Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap

Abstract

Pathway enrichment analysis helps researchers gain mechanistic insight into gene lists generated from genome-scale (omics) experiments. This method identifies biological pathways that are enriched in a gene list more than would be expected by chance. We explain the procedures of pathway enrichment analysis and present a practical step-by-step guide to help interpret gene lists resulting from RNA-seq and genome-sequencing experiments. The protocol comprises three major steps: definition of a gene list from omics data, determination of statistically enriched pathways, and visualization and interpretation of the results. We describe how to use this protocol with published examples of differentially expressed genes and mutated cancer genes; however, the principles can be applied to diverse types of omics data. The protocol describes innovative visualization techniques, provides comprehensive background and troubleshooting guidelines, and uses freely available and frequently updated software, including g:Profiler, Gene Set Enrichment Analysis (GSEA), Cytoscape and EnrichmentMap. The complete protocol can be performed in ~4.5 h and is designed for use by biologists with no prior bioinformatics training.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Protocol overview.
Fig. 2: Screenshot of g:Profiler user interface.
Fig. 3: Screenshot of GSEA user interface.
Fig. 4: GSEA output overview.
Fig. 5: Class/phenotype-specific GSEA output.
Fig. 6: Screenshot of the EnrichmentMap software user interface.
Fig. 7: Resulting enrichment maps (no manual formatting).
Fig. 8: Overview of EnrichmentMap panels in Cytoscape.
Fig. 9: Example heat map in EnrichmentMap.
Fig. 10: Resulting publication-ready enrichment map.
Fig. 11: Collapsed enrichment map.
Fig. 12: Subnetwork example.
Fig. 13: Generic enrichment map legend.

Similar content being viewed by others

Data availability

The protocol uses publicly available software packages (GSEA v.3.0 or higher, g:Profiler, Enrichment Map v.3.0 or higher, Cytoscape v.3.6.0 or higher) and custom R scripts that apply publicly available R packages (edgeR, Roast, Limma, Camera). Custom scripts are available in the Supplementary Protocols and at our GitHub web sites (https://github.com/BaderLab/Cytoscape_workflows/tree/master/EnrichmentMapPipeline and https://baderlab.github.io/Cytoscape_workflows/EnrichmentMapPipeline/index.html).

References

  1. Lander, E. S. Initial impact of the sequencing of the human genome. Nature 470, 187–197 (2011).

    CAS  PubMed  Google Scholar 

  2. Stephens, Z. D. et al. Big data: astronomical or genomical? PLoS Biol. 13, e1002195 (2015).

    PubMed  PubMed Central  Google Scholar 

  3. Mack, S. C. et al. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature 506, 445–450 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Pinto, D. et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature 466, 368–372 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Pinto, D. et al. Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am. J. Hum. Genet. 94, 677–694 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Verhaak, R. G. et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J. Clin. Invest. 123, 517–525 (2013).

    CAS  PubMed  Google Scholar 

  8. The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

    PubMed Central  Google Scholar 

  9. Cline, M. S. et al. Integration of biological networks and gene expression data using Cytoscape. Nat. Protoc. 2, 2366–2382 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Creixell, P. et al. Pathway and network analysis of cancer genomes. Nat Methods 12, 615–621 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wadi, L., Meyer, M., Weiser, J., Stein, L. D. & Reimand, J. Impact of outdated gene annotations on pathway enrichment analysis. Nat. Methods 13, 705–706 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Reyna, M. A. et al. Pathway and network analysis of more than 2,500 whole cancer genomes. Preprint at https://www.biorxiv.org/content/early/2018/08/07/385294 (2018).

  13. Reimand, J. et al. g:Profiler-a web server for functional interpretation of gene lists (2016 update). Nucleic Acids Res. 44, W83–89 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G. D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS ONE 5, e13984 (2010).

