Proteogenomics of glioblastoma associates molecular patterns with survival. Cell Reports 2021, 34
(9)
, 108787. https://doi.org/10.1016/j.celrep.2021.108787
- Saul Lema A, Marina Klemenčič, Franziska Völlmy, Maarten Altelaar, Christiane Funk. The Role of Pseudo-Orthocaspase (SyOC) of Synechocystis sp. PCC 6803 in Attenuating the Effect of Oxidative Stress. Frontiers in Microbiology 2021, 12 https://doi.org/10.3389/fmicb.2021.634366
- Kinza Rian, Marta R. Hidalgo, Cankut Çubuk, Matias M. Falco, Carlos Loucera, Marina Esteban-Medina, Inmaculada Alamo-Alvarez, María Peña-Chilet, Joaquín Dopazo. Genome-scale mechanistic modeling of signaling pathways made easy: A bioconductor/cytoscape/web server framework for the analysis of omic data. Computational and Structural Biotechnology Journal 2021, 19 , 2968-2978. https://doi.org/10.1016/j.csbj.2021.05.022
- Bruno Pinto, Giovanni Morelli, Mohit Rastogi, Annalisa Savardi, Amos Fumagalli, Andrea Petretto, Martina Bartolucci, Emilio Varea, Tiziano Catelani, Andrea Contestabile, Laura E. Perlini, Laura Cancedda. Rescuing Over-activated Microglia Restores Cognitive Performance in Juvenile Animals of the Dp(16) Mouse Model of Down Syndrome. Neuron 2020, 108
(5)
, 887-904.e12. https://doi.org/10.1016/j.neuron.2020.09.010
- Cristian De Gregorio, David Contador, Diego Díaz, Constanza Cárcamo, Daniela Santapau, Lorena Lobos-Gonzalez, Cristian Acosta, Mario Campero, Daniel Carpio, Caterina Gabriele, Marco Gaspari, Victor Aliaga-Tobar, Vinicius Maracaja-Coutinho, Marcelo Ezquer, Fernando Ezquer. Human adipose-derived mesenchymal stem cell-conditioned medium ameliorates polyneuropathy and foot ulceration in diabetic BKS db/db mice. Stem Cell Research & Therapy 2020, 11
(1)
https://doi.org/10.1186/s13287-020-01680-0
- Timothy R. Howard, Ileana M. Cristea. Interrogating Host Antiviral Environments Driven by Nuclear DNA Sensing: A Multiomic Perspective. Biomolecules 2020, 10
(12)
, 1591. https://doi.org/10.3390/biom10121591
- Sung‐Huan Yu, Daniela Ferretti, Julia P. Schessner, Jan Daniel Rudolph, Georg H. H. Borner, Jürgen Cox. Expanding the Perseus Software for Omics Data Analysis With Custom Plugins. Current Protocols in Bioinformatics 2020, 71
(1)
https://doi.org/10.1002/cpbi.105
- Syed Feroj Ahmed, Lori Buetow, Mads Gabrielsen, Sergio Lilla, Chatrin Chatrin, Gary J. Sibbet, Sara Zanivan, Danny T. Huang. DELTEX2 C-terminal domain recognizes and recruits ADP-ribosylated proteins for ubiquitination. Science Advances 2020, 6
(34)
https://doi.org/10.1126/sciadv.abc0629
- Joel D. Federspiel, Katelyn C. Cook, Michelle A. Kennedy, Samvida S. Venkatesh, Clayton J. Otter, William A. Hofstadter, Pierre M. Jean Beltran, Ileana M. Cristea. Mitochondria and Peroxisome Remodeling across Cytomegalovirus Infection Time Viewed through the Lens of Inter-ViSTA. Cell Reports 2020, 32
(4)
, 107943. https://doi.org/10.1016/j.celrep.2020.107943
- Nellie A. Martin, Kirsten H. Hyrlov, Maria L. Elkjaer, Eva K. Thygesen, Agnieszka Wlodarczyk, Kirstine J. Elbaek, Christopher Aboo, Justyna Okarmus, Eirikur Benedikz, Richard Reynolds, Zoltan Hegedus, Allan Stensballe, Åsa Fex Svenningsen, Trevor Owens, Zsolt Illes. Absence of miRNA-146a Differentially Alters Microglia Function and Proteome. Frontiers in Immunology 2020, 11 https://doi.org/10.3389/fimmu.2020.01110
- Jason M. Held. Redox Systems Biology: Harnessing the Sentinels of the Cysteine Redoxome. Antioxidants & Redox Signaling 2020, 32
(10)
, 659-676. https://doi.org/10.1089/ars.2019.7725
- Chen Chen, Jie Hou, John J. Tanner, Jianlin Cheng. Bioinformatics Methods for Mass Spectrometry-Based Proteomics Data Analysis. International Journal of Molecular Sciences 2020, 21
(8)
, 2873. https://doi.org/10.3390/ijms21082873
- Chenyang Li, Jayaseelan Murugaiyan, Christian Thomas, Thomas Alter, Carolin Riedel. Isolate Specific Cold Response of Yersinia enterocolitica in Transcriptional, Proteomic, and Membrane Physiological Changes. Frontiers in Microbiology 2020, 10 https://doi.org/10.3389/fmicb.2019.03037
- Christof Rampitsch, Mei Huang, Slavica Djuric-Cignaovic, Xiben Wang, Ursla Fernando. Temporal Quantitative Changes in the Resistant and Susceptible Wheat Leaf Apoplastic Proteome During Infection by Wheat Leaf Rust (Puccinia triticina). Frontiers in Plant Science 2019, 10 https://doi.org/10.3389/fpls.2019.01291
- Michal Harel, Rona Ortenberg, Siva Karthik Varanasi, Kailash Chandra Mangalhara, Mariya Mardamshina, Ettai Markovits, Erez N. Baruch, Victoria Tripple, May Arama-Chayoth, Eyal Greenberg, Anjana Shenoy, Ruveyda Ayasun, Naama Knafo, Shihao Xu, Liat Anafi, Gali Yanovich-Arad, Georgina D. Barnabas, Shira Ashkenazi, Michal J. Besser, Jacob Schachter, Marcus Bosenberg, Gerald S. Shadel, Iris Barshack, Susan M. Kaech, Gal Markel, Tamar Geiger. Proteomics of Melanoma Response to Immunotherapy Reveals Mitochondrial Dependence. Cell 2019, 179
(1)
, 236-250.e18. https://doi.org/10.1016/j.cell.2019.08.012
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 49 other publications.
- 1Bar-Yam, Y. General Features of Complex Systems. Knowledge Management, Organizational Intelligence and Learning, and Complexity, Vol. I; Encyclopedia of Life Support Systems, 1997; www.eolss.netThere is no corresponding record for this reference.
- 2O’Connor, T.; Wong, H. Y. Emergent Properties. Stanford Encyclopedia of Philosophy, Spring 2012 ed.; Metaphysics Research Lab, Stanford University, 2012; https://plato.stanford.edu/archives/spr2012/entries/properties-emergent/There is no corresponding record for this reference.
- 3Grandjean, M. A social network analysis of Twitter: Mapping the digital humanities community. Cogent Arts Humanit. 2016, 3, 1171458, DOI: 10.1080/23311983.2016.1171458There is no corresponding record for this reference.
- 4Goffman, C. And What is Your Erdös Number?. Am. Math. Mon. 1969, 76, 791, DOI: 10.1080/00029890.1969.12000324There is no corresponding record for this reference.
- 5Sporns, O.; Tononi, G.; Kötter, R. The human connectome: A structural description of the human brain. PLoS Comput. Biol. 2005, 1, e42, DOI: 10.1371/journal.pcbi.0010042There is no corresponding record for this reference.
- 6Seung, S. I am my connectome. TED: Ideas Worth Spreading , 2010; https://www.ted.com/talks/sebastian_seung.There is no corresponding record for this reference.
- 7Seung, H. S. Connectome: How the Brain’s Wiring Makes Us Who We Are; Penguin Books Ltd., 2013.There is no corresponding record for this reference.
- 8Barabási, A.-L.; Oltvai, Z. N. Network biology: understanding the cell’s functional organization. Nat. Rev. Genet. 2004, 5, 101– 113, DOI: 10.1038/nrg12728https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXovV2mtg%253D%253D&md5=2ac9082dfaf88d56da1fdd75b9edddb4Network biology: Understanding the cell's functional organizationBarabasi, Albert-Laszlo; Oltvai, Zoltan N.Nature Reviews Genetics (2004), 5 (2), 101-113CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)A key aim of postgenomic biomedical research is to systematically catalog all mols. and their interactions within a living cell. There is a clear need to understand how these mols. and the interactions between them det. the function of this enormously complex machinery, both in isolation and when surrounded by other cells. Rapid advances in network biol. indicate that cellular networks are governed by universal laws and offer a new conceptual framework that could potentially revolutionize our view of biol. and disease pathologies in the twenty-first century.
- 9Butte, A. J.; Kohane, I. S. Mutual Information Relevance Networks: Functional Genomic Clustering Using Pairwise Entropy Measurements. Biocomputing 2000, 2000, 418– 429, DOI: 10.1142/9789814447331_0040There is no corresponding record for this reference.
- 10Sinitcyn, P.; Rudolph, J. D.; Cox, J. Computational Methods for Understanding Mass Spectrometry-Based Shotgun Proteomics Data. Annu. Rev. Biomed. Data Sci. 2018, 1, 207– 234, DOI: 10.1146/annurev-biodatasci-080917-013516There is no corresponding record for this reference.
- 11Tyanova, S. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 2016, 13, 731– 40, DOI: 10.1038/nmeth.390111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVKntbnN&md5=f8c3e2876e4d724518054bb1a2d1e6eeThe Perseus computational platform for comprehensive analysis of (prote)omics dataTyanova, Stefka; Temu, Tikira; Sinitcyn, Pavel; Carlson, Arthur; Hein, Marco Y.; Geiger, Tamar; Mann, Matthias; Cox, JuergenNature Methods (2016), 13 (9), 731-740CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)A main bottleneck in proteomics is the downstream biol. anal. of highly multivariate quant. protein abundance data generated using mass-spectrometry-based anal. We developed the Perseus software platform (http://www.perseus-framework.org) to support biol. and biomedical researchers in interpreting protein quantification, interaction and post-translational modification data. Perseus contains a comprehensive portfolio of statistical tools for high-dimensional omics data anal. covering normalization, pattern recognition, time-series anal., cross-omics comparisons and multiple-hypothesis testing. A machine learning module supports the classification and validation of patient groups for diagnosis and prognosis, and it also detects predictive protein signatures. Central to Perseus is a user-friendly, interactive workflow environment that provides complete documentation of computational methods used in a publication. All activities in Perseus are realized as plugins, and users can extend the software by programming their own, which can be shared through a plugin store. We anticipate that Perseus's arsenal of algorithms and its intuitive usability will empower interdisciplinary anal. of complex large data sets.
- 12Tyanova, S.; Cox, J. Methods in Molecular Biology Springer, 2018; Vol. 1711, pp 133– 148.There is no corresponding record for this reference.
- 13Shannon, P. Cytoscape: A software Environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498– 2504, DOI: 10.1101/gr.123930313https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXovFWrtr4%253D&md5=2bcbca9a3bd04717761f0424c0209e43Cytoscape: A software environment for integrated models of biomolecular interaction networksShannon, Paul; Markiel, Andrew; Ozier, Owen; Baliga, Nitin S.; Wang, Jonathan T.; Ramage, Daniel; Amin, Nada; Schwikowski, Benno; Ideker, TreyGenome Research (2003), 13 (11), 2498-2504CODEN: GEREFS; ISSN:1088-9051. (Cold Spring Harbor Laboratory Press)Cytoscape is an open source software project for integrating biomol. interaction networks with high-throughput expression data and other mol. states into a unified conceptual framework. Although applicable to any system of mol. components and interactions, Cytoscape is most powerful when used in conjunction with large databases of protein-protein, protein-DNA, and genetic interactions that are increasingly available for humans and model organisms. Cytoscape's software Core provides basic functionality to layout and query the network; to visually integrate the network with expression profiles, phenotypes, and other mol. states; and to link the network to databases of functional annotations. The Core is extensible through a straightforward plug-in architecture, allowing rapid development of addnl. computational analyses and features. Several case studies of Cytoscape plug-ins are surveyed, including a search for interaction pathways correlating with changes in gene expression, a study of protein complexes involved in cellular recovery to DNA damage, inference of a combined phys./functional interaction network for Halobacterium, and an interface to detailed stochastic/kinetic gene regulatory models.
- 14Kloet, S. L. The dynamic interactome and genomic targets of Polycomb complexes during stem-cell differentiation. Nat. Struct. Mol. Biol. 2016, 23, 682– 690, DOI: 10.1038/nsmb.324814https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsVCku7c%253D&md5=b5b35be17f21b312c593a16bedc27026The dynamic interactome and genomic targets of Polycomb complexes during stem-cell differentiationKloet, Susan L.; Makowski, Matthew M.; Baymaz, H. Irem; van Voorthuijsen, Lisa; Karemaker, Ino D.; Santanach, Alexandra; Jansen, Pascal W. T. C.; Di Croce, Luciano; Vermeulen, MichielNature Structural & Molecular Biology (2016), 23 (7), 682-690CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Although the core subunits of Polycomb group (PcG) complexes are well characterized, little is known about the dynamics of these protein complexes during cellular differentiation. We used quant. interaction proteomics and genome-wide profiling to study PcG proteins in mouse embryonic stem cells (ESCs) and neural progenitor cells (NPCs). We found that the stoichiometry and genome-wide binding of PRC1 and PRC2 were highly dynamic during neural differentiation. Intriguingly, we obsd. a downregulation and loss of PRC2 from chromatin marked with trimethylated histone H3 K27 (H3K27me3) during differentiation, whereas PRC1 was retained at these sites. Addnl., we found PRC1 at enhancer and promoter regions independently of PRC2 binding and H3K27me3. Finally, overexpression of NPC-specific PRC1 interactors in ESCs led to increased Ring1b binding to, and decreased expression of, NPC-enriched Ring1b-target genes. In summary, our integrative analyses uncovered dynamic PcG subcomplexes and their widespread colocalization with active chromatin marks during differentiation.