    PubMed  PubMed Central  Google Scholar 

  17. Anders, S. et al. Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat. Protoc. 8, 1765–1786 (2013).

    PubMed  Google Scholar 

  18. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  19. Silva, T. S. & Richard, N. Visualization and differential analysis of protein expression data using R. Methods Mol. Biol. 1362, 105–118 (2016).

    CAS  PubMed  Google Scholar 

  20. Schubert, O. T., Rost, H. L., Collins, B. C., Rosenberger, G. & Aebersold, R. Quantitative proteomics: challenges and opportunities in basic and applied research. Nat. Protoc. 12, 1289–1294 (2017).

    CAS  PubMed  Google Scholar 

  21. MacArthur, D. G. et al. Guidelines for investigating causality of sequence variants in human disease. Nature 508, 469–476 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Gonzalez-Perez, A. et al. Computational approaches to identify functional genetic variants in cancer genomes. Nat. Methods 10, 723–729 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Yang, H. & Wang, K. Genomic variant annotation and prioritization with ANNOVAR and wANNOVAR. Nat. Protoc. 10, 1556–1566 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Assenov, Y. et al. Comprehensive analysis of DNA methylation data with RnBeads. Nat. Methods 11, 1138–1140 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Laird, P. W. Principles and challenges of genomewide DNA methylation analysis. Nat. Rev. Genet. 11, 191–203 (2010).

    CAS  PubMed  Google Scholar 

  26. Rapaport, F. et al. Comprehensive evaluation of differential gene expression analysis methods for RNA-seq data. Genome Biol. 14, R95 (2013).

    PubMed  PubMed Central  Google Scholar 

  27. Conesa, A. et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 17, 13 (2016).

    PubMed  PubMed Central  Google Scholar 

  28. Bullard, J. H., Purdom, E., Hansen, K. D. & Dudoit, S. Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinformatics 11, 94 (2010).

    PubMed  PubMed Central  Google Scholar 

  29. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  30. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Smyth, G. K. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol 3, Article3 (2004).

    PubMed  Google Scholar 

  32. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    PubMed  PubMed Central  Google Scholar 

  33. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Hochberg, Y. & Benjamini, Y. More powerful procedures for multiple significance testing. Stat. Med. 9, 811–818 (1990).

    CAS  PubMed  Google Scholar 

  35. Chen, J., Bardes, E. E., Aronow, B. J. & Jegga, A. G. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 37, W305–W311 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    CAS  Google Scholar 

  37. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Mi, H., Muruganujan, A. & Thomas, P. D. PANTHER in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res. 41, D377–D386 (2013).

    CAS  PubMed  Google Scholar 

  39. Reimand, J., Kull, M., Peterson, H., Hansen, J. & Vilo, J. g:Profiler—a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res. 35, W193–W200 (2007).

    PubMed  PubMed Central  Google Scholar 

  40. Bindea, G. et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25, 1091–1093 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Maere, S., Heymans, K. & Kuiper, M. BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics 21, 3448–3449 (2005).

    Google Scholar 

  42. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10, 48 (2009).

    PubMed  PubMed Central  Google Scholar 

  43. Wang, J., Duncan, D., Shi, Z. & Zhang, B. WEB-based GEne SeT AnaLysis Toolkit (WebGestalt): update 2013. Nucleic Acids Res. 41, W77–W83 (2013).

    PubMed  PubMed Central  Google Scholar 

  44. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  45. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    PubMed  Google Scholar 

  46. Fabregat, A. et al. The Reactome pathway Knowledgebase. Nucleic Acids Res. 46, D649–D655 (2018).

    CAS  PubMed  Google Scholar 

  47. Kanehisa, M., Goto, S., Sato, Y., Furumichi, M. & Tanabe, M. KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40, D109–D114 (2012).

    CAS  PubMed  Google Scholar 

  48. Kelder, T. et al. WikiPathways: building research communities on biological pathways. Nucleic Acids Res. 40, D1301–D1307 (2012).