- 15Smith, C. L.; Blake, J. A.; Kadin, J. A.; Richardson, J. E.; Bult, C. J. Mouse Genome Database (MGD)-2018: Knowledgebase for the laboratory mouse. Nucleic Acids Res. 2018, 46, D836– 42, DOI: 10.1093/nar/gkx100615https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGit77M&md5=083422379b7c3bf55592a79bedb59056Mouse genome database (MGD)-2018: knowledge-base for the laboratory mouseSmith, Cynthia L.; Blake, Judith A.; Kadin, James A.; Richardson, Joel E.; Bult, Carol J.; Anagnostopoulos, A.; Andrews, A.; Baldarelli, R. M.; Beal, J. S.; Bello, S. M.; Blodgett, O.; Butler, N. E.; Christie, K.; Corbani, L. E.; Drabkin, H. J.; Espinoza, R.; Franco, J.; Giannatto, S. L.; Hale, P.; Hill, D. P.; Hutchins, L.; Law, M.; Lewis, J. R.; Mcandrews, M.; Mez, N.; Miers, D.; Motenko, H.; Ni, L.; Onda, H.; Perry, M.; Recla, J. M.; Reed, D. J.; Richards-Smith, B.; Sitnikov, D.; Tomczuk, M.; Wilming, L.; Zhu, Y.Nucleic Acids Research (2018), 46 (D1), D836-D842CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The Mouse Genome Database is the key community mouse database which supports basic, translational and computational research by providing integrated data on the genetics, genomics, and biol. of the lab. mouse. MGD serves as the source for biol. ref. data sets related to mouse genes, gene functions, phenotypes and disease models with an increasing emphasis on the assocn. of these data to human biol. and disease. We report here on recent enhancements to this resource, including improved access to mouse disease model and human phenotype data and enhanced relationships of mouse models to human disease.
- 16Langfelder, P.; Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinf. 2008, 9, 559, DOI: 10.1186/1471-2105-9-55916https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M%252FpvFOitw%253D%253D&md5=8e22feffdc9e990fe5a4aeecebfe4a7eWGCNA: an R package for weighted correlation network analysisLangfelder Peter; Horvath SteveBMC bioinformatics (2008), 9 (), 559 ISSN:.BACKGROUND: Correlation networks are increasingly being used in bioinformatics applications. For example, weighted gene co-expression network analysis is a systems biology method for describing the correlation patterns among genes across microarray samples. Weighted correlation network analysis (WGCNA) can be used for finding clusters (modules) of highly correlated genes, for summarizing such clusters using the module eigengene or an intramodular hub gene, for relating modules to one another and to external sample traits (using eigengene network methodology), and for calculating module membership measures. Correlation networks facilitate network based gene screening methods that can be used to identify candidate biomarkers or therapeutic targets. These methods have been successfully applied in various biological contexts, e.g. cancer, mouse genetics, yeast genetics, and analysis of brain imaging data. While parts of the correlation network methodology have been described in separate publications, there is a need to provide a user-friendly, comprehensive, and consistent software implementation and an accompanying tutorial. RESULTS: The WGCNA R software package is a comprehensive collection of R functions for performing various aspects of weighted correlation network analysis. The package includes functions for network construction, module detection, gene selection, calculations of topological properties, data simulation, visualization, and interfacing with external software. Along with the R package we also present R software tutorials. While the methods development was motivated by gene expression data, the underlying data mining approach can be applied to a variety of different settings. CONCLUSION: The WGCNA package provides R functions for weighted correlation network analysis, e.g. co-expression network analysis of gene expression data. The R package along with its source code and additional material are freely available at http://www.genetics.ucla.edu/labs/horvath/CoexpressionNetwork/Rpackages/WGCNA.
- 17Rudolph, J. D.; de Graauw, M.; van de Water, B.; Geiger, T.; Sharan, R. Elucidation of Signaling Pathways from Large-Scale Phosphoproteomic Data Using Protein Interaction Networks. Cell Syst. 2016, 3, 585– 593, DOI: 10.1016/j.cels.2016.11.00517https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFamu78%253D&md5=4bc19764bdb6820543b579e86d00df0dElucidation of Signaling Pathways from Large-Scale Phosphoproteomic Data Using Protein Interaction NetworksRudolph, Jan Daniel; de Graauw, Marjo; van de Water, Bob; Geiger, Tamar; Sharan, RodedCell Systems (2016), 3 (6), 585-593.e3CODEN: CSEYA4; ISSN:2405-4712. (Cell Press)Phosphoproteomic expts. typically identify sites within a protein that are differentially phosphorylated between two or more cell states. However, the interpretation of these data is hampered by the lack of methods that can translate site-specific information into global maps of active proteins and signaling networks, esp. as the phosphoproteome is often undersampled. Here, we describe PHOTON, a method for interpreting phosphorylation data within their signaling context, as captured by protein-protein interaction networks, to identify active proteins and pathways and pinpoint functional phosphosites. We apply PHOTON to interpret existing and novel phosphoproteomic datasets related to epidermal growth factor and insulin responses. PHOTON substantially outperforms the widely used cutoff approach, providing highly reproducible predictions that are more in line with current biol. knowledge. Altogether, PHOTON overcomes the fundamental challenge of delineating signaling pathways from large-scale phosphoproteomic data, thereby enabling translation of environmental cues to downstream cellular responses.
- 18Hornbeck, P. V. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 2015, 43, D512– 20, DOI: 10.1093/nar/gku126718https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVymt7rO&md5=df6fdbeb9f2e8cf025f1dea5179220afPhosphoSitePlus, 2014: mutations, PTMs and recalibrationsHornbeck, Peter V.; Zhang, Bin; Murray, Beth; Kornhauser, Jon M.; Latham, Vaughan; Skrzypek, ElzbietaNucleic Acids Research (2015), 43 (D1), D512-D520CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)PhosphoSitePlus (PSP), a knowledge-base dedicated to mammalian post-translational modifications (PTMs), contains over 330000 non-redundant PTMs, including phospho, acetyl, ubiquityl and Me groups. Over 95% of the sites are from mass spectrometry (MS) expts. In order to improve data reliability, early MS data have been reanalyzed, applying a common std. of anal. across over 1000000 spectra. Site assignments with P > 0.05 were filtered out. Two new downloads are available from PSP. The 'Regulatory sites' dataset includes curated information about modification sites that regulate downstream cellular processes, mol. functions and protein-protein interactions. The 'PTMVar' dataset, an intersect of missense mutations and PTMs from PSP, identifies over 25000 PTMVars (PTMs Impacted by Variants) that can rewire signaling pathways. The PTMVar data include missense mutations from UniPROTKB, TCGA and other sources that cause over 2000 diseases or syndromes (MIM) and polymorphisms, or are assocd. with hundreds of cancers. PTMVars include 18548 phosphorlyation sites, 3412 ubiquitylation sites, 2316 acetylation sites, 685 methylation sites and 245 succinylation sites.
- 19Yanovich, G. Clinical Proteomics of Breast Cancer Reveals a Novel Layer of Breast Cancer Classification. Cancer Res. 2018, 78, 6001– 6010, DOI: 10.1158/0008-5472.CAN-18-107919https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGhtbzL&md5=0a4b0f1f9fbd03b93e51335d002b7c12Clinical proteomics of breast cancer reveals a novel layer of breast cancer classificationYanovich, Gali; Agmon, Hadar; Harel, Michal; Sonnenblick, Amir; Peretz, Tamar; Geiger, TamarCancer Research (2018), 78 (20), 6001-6010CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Breast cancer classification has been the focus of numerous worldwide efforts, analyzing the mol. basis of breast cancer subtypes and aiming to assoc. them with clin. outcome and to improve the current diagnostic routine. Genomic and transcriptomic profiles of breast cancer have been well established, however the proteomic contribution to these profiles has yet to be elucidated. In this work, we utilized mass spectrometry-based proteomic anal. on more than 130 clin. breast samples to demonstrate intertumor heterogeneity across three breast cancer subtypes and healthy tissue. Unsupervised anal. identified four proteomic clusters, among them, one that represents a novel luminal subtype characterized by increased PI3K signaling. This subtype was further validated using an independent protein-based dataset, but not in two independent transcriptome cohorts. These results demonstrate the importance of deep proteomic anal., which may affect cancer treatment decision making.
- 20Szklarczyk, D. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362– D368, DOI: 10.1093/nar/gkw93720https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWhs70%253D&md5=cef71d32dbd02a49839447f168683d39The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessibleSzklarczyk, Damian; Morris, John H.; Cook, Helen; Kuhn, Michael; Wyder, Stefan; Simonovic, Milan; Santos, Alberto; Doncheva, Nadezhda T.; Roth, Alexander; Bork, Peer; Jensen, Lars J.; von Mering, ChristianNucleic Acids Research (2017), 45 (D1), D362-D368CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A system-wide understanding of cellular function requires knowledge of all functional interactions between the expressed proteins. The STRING database aims to collect and integrate this information, by consolidating known and predicted protein-protein assocn. data for a large no. of organisms. The assocns. in STRING include direct (phys.) interactions, as well as indirect (functional) interactions, as long as both are specific and biol. meaningful. Apart from collecting and reassessing available exptl. data on protein-protein interactions, and importing known pathways and protein complexes from curated databases, interaction predictions are derived from the following sources: (i) systematic coexpression anal., (ii) detection of shared selective signals across genomes, (iii) automated text-mining of the scientific literature and (iv) computational transfer of interaction knowledge between organisms based on gene orthol. In the latest version 10.5 of STRING, the biggest changes are concerned with data dissemination: the web front-end has been completely redesigned to reduce dependency on outdated browser technologies, and the database can now also be queried from inside the popular Cytoscape software framework. Further improvements include automated background anal. of user inputs for functional enrichments, and streamlined download options. The STRING resource is available online, at http://string-db.org/.
- 21Chatr-Aryamontri, A. The BioGRID interaction database: 2017 update. Nucleic Acids Res. 2017, 45, D369– D379, DOI: 10.1093/nar/gkw110221https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWhsrk%253D&md5=96ab82537dc1a8e03f383f8ad0a78298The BioGRID interaction database: 2017 updateChatr-aryamontri, Andrew; Oughtred, Rose; Boucher, Lorrie; Rust, Jennifer; Chang, Christie; Kolas, Nadine K.; O'Donnell, Lara; Oster, Sara; Theesfeld, Chandra; Sellam, Adnane; Stark, Chris; Breitkreutz, Bobby-Joe; Dolinski, Kara; Tyers, MikeNucleic Acids Research (2017), 45 (D1), D369-D379CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The Biol. General Repository for Interaction Datasets (BioGRID: https://thebiogrid.org) is an open access database dedicated to the annotation and archival of protein, genetic and chem. interactions for all major model organism species and humans. As of Sept. 2016 (build 3.4.140), the BioGRID contains 1 072 173 genetic and protein interactions, and 38 559 post-translational modifications, as manually annotated from 48 114 publications. This dataset represents interaction records for 66 model organisms and represents a 30% increase compared to the previous 2015 BioGRID update. BioGRID curates the biomedical literature for major model organism species, including humans, with a recent emphasis on central biol. processes and specific human diseases. To facilitate network-based approaches to drug discovery, BioGRID now incorporates 27 501 chem.-protein interactions for human drug targets, as drawn from the DrugBank database. A new dynamic interaction network viewer allows the easy navigation and filtering of all genetic and protein interaction data, as well as for bioactive compds. and their established targets. BioGRID data are directly downloadable without restriction in a variety of standardized formats and are freely distributed through partner model organism databases and meta-databases.
- 22Orchard, S. The MIntAct project - IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Res. 2014, 42, D358– 63, DOI: 10.1093/nar/gkt111522https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXoslKg&md5=f2964108748995acc8bc98b67880c95aThe MIntAct project-IntAct as a common curation platform for 11 molecular interaction databasesOrchard, Sandra; Ammari, Mais; Aranda, Bruno; Breuza, Lionel; Briganti, Leonardo; Broackes-Carter, Fiona; Campbell, Nancy H.; Chavali, Gayatri; Chen, Carol; del-Toro, Noemi; Duesbury, Margaret; Dumousseau, Marine; Galeota, Eugenia; Hinz, Ursula; Iannuccelli, Marta; Jagannathan, Sruthi; Jimenez, Rafael; Khadake, Jyoti; Lagreid, Astrid; Licata, Luana; Lovering, Ruth C.; Meldal, Birgit; Melidoni, Anna N.; Milagros, Mila; Peluso, Daniele; Perfetto, Livia; Porras, Pablo; Raghunath, Arathi; Ricard-Blum, Sylvie; Roechert, Bernd; Stutz, Andre; Tognolli, Michael; van Roey, Kim; Cesareni, Gianni; Hermjakob, HenningNucleic Acids Research (2014), 42 (D1), D358-D363CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)IntAct (freely available at http://www.ebi.ac.uk/intact) is an open-source, open data mol. interaction database populated by data either curated from the literature or from direct data depositions. IntAct has developed a sophisticated web-based curation tool, capable of supporting both IMEx- and MIMIx-level curation. This tool is now utilized by multiple addnl. curation teams, all of whom annotate data directly into the IntAct database. Members of the IntAct team supply appropriate levels of training, perform quality control on entries and take responsibility for long-term data maintenance. Recently, the MINT and IntAct databases decided to merge their sep. efforts to make optimal use of limited developer resources and maximize the curation output. All data manually curated by the MINT curators have been moved into the IntAct database at EMBL-EBI and are merged with the existing IntAct dataset. Both IntAct and MINT are active contributors to the IMEx consortium (http://www.imexconsortium.org).
- 23Ruepp, A. CORUM: The comprehensive resource of mammalian protein complexes-2009. Nucleic Acids Res. 2010, 38, D497– 501, DOI: 10.1093/nar/gkp91423https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXktlynug%253D%253D&md5=8803df58fa6353157f8a058f036d9145CORUM: the comprehensive resource of mammalian protein complexes-2009Ruepp, Andreas; Waegele, Brigitte; Lechner, Martin; Brauner, Barbara; Dunger-Kaltenbach, Irmtraud; Fobo, Gisela; Frishman, Goar; Montrone, Corinna; Mewes, H.-WernerNucleic Acids Research (2010), 38 (Database Iss), D497-D501CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)CORUM is a database that provides a manually curated repository of exptl. characterized protein complexes from mammalian organisms, mainly human (64%), mouse (16%) and rat (12%). Protein complexes are key mol. entities that integrate multiple gene products to perform cellular functions. The new CORUM 2.0 release encompasses 2837 protein complexes offering the largest and most comprehensive publicly available dataset of mammalian protein complexes. The CORUM dataset is built from 3198 different genes, representing ∼16% of the protein coding genes in humans. Each protein complex is described by a protein complex name, subunit compn., function as well as the literature ref. that characterizes the resp. protein complex. Recent developments include mapping of functional annotation to Gene Ontol. terms as well as cross-refs. to Entrez Gene identifiers. In addn., a Phylogenetic Conservation' anal. tool was implemented that analyses the potential occurrence of orthologous protein complex subunits in mammals and other selected groups of organisms. This allows one to predict the occurrence of protein complexes in different phylogenetic groups. CORUM is freely accessible at (http://mips.helmholtz-muenchen.de/genre/proj/corum/index.html).