    CAS  PubMed  Google Scholar 

  49. Kutmon, M. et al. PathVisio 3: an extendable pathway analysis toolbox. PLoS Comput. Biol. 11, e1004085 (2015).

    PubMed  PubMed Central  Google Scholar 

  50. Szklarczyk, D. et al. STRINGv10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43, D447–D452 (2015).

    CAS  PubMed  Google Scholar 

  51. Warde-Farley, D. et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38, W214–W220 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lechman, E. R. et al. Attenuation of miR-126 activity expands HSC in vivo without exhaustion. Cell Stem Cell 11, 799–811 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Jhas, B. et al. Metabolic adaptation to chronic inhibition of mitochondrial protein synthesis in acute myeloid leukemia cells. PLoS ONE 8, e58367 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Ballouz, S., Pavlidis, P. & Gillis, J. Using predictive specificity to determine when gene set analysis is biologically meaningful. Nucleic Acids Res. 45, e20 (2017).

    PubMed  Google Scholar 

  55. Krzywinski, M. & Altman, N. Power and sample size. Nat. Methods 10, 1139–1140 (2013).

    CAS  Google Scholar 

  56. Liu, Y., Zhou, J. & White, K. P. RNA-seq differential expression studies: more sequence or more replication? Bioinformatics 30, 301–304 (2014).

    CAS  PubMed  Google Scholar 

  57. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Fabregat, A. et al. The Reactome pathway Knowledgebase. Nucleic Acids Res. 44, D481–D487 (2016).

    CAS  PubMed  Google Scholar 

  59. Caspi, R. et al. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Res. 44, D471–D480 (2016).

    CAS  PubMed  Google Scholar 

  60. Kandasamy, K. et al. NetPath: a public resource of curated signal transduction pathways. Genome Biol. 11, R3 (2010).

    PubMed  PubMed Central  Google Scholar 

  61. Rhee, S. Y., Wood, V., Dolinski, K. & Draghici, S. Use and misuse of the gene ontology annotations. Nat. Rev. Genet. 9, 509–515 (2008).

    CAS  PubMed  Google Scholar 

  62. Skunca, N., Altenhoff, A. & Dessimoz, C. Quality of computationally inferred gene ontology annotations. PLoS Comput. Biol. 8, e1002533 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Wojtowicz, E. E. et al. Ectopic miR-125a expression induces long-term repopulating stem cell capacity in mouse and human hematopoietic progenitors. Cell Stem Cell 19, 383–396 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Tong, J. et al. Integrated analysis of proteome, phosphotyrosine-proteome, tyrosine-kinome, and tyrosine-phosphatome in acute myeloid leukemia. Proteomics 17, 1600361 (2017).

  65. Kamdar, S. N. et al. Dynamic interplay between locus-specific DNA methylation and hydroxymethylation regulates distinct biological pathways in prostate carcinogenesis. Clin. Epigenetics 8, 32 (2016).

    PubMed  PubMed Central  Google Scholar 

  66. Liu, Y. et al. Metabolomic profiling in liver of adiponectin-knockout mice uncovers lysophospholipid metabolism as an important target of adiponectin action. Biochem. J. 469, 71–82 (2015).

    CAS  PubMed  Google Scholar 

  67. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Raychaudhuri, S. et al. Accurately assessing the risk of schizophrenia conferred by rare copy-number variation affecting genes with brain function. PLoS Genet. 6, e1001097 (2010).

    PubMed  PubMed Central  Google Scholar 

  69. Lee, P. H., O’Dushlaine, C., Thomas, B. & Purcell, S. M. INRICH: interval-based enrichment analysis for genome-wide association studies. Bioinformatics 28, 1797–1799 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Khatri, P., Sirota, M. & Butte, A. J. Ten years of pathway analysis: current approaches and outstanding challenges. PLoS Comput. Biol. 8, e1002375 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Wu, D. & Smyth, G. K. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 40, e133 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Young, M. D., Wakefield, M. J., Smyth, G. K. & Oshlack, A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol. 11, R14 (2010).

    PubMed  PubMed Central  Google Scholar 

  73. Gu, Z. & Wang, J. CePa: an R package for finding significant pathways weighted by multiple network centralities. Bioinformatics 29, 658–660 (2013).