- 24Gingras, A. C.; Gstaiger, M.; Raught, B.; Aebersold, R. Analysis of protein complexes using mass spectrometry. Nat. Rev. Mol. Cell Biol. 2007, 8, 645– 654, DOI: 10.1038/nrm220824https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXotVahtro%253D&md5=0f6219bd2475259e258ea56238a8c6dcAnalysis of protein complexes using mass spectrometryGingras, Anne-Claude; Gstaiger, Matthias; Raught, Brian; Aebersold, RuediNature Reviews Molecular Cell Biology (2007), 8 (8), 645-654CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. The versatile combination of affinity purifn. and mass spectrometry (AP-MS) has recently been applied to the detailed characterization of many protein complexes and large protein-interaction networks. The combination of AP-MS with other techniques, such as biochem. fractionation, intact mass measurement and chem. crosslinking, can help to decipher the supramol. organization of protein complexes. AP-MS can also be combined with quant. proteomics approaches to better understand the dynamics of protein-complex assembly.
- 25Dunham, W. H.; Mullin, M.; Gingras, A. C. Affinity-purification coupled to mass spectrometry: Basic principles and strategies. Proteomics 2012, 12, 1576, DOI: 10.1002/pmic.20110052325https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xoslehur0%253D&md5=602ff89e9f3496a7934f4f52a3abc1e5Affinity-purification coupled to mass spectrometry: Basic principles and strategiesDunham, Wade H.; Mullin, Michael; Gingras, Anne-ClaudeProteomics (2012), 12 (10), 1576-1590CODEN: PROTC7; ISSN:1615-9853. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Identifying the interactions established by a protein of interest can be a crit. step in understanding its function. This is esp. true when an unknown protein of interest is demonstrated to phys. interact with proteins of known function. While many techniques have been developed to characterize protein-protein interactions, one strategy that has gained considerable momentum over the past decade for identification and quantification of protein-protein interactions, is affinity-purifn. followed by mass spectrometry (AP-MS). Here, we briefly review the basic principles used in affinity-purifn. coupled to mass spectrometry, with an emphasis on tools (both biochem. and computational), which enable the discovery and reporting of high quality protein-protein interactions.
- 26Hein, M. Y. A Human Interactome in Three Quantitative Dimensions Organized by Stoichiometries and Abundances. Cell 2015, 163, 712– 723, DOI: 10.1016/j.cell.2015.09.05326https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslWrsLnK&md5=599f46801c2006cbc7b26d6ff8a00d6dA Human Interactome in Three Quantitative Dimensions Organized by Stoichiometries and AbundancesHein, Marco Y.; Hubner, Nina C.; Poser, Ina; Cox, Juergen; Nagaraj, Nagarjuna; Toyoda, Yusuke; Gak, Igor A.; Weisswange, Ina; Mansfeld, Joerg; Buchholz, Frank; Hyman, Anthony A.; Mann, MatthiasCell (Cambridge, MA, United States) (2015), 163 (3), 712-723CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The organization of a cell emerges from the interactions in protein networks. The interactome is critically dependent on the strengths of interactions and the cellular abundances of the connected proteins, both of which span orders of magnitude. However, these aspects have not yet been analyzed globally. Here, the authors have generated a library of HeLa cell lines expressing 1125 GFP-tagged proteins under near-endogenous control, which the authors used as input for a next-generation interaction survey. Using quant. proteomics, the authors detect specific interactions, est. interaction stoichiometries, and measure cellular abundances of interacting proteins. These three quant. dimensions reveal that the protein network is dominated by weak, substoichiometric interactions that play a pivotal role in defining network topol. The minority of stable complexes can be identified by their unique stoichiometry signature. This study provides a rich interaction dataset connecting thousands of proteins and introduces a framework for quant. network anal.
- 27Huttlin, E. L. The BioPlex Network: A Systematic Exploration of the Human Interactome. Cell 2015, 162, 425– 440, DOI: 10.1016/j.cell.2015.06.04327https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1KgtL3I&md5=79b8d96037646f6679baab3b966b9d47The BioPlex Network: A Systematic Exploration of the Human InteractomeHuttlin, Edward L.; Ting, Lily; Bruckner, Raphael J.; Gebreab, Fana; Gygi, Melanie P.; Szpyt, John; Tam, Stanley; Zarraga, Gabriela; Colby, Greg; Baltier, Kurt; Dong, Rui; Guarani, Virginia; Vaites, Laura Pontano; Ordureau, Alban; Rad, Ramin; Erickson, Brian K.; Wuhr, Martin; Chick, Joel; Zhai, Bo; Kolippakkam, Deepak; Mintseris, Julian; Obar, Robert A.; Harris, Tim; Artavanis-Tsakonas, Spyros; Sowa, Mathew E.; De Camilli, Pietro; Paulo, Joao A.; Harper, J. Wade; Gygi, Steven P.Cell (Cambridge, MA, United States) (2015), 162 (2), 425-440CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Protein interactions form a network whose structure drives cellular function and whose organization informs biol. inquiry. Using high-throughput affinity-purifn. mass spectrometry, the authors identify interacting partners for 2594 human proteins in HEK293T cells. The resulting network (BioPlex) contains 23,744 interactions among 7668 proteins with 86% previously undocumented. BioPlex accurately depicts known complexes, attaining 80%-100% coverage for most CORUM complexes. The network readily subdivides into communities that correspond to complexes or clusters of functionally related proteins. More generally, network architecture reflects cellular localization, biol. process, and mol. function, enabling functional characterization of thousands of proteins. Network structure also reveals assocns. among thousands of protein domains, suggesting a basis for examg. structurally related proteins. Finally, BioPlex, in combination with other approaches, can be used to reveal interactions of biol. or clin. significance. For example, mutations in the membrane protein VAPB implicated in familial amyotrophic lateral sclerosis perturb a defined community of interactors.
- 28Hubner, N. C. Quantitative proteomics combined with BAC TransgeneOmics reveals in vivo protein interactions. J. Cell Biol. 2010, 189, 739– 754, DOI: 10.1083/jcb.20091109128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmslKktrg%253D&md5=4cf589eedd456fb8a9b141ef0d5bc4f4Quantitative proteomics combined with BAC TransgeneOmics reveals in vivo protein interactionsHubner, Nina C.; Bird, Alexander W.; Cox, Juergen; Splettstoesser, Bianca; Bandilla, Peter; Poser, Ina; Hyman, Anthony; Mann, MatthiasJournal of Cell Biology (2010), 189 (4), 739-754CODEN: JCLBA3; ISSN:0021-9525. (Rockefeller University Press)Protein interactions are involved in all cellular processes. Their efficient and reliable characterization is therefore essential for understanding biol. mechanisms. In this study, we show that combining bacterial artificial chromosome (BAC) TransgeneOmics with quant. interaction proteomics, which we call quant. BAC-green fluorescent protein interactomics (QUBIC), allows specific and highly sensitive detection of interactions using rapid, generic, and quant. procedures with minimal material. We applied this approach to identify known and novel components of well-studied complexes such as the anaphase-promoting complex. Furthermore, we demonstrate second generation interaction proteomics by incorporating directed mutational transgene modification and drug perturbation into QUBIC. These methods identified domain/isoform-specific interactors of pericentrin- and phosphorylation-specific interactors of TACC3, which are necessary for its recruitment to mitotic spindles. The scalability, simplicity, cost effectiveness, and sensitivity of this method provide a basis for its general use in small-scale expts. and in mapping the human protein interactome.
- 29Noble, W. S. How does multiple testing correction work?. Nat. Biotechnol. 2009, 27, 1135– 1137, DOI: 10.1038/nbt1209-113529https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFemtr3N&md5=7376f0b000468adffe6e6c681ef84c95How does multiple testing correction work?Noble, William S.Nature Biotechnology (2009), 27 (12), 1135-1137CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. When prioritizing hits from a high-throughput expt., it is important to correct for random events that falsely appear significant. How is this done and what methods should be used. Techniques for doing this are discussed here.
- 30Tusher, V. G.; Tibshirani, R.; Chu, G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 5116– 5121, DOI: 10.1073/pnas.09106249830https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXjt1Ons7w%253D&md5=819240102af66293c77046878e4ca3c3Significance analysis of microarrays applied to the ionizing radiation responseTusher, Virginia Goss; Tibshirani, Robert; Chu, GilbertProceedings of the National Academy of Sciences of the United States of America (2001), 98 (9), 5116-5121CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Microarrays can measure the expression of thousands of genes to identify changes in expression between different biol. states. Methods are needed to det. the significance of these changes while accounting for the enormous no. of genes. We describe a method, Significance Anal. of Microarrays (SAM), that assigns a score to each gene on the basis of change in gene expression relative to the std. deviation of repeated measurements. For genes with scores greater than an adjustable threshold, SAM uses permutations of the repeated measurements to est. the percentage of genes identified by chance, the false discovery rate (FDR). When the transcriptional response of human cells to ionizing radiation was measured by microarrays, SAM identified 34 genes that changed at least 1.5-fold with an estd. FDR of 12%, compared with FDRs of 60 and 84% by using conventional methods of anal. Of the 34 genes, 19 were involved in cell cycle regulation and 3 in apoptosis. Surprisingly, four nucleotide excision repair genes were induced, suggesting that this repair pathway for UV-damaged DNA might play a previously unrecognized role in repairing DNA damaged by ionizing radiation.
- 31Franz, M. Cytoscape.js: A graph theory library for visualisation and analysis. Bioinformatics 2015, 32, 309– 311, DOI: 10.1093/bioinformatics/btv557There is no corresponding record for this reference.
- 32Dogrusoz, U.; Giral, E.; Cetintas, A.; Civril, A.; Demir, E. A layout algorithm for undirected compound graphs. Inf. Sci. (N. Y.) 2009, 179, 980– 994, DOI: 10.1016/j.ins.2008.11.017There is no corresponding record for this reference.
- 33Pedamallu, C. S.; Ozdamar, L. A Review on protein-protein interaction network databases. In Springer Proceedings in Mathematics and Statistics; Springer, 2014; Vol. 73, pp 511– 519.There is no corresponding record for this reference.
- 34Alanis-Lobato, G.; Andrade-Navarro, M. A.; Schaefer, M. H. HIPPIE v2.0: Enhancing meaningfulness and reliability of protein-protein interaction networks. Nucleic Acids Res. 2017, 45, D408– 14, DOI: 10.1093/nar/gkw98534https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWht74%253D&md5=400725d91e68e757a112622623fb5e63HIPPIE v2.0: enhancing meaningfulness and reliability of protein-protein interaction networksAlanis-Lobato, Gregorio; Andrade-Navarro, Miguel A.; Schaefer, Martin H.Nucleic Acids Research (2017), 45 (D1), D408-D414CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The increasing no. of exptl. detected interactions between proteins makes it difficult for researchers to ext. the interactions relevant for specific biol. processes or diseases. This makes it necessary to accompany the large-scale detection of protein-protein interactions (PPIs) with strategies and tools to generate meaningful PPI subnetworks. To this end, we generated the Human Integrated Protein-Protein Interaction ref. or HIPPIE (http://cbdm.uni-mainz.de/hippie/). HIPPIE is a one-stop resource for the generation and interpretation of PPI networks relevant to a specific research question. We provide means to generate highly reliable, context-specific PPI networks and to make sense out of them. We just released the second major update of HIPPIE, implementing various new features. HIPPIE grew substantially over the last years and now contains more than 270 000 confidence scored and annotated PPIs. We integrated different types of exptl. information for the confidence scoring and the construction of context-specific networks. We implemented basic graph algorithms that highlight important proteins and interactions. HIPPIE's graphical interface implements several ways for wet lab and computational scientists alike to access the PPI data.
- 35Gene Ontology Consortium, C. Gene Ontology Consortium: going forward. Nucleic Acids Res. 2015, 43, D1049– 56, DOI: 10.1093/nar/gku1179There is no corresponding record for this reference.
- 36Clauset, A.; Shalizi, C. R.; Newman, M. E. J. Power-law distributions in empirical data. SIAM Rev. 2009, 51, 661, DOI: 10.1137/070710111There is no corresponding record for this reference.
- 37Albert, R. Scale-free networks in cell biology. J. Cell Sci. 2005, 118, 4947, DOI: 10.1242/jcs.0271437https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1ynsbjI&md5=966ea6581f396e2515be7c229748fbf9Scale-free networks in cell biologyAlbert, RekaJournal of Cell Science (2005), 118 (21), 4947-4957CODEN: JNCSAI; ISSN:0021-9533. (Company of Biologists Ltd.)A review. A cell's behavior is a consequence of the complex interactions between its numerous constituents, such as DNA, RNA, proteins and small mols. Cells use signaling pathways and regulatory mechanisms to coordinate multiple processes, allowing them to respond to and adapt to an ever-changing environment. The large no. of components, the degree of interconnectivity and the complex control of cellular networks are becoming evident in the integrated genomic and proteomic analyses that are emerging. It is increasingly recognized that the understanding of properties that arise from whole-cell function require integrated, theor. descriptions of the relationships between different cellular components. Recent theor. advances allow us to describe cellular network structure with graph concepts and have revealed organizational features shared with numerous non-biol. networks. We now have the opportunity to describe quant. a network of hundreds or thousands of interacting components. Moreover, the obsd. topologies of cellular networks give us clues about their evolution and how their organization influences their function and dynamic responses.