    CAS  PubMed  Google Scholar 

  74. Fang, Z., Tian, W. & Ji, H. A network-based gene-weighting approach for pathway analysis. Cell Res. 22, 565–580 (2012).

    CAS  PubMed  Google Scholar 

  75. Farfan, F., Ma, J., Sartor, M. A., Michailidis, G. & Jagadish, H. V. THINK Back: KNowledge-based Interpretation of High Throughput data. BMC Bioinformatics 13(Suppl. 2), S4 (2012).

    PubMed  PubMed Central  Google Scholar 

  76. Tarca, A. L. et al. A novel signaling pathway impact analysis. Bioinformatics 25, 75–82 (2009).

    CAS  PubMed  Google Scholar 

  77. Draghici, S. et al. A systems biology approach for pathway level analysis. Genome Res. 17, 1537–1545 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Glaab, E., Baudot, A., Krasnogor, N., Schneider, R. & Valencia, A. EnrichNet: network-based gene set enrichment analysis. Bioinformatics 28, i451–i457 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Schaefer, C. F. et al. PID: the Pathway Interaction Database. Nucleic Acids Res. 37, D674–D679 (2009).

    CAS  PubMed  Google Scholar 

  80. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bader, G. D., Cary, M. P. & Sander, C. Pathguide: a pathway resource list. Nucleic Acids Res. 34, D504–D506 (2006).

    CAS  PubMed  Google Scholar 

  83. Kanehisa, M., Furumichi, M., Tanabe, M., Sato, Y. & Morishima, K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45, D353–D361 (2017).

    CAS  PubMed  Google Scholar 

  84. Tavazoie, S., Hughes, J. D., Campbell, M. J., Cho, R. J. & Church, G. M. Systematic determination of genetic network architecture. Nat. Genet. 22, 281–285 (1999).

    CAS  PubMed  Google Scholar 

  85. Goeman, J. J. & Bühlmann, P. Analyzing gene expression data in terms of gene sets: methodological issues. Bioinformatics 23, 980–987 (2007).

    CAS  PubMed  Google Scholar 

  86. Bansal, V., Libiger, O., Torkamani, A. & Schork, N. J. Statistical analysis strategies for association studies involving rare variants. Nat. Rev. Genet. 11, 773–785 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are grateful to J. Mesirov for comments on the manuscript. This project was supported by an Investigator Award to J.R. from the Ontario Institute for Cancer Research through funding from the Government of Ontario and by a Natural Sciences and Engineering Research Council of Canada (NSERC) Discovery Grant to J.R. (RGPIN-2016-06485). This work was supported by US National Institutes of Health grants P41 GM103504, R01 GM070743, U41 HG006623 and R01 CA121941 to G.D.B.

Author information

Authors and Affiliations

Authors

Contributions

J.R., R.I., V.V., A.R., D.M. and G.D.B. wrote the manuscript. R.I. created the step-by-step protocols, figures, R scripts and R notebooks, except for g:Profiler (J.R.). M.K. and C.T.-L. developed EnrichmentMap 3.0 and AutoAnnotate Cytoscape applications. L.W., M.M., J.W., C.X. and V.V. tested the protocol. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Gary D. Bader.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Pinto, D. et al. Nature 466, 368–372 (2010): https://doi.org/10.1038/nature09146

Pajtler, K. W. et al. Cancer Cell 27, P728–P743 (2015): https://doi.org/10.1016/j.ccell.2015.04.002

Cavalli, F. M. G. et al. Cancer Cell 31, P737–P754 (2017): https://doi.org/10.1016/j.ccell.2017.05.005

Supplementary information

Supplementary Tables and Methods

Supplementary Tables 1–13 and Supplementary Protocols 1–4

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reimand, J., Isserlin, R., Voisin, V. et al. Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat Protoc 14, 482–517 (2019). https://doi.org/10.1038/s41596-018-0103-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0103-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research