- 38Riley, N. M.; Coon, J. J. Phosphoproteomics in the Age of Rapid and Deep Proteome Profiling. Anal. Chem. 2016, 88, 74– 94, DOI: 10.1021/acs.analchem.5b0412338https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslygur%252FE&md5=5b91915e258ce17207c3d433adae955bPhosphoproteomics in the Age of Rapid and Deep Proteome ProfilingRiley, Nicholas M.; Coon, Joshua J.Analytical Chemistry (Washington, DC, United States) (2016), 88 (1), 74-94CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A review. Protein phosphorylation is a post-translational modification (PTM) that orchestrates a diverse array of cellular processes. Because this modification serves as a rapid and reversible means to modulate protein activity and transduce signals, the regulation of phosphorylation is a central mechanism in cell health and disease. The addn. and removal of phosphoryl modifications via kinases and phosphates, resp., makes the landscape of phosphorylation particularly dynamic. Understanding the complex networks and functions coordinated by phosphorylation requires knowledge of specific amino acid modifications with both spatial and temporal resoln. - a task that remains a challenging anal. endeavor. Mass spectrometry (MS) has emerged as the premier tool for global PTM anal., boasting high sensitivity, considerable throughput, and the capacity to localize modifications to a single residue. Indeed, MS-centric phosphoproteomics has become a std. approach for investigating protein phosphorylation in labs. worldwide. Anal. Chem. last reviewed the contribution of MS and related technologies to phosphoproteomics in 2011. Since that time, MS methodol. has developed at an impressive pace. While routine proteomic expts. can now analyze thousands of proteins in just a few hours, rather than days or weeks, characterizing the global phosphoproteome is significantly more challenging than measuring non-modified proteins. The relative low abundance of phosphorylated peptides and the need for residue-specific information require special considerations in sample handling, data acquisition, and post-acquisition processing that constrain reproducibility, quant. efficacy, throughput, and depth in phosphoproteomic workflows. Advances in MS-based approaches have remarkably improved our abilities to investigate the many roles of protein phosphorylation across a diverse set of biol. contexts, but many tech. obstacles still exist. Poor run-to-run overlap, challenges in confident phosphosite assignment, and complications inherent to various quant. strategies limit biol. insight, despite ever-increasing nos. of detected phosphopeptides. Focusing on work from the past two years (2013-2015), this review examines major developments in MS technol. that have enabled the characterization of tens of thousands of phosphopeptides in a given expt., and considers the contribution of this anal. power to translational research. We also discuss how future innovation can address tech. challenges of today's methods, and we offer our perspective on how phosphoproteomics will continue to mature.
- 39Casado, P. Kinase-substrate enrichment analysis provides insights into the heterogeneity of signaling pathway activation in leukemia cells. Sci. Signaling 2013, 6, rs6– rs6, DOI: 10.1126/scisignal.2003573There is no corresponding record for this reference.
- 40Hernandez-Armenta, C.; Ochoa, D.; Gonçalves, E.; Saez-Rodriguez, J.; Beltrao, P. Benchmarking substrate-based kinase activity inference using phosphoproteomic data. Bioinformatics 2017, 33, 1845– 1851, DOI: 10.1093/bioinformatics/btx08240https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFagsbfI&md5=dafb0673d76db1772c48306366ba37c0Benchmarking substrate-based kinase activity inference using phosphoproteomic dataHernandez-Armenta, Claudia; Ochoa, David; Goncalves, Emanuel; Saez-Rodriguez, Julio; Beltrao, PedroBioinformatics (2017), 33 (12), 1845-1851CODEN: BOINFP; ISSN:1460-2059. (Oxford University Press)Motivation: Phosphoproteomic expts. are increasingly used to study the changes in signaling occurring across different conditions. It has been proposed that changes in phosphorylation of kinase target sites can be used to infer when a kinase activity is under regulation. However, these approaches have not yet been benchmarked due to a lack of appropriate benchmarking strategies. Results: We used curated phosphoproteomic expts. and a gold std. dataset contg. a total of 184 kinase-condition pairs where regulation is expected to occur to benchmark and compare different kinase activity inference strategies: Z-test, Kolmogorov Smirnov test, Wilcoxon rank sum test, gene set enrichment anal. (GSEA), and a multiple linear regression model. We also tested weighted variants of the Z-test and GSEA that include information on kinase sequence specificity as proxy for affinity. Finally, we tested how the no. of known substrates and the type of evidence (in vivo, in vitro or in silico) supporting these influence the predictions. Conclusions: Most models performed well with the Z-test and the GSEA performing best as detd. by the area under the ROC curve (Mean AUC 1/4 0.722). Weighting kinase targets by the kinase target sequence preference improves the results marginally. However, the no. of known substrates and the evidence supporting the interactions has a strong effect on the predictions.
- 41Herranz, N. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 2015, 17, 1205, DOI: 10.1038/ncb322541https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlCksbzL&md5=18eaa8c3ea3ee236774ef90e489535bcmTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotypeHerranz, Nicolas; Gallage, Suchira; Mellone, Massimiliano; Wuestefeld, Torsten; Klotz, Sabrina; Hanley, Christopher J.; Raguz, Selina; Acosta, Juan Carlos; Innes, Andrew J.; Banito, Ana; Georgilis, Athena; Montoya, Alex; Wolter, Katharina; Dharmalingam, Gopuraja; Faull, Peter; Carroll, Thomas; Martinez-Barbera, Juan Pedro; Cutillas, Pedro; Reisinger, Florian; Heikenwalder, Mathias; Miller, Richard A.; Withers, Dominic; Zender, Lars; Thomas, Gareth J.; Gil, JesusNature Cell Biology (2015), 17 (9), 1205-1217CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)Senescent cells secrete a combination of factors collectively known as the senescence-assocd. secretory phenotype (SASP). The SASP reinforces senescence and activates an immune surveillance response, but it can also show pro-tumorigenic properties and contribute to age-related pathologies. In a drug screen to find new SASP regulators, we uncovered the mTOR inhibitor rapamycin as a potent SASP suppressor. Here we report a mechanism by which mTOR controls the SASP by differentially regulating the translation of the MK2 (also known as MAPKAPK2) kinase through 4EBP1. In turn, MAPKAPK2 phosphorylates the RNA-binding protein ZFP36L1 during senescence, inhibiting its ability to degrade the transcripts of numerous SASP components. Consequently, mTOR inhibition or constitutive activation of ZFP36L1 impairs the non-cell-autonomous effects of senescent cells in both tumor-suppressive and tumor-promoting contexts. Altogether, our results place regulation of the SASP as a key mechanism by which mTOR could influence cancer, age-related diseases and immune responses.
- 42Wilkes, E. H.; Terfve, C.; Gribben, J. G.; Saez-Rodriguez, J.; Cutillas, P. R. Empirical inference of circuitry and plasticity in a kinase signaling network. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 7719, DOI: 10.1073/pnas.142334411242https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXpslyjsLw%253D&md5=e144af280676314d91f41d0b06f79a3dEmpirical inference of circuitry and plasticity in a kinase signaling networkWilkes, Edmund H.; Terfve, Camille; Gribben, John G.; Saez-Rodriguez, Julio; Cutillas, Pedro RodriguezProceedings of the National Academy of Sciences of the United States of America (2015), 112 (25), 7719-7724CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Our understanding of physiol. and disease is hampered by the difficulty of measuring the circuitry and plasticity of signaling networks that regulate cell biol., and how these relate to phenotypes. Here, using mass spectrometry-based phosphoproteomics, we systematically characterized the topol. of a network comprising the PI3K/Akt/mTOR and MEK/ERK signaling axes and confirmed its biol. relevance by assessing its dynamics upon EGF and IGF1 stimulation. Measuring the activity of this network in models of acquired drug resistance revealed that cells chronically treated with PI3K or mTORC1/2 inhibitors differed in the way their networks were remodeled. Unexpectedly, we also obsd. a degree of heterogeneity in the network state between cells resistant to the same inhibitor, indicating that even identical and carefully controlled exptl. conditions can give rise to the evolution of distinct kinase network statuses. These data suggest that the initial conditions of the system do not necessarily det. the mechanism by which cancer cells become resistant to PI3K/mTOR targeted therapies. The patterns of signaling network activity obsd. in the resistant cells mirrored the patterns of response to several drug combination treatments, suggesting that the activity of the defined signaling network truly reflected the evolved phenotypic diversity.
- 43Linding, R. NetworKIN: A resource for exploring cellular phosphorylation networks. Nucleic Acids Res. 2007, 36, D695– 99, DOI: 10.1093/nar/gkm902There is no corresponding record for this reference.
- 44Wiredja, D. D.; Koyutürk, M.; Chance, M. R. The KSEA App: a web-based tool for kinase activity inference from quantitative phosphoproteomics. Bioinformatics 2017, 33, 3489, DOI: 10.1093/bioinformatics/btx41544https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFWmsbnO&md5=7500765789d1dd6304968f51b89958bcThe KSEA app: A web-based tool for kinase activity inference from quantitative phosphoproteomicsWiredja, Danica D.; Koyuturk, Mehmet; Chance, Mark R.Bioinformatics (2017), 33 (21), 3489-3491CODEN: BOINFP; ISSN:1367-4811. (Oxford University Press)Summary: Computational characterization of differential kinase activity from phosphoproteomics datasets is crit. for correctly inferring cellular circuitry and how signaling cascades are altered in drug treatment and/or disease. Kinase-Substrate Enrichment Anal. (KSEA) offers a powerful approach to estg. changes in a kinase's activity based on the collective phosphorylation changes of its identified substrates. However, KSEA has been limited to programmers who are able to implement the algorithms. Thus, to make it accessible to the larger scientific community, we present a web-based application of this method: the KSEA App. Overall, we expect that this tool will offer a quick and user-friendly way of generating kinase activity ests. from high-throughput phosphoproteomics datasets.
- 45Zhang, B.; Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 2005, 4, 17, DOI: 10.2202/1544-6115.1128There is no corresponding record for this reference.
- 46Cox, J.; Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 2008, 26, 1367– 1372, DOI: 10.1038/nbt.151146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVWjtLzJ&md5=675d31ca84e3a7e4fb9bdd601d8075eaMaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantificationCox, Juergen; Mann, MatthiasNature Biotechnology (2008), 26 (12), 1367-1372CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Efficient anal. of very large amts. of raw data for peptide identification and protein quantification is a principal challenge in mass spectrometry (MS)-based proteomics. Here we describe MaxQuant, an integrated suite of algorithms specifically developed for high-resoln., quant. MS data. Using correlation anal. and graph theory, MaxQuant detects peaks, isotope clusters and stable amino acid isotope-labeled (SILAC) peptide pairs as three-dimensional objects in m/z, elution time and signal intensity space. By integrating multiple mass measurements and correcting for linear and nonlinear mass offsets, we achieve mass accuracy in the p.p.b. range, a sixfold increase over std. techniques. We increase the proportion of identified fragmentation spectra to 73% for SILAC peptide pairs via unambiguous assignment of isotope and missed-cleavage state and individual mass precision. MaxQuant automatically quantifies several hundred thousand peptides per SILAC-proteome expt. and allows statistically robust identification and quantification of >4000 proteins in mammalian cell lysates.
- 47Sinitcyn, P. MaxQuant goes Linux. Nat. Methods 2018, 15, 401, DOI: 10.1038/s41592-018-0018-y47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFOmtL%252FI&md5=d1aaed94810a18c96d0da5de8a9714d4MaxQuant goes LinuxSinitcyn, Pavel; Tiwary, Shivani; Rudolph, Jan; Gutenbrunner, Petra; Wichmann, Christoph; Yilmaz, Sule; Hamzeiy, Hamid; Salinas, Favio; Cox, JuergenNature Methods (2018), 15 (6), 401CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)There is no expanded citation for this reference.
- 48Kristensen, A. R.; Foster, L. J. Protein correlation profiling-SILAC to study protein-protein interactions. Methods Mol. Biol. 2014, 1188, 263, DOI: 10.1007/978-1-4939-1142-4_1848https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cbmtVOktA%253D%253D&md5=f8ed51d259f10f8fbfe305629a531e85Protein correlation profiling-SILAC to study protein-protein interactionsKristensen Anders R; Foster Leonard JMethods in molecular biology (Clifton, N.J.) (2014), 1188 (), 263-70 ISSN:.An interactome describes the global organization of protein interactions within a cell and is typically generated using affinity purification-mass spectrometry (AP-MS), yeast two-hybrid screening, or protein-fragment complementation assays (Gavin et al. Nature 440: 631-636, 2006; Krogan et al. Nature 440: 637-643, 2006; Uetz et al. Nature 403: 623-627, 2000; Tarassov et al. Science 320: 1465-1470, 2008). These techniques have been widely used to depict the interactome as we know it today but current models of interactomes do not contain stoichiometric or temporal information. In this chapter we describe size-exclusion chromatography (SEC) combined with protein correlation profiling-stable isotope labeling by amino acids in cell culture (PCP-SILAC) to generate dynamic chromatographs for thousands of proteins (Kristensen et al. Nat Methods 9: 907-909, 2012). Using the precise co-elution of two proteins as evidence that they interact, it is possible to identify similar numbers of protein interactions without overexpression or creating fusion proteins as other high-throughput techniques require. In addition, triplex SILAC allows us to quantify protein stoichiometry and temporal changes to the interactome following perturbation. Finally, SEC-PCP-SILAC is very time efficient since it generates two orders of magnitude fewer samples for LC-MS analysis and avoids the tedious tagging and purification steps, making it possible for everyone with a single mass spectrometer to study the interactome.
- 49Liu, F.; Lössl, P.; Scheltema, R.; Viner, R.; Heck, A. J. R. Optimized fragmentation schemes and data analysis strategies for proteome-wide cross-link identification. Nat. Commun. 2017, 8, 15473, DOI: 10.1038/ncomms1547349https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXot1Wlurw%253D&md5=ca5c671ac595773e1aecefa279536e80Optimized fragmentation schemes and data analysis strategies for proteome-wide cross-link identificationLiu, Fan; Loessl, Philip; Scheltema, Richard; Viner, Rosa; Heck, Albert J. R.Nature Communications (2017), 8 (), 15473CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)We describe optimized fragmentation schemes and data anal. strategies substantially enhancing the depth and accuracy in identifying protein cross-links using non-restricted whole proteome databases. These include a novel hybrid data acquisition strategy to sequence cross-links at both MS2 and MS3 level and a new algorithmic design XlinkX v2.0 for data anal. As proof-of-concept we investigated proteome-wide protein interactions in E. coli and HeLa cell lysates, resp., identifying 1,158 and 3,301 unique cross-links at ~ 1% false discovery rate. These protein interaction repositories provide meaningful structural information on many endogenous macromol. assemblies, as we showcase on several protein complexes involved in translation, protein folding and carbohydrate metab.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jproteome.8b00927.
Figure S1, Graphical workflow combining matrix and network activities; Figure S2, Organization of the new Perseus plugin API for networks; Figure S3, Context-specific documentation; and explanations of Tables S1–S3 (PDF)
Table S1, AP-MS pull screen (TXT)
Table S2, phosphoproteomics of EGF stimulation (TXT)
Table S3, clinical proteomics dataset (TXT)
Supplementary Data 1, Perseus network collection data format: example of a network collection describing three small, randomly generated networks (ZIP)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.