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.

  • Resource
  • Published:

Enabling single-cell trajectory network enrichment

Abstract

Single-cell sequencing (scRNA-seq) technologies allow the investigation of cellular differentiation processes with unprecedented resolution. Although powerful software packages for scRNA-seq data analysis exist, systems biology-based tools for trajectory analysis are rare and typically difficult to handle. This hampers biological exploration and prevents researchers from gaining deeper insights into the molecular control of developmental processes. Here, to address this, we have developed Scellnetor; a network-constraint time-series clustering algorithm. It allows extraction of temporal differential gene expression network patterns (modules) that explain the difference in regulation of two developmental trajectories. Using well-characterized experimental model systems, we demonstrate the capacity of Scellnetor as a hypothesis generator to identify putative mechanisms driving haematopoiesis or mechanistically interpretable subnetworks driving dysfunctional CD8 T-cell development in chronic infections. Altogether, Scellnetor allows for single-cell trajectory network enrichment, which effectively lifts scRNA-seq data analysis to a systems biology level.

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

Access options

Buy this article

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

Fig. 1: Workflow of Scellnetor.
Fig. 2: Computation of hyper-similarity matrix for gene module discovery.
Fig. 3: Comparison of neutrophil and erythrocyte differentiation trajectories.
Fig. 4: Differentiation of progenitor cells to dysfunctional CD8 T cells.

Similar content being viewed by others

Data availability

The scRNA-seq data used for the Scellnetor haematopoiesis analysis is from GEO (GSE72857). The scRNA-seq used for the clustering of exhausted CD8 T cells in chronic infections is also from GEO (GSE137007). Scellnetor results can be downloaded from ref. 53 and from GitLab.

Code availability

Scellnetor is freely available as an online tool at https://exbio.wzw.tum.de/scellnetor/ and can be downloaded as a standalone program from ref. 53 and from GitLab.

References

  1. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. https://doi.org/10.1186/s13059-019-1663-x (2019).

  2. Haghverdi, L., Büttner, M., Wolf, F. A., Buettner, F. & Theis, F. J. Diffusion pseudotime robustly reconstructs lineage branching. Nat. Methods https://doi.org/10.1038/nmeth.3971 (2016).

  3. Kiselev, V. Y., Andrews, T. S. & Hemberg, M. Challenges in unsupervised clustering of single-cell RNA-seq data. Nat. Rev. Genet. https://doi.org/10.1038/s41576-018-0088-9 (2019).

  4. Haghverdi, L., Buettner, F. & Theis, F. J. Diffusion maps for high-dimensional single-cell analysis of differentiation data. Bioinformatics https://doi.org/10.1093/bioinformatics/btv325 (2015).

  5. Tritschler, S. et al. Concepts and limitations for learning developmental trajectories from single cell genomics. Development https://doi.org/10.1242/dev.170506 (2019).

  6. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. https://doi.org/10.1186/s13059-017-1382-0 (2018).

  7. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies and species. Nat. Biotechnol. https://doi.org/10.1038/nbt.4096 (2018).

  8. Guo, M., Wang, H., Potter, S. S., Whitsett, J. A. & Xu, Y. SINCERA: a pipeline for single-cell RNA-Seq profiling analysis. PLoS Comput. Biol. https://doi.org/10.1371/journal.pcbi.1004575 (2015).

  9. Chen, G., Ning, B. & Shi, T. Single-cell RNA-seq technologies and related computational data analysis. Front. Genet. https://doi.org/10.3389/fgene.2019.00317 (2019).

  10. Kanev, K. et al. Proliferation-competent Tcf1+ CD8 T cells in dysfunctional populations are CD4 T cell help independent. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.1902701116 (2019).

  11. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. https://doi.org/10.1038/s41591-018-0045-3 (2018).

  12. Luecken, M. D. & Theis, F. J. Current best practices in single‐cell RNA‐seq analysis: a tutorial. Mol. Syst. Biol. https://doi.org/10.15252/msb.20188746 (2019).

  13. Hwang, B., Lee, J. H. & Bang, D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med. https://doi.org/10.1038/s12276-018-0071-8 (2018).

  14. Stegle, O., Teichmann, S. A. & Marioni, J. C. Computational and analytical challenges in single-cell transcriptomics. Nat. Rev. Genet. https://doi.org/10.1038/nrg3833 (2015).

  15. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell https://doi.org/10.1016/j.cell.2015.11.013 (2015).

  16. Campbell, K. R. & Yau, C. Switchde: inference of switch-like differential expression along single-cell trajectories. Bioinformatics https://doi.org/10.1093/bioinformatics/btw798 (2017).

  17. Matsumoto, H. et al. SCODE: an efficient regulatory network inference algorithm from single-cell RNA-seq during differentiation. Bioinformatics https://doi.org/10.1093/bioinformatics/btx194 (2017).

  18. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods https://doi.org/10.1038/nmeth.4463 (2017).

  19. Chan, T. E., Stumpf, M. P. H. & Babtie, A. C. Gene regulatory network inference from single-cell data using multivariate information measures. Cell Syst. https://doi.org/10.1016/j.cels.2017.08.014 (2017).

  20. Alcaraz, N. et al. De novo pathway-based biomarker identification. Nucleic Acids Res. https://doi.org/10.1093/nar/gkx642 (2017).

  21. Breitling, R., Amtmann, A. & Herzyk, P. Graph-based iterative group analysis enhances microarray interpretation. BMC Bioinformatics https://doi.org/10.1186/1471-2105-5-100 (2004).

  22. Ideker, T., Ozier, O., Schwikowski, B. & Siegel, A. F. Discovering regulatory and signalling circuits in molecular interaction networks. Bioinformatics https://doi.org/10.1093/bioinformatics/18.suppl_1.S233 (2002).

  23. Klimm, F. et al. Functional module detection through integration of single-cell RNA sequencing data with protein–protein interaction networks. BMC Genomics https://doi.org/10.1186/s12864-020-07144-2 (2020).

  24. Oughtred, R. et al. The BioGRID interaction database: 2019 update. Nucleic Acids Res. https://doi.org/10.1093/nar/gky1079 (2019).

  25. Jacomy, M., Venturini, T., Heymann, S. & Bastian, M. ForceAtlas2, a continuous graph layout algorithm for handy network visualization designed for the Gephi software. PLoS ONE https://doi.org/10.1371/journal.pone.0098679 (2014).

  26. Ribeiro, D. M. & Sonati, M. F. Regulation of human α-globin gene expression and α-thalassemia. Genet. Mol. Res. https://doi.org/10.4238/vol7-4gmr472 (2008).

  27. Shah, D. I. et al. Mitochondrial Atpif1 regulates haem synthesis in developing erythroblasts. Nature https://doi.org/10.1038/nature11536 (2012).

  28. Tanimura, A. et al. Mitochondrial activity and unfolded protein response are required for neutrophil differentiation. Cell. Physiol. Biochem. https://doi.org/10.1159/000491464 (2018).

  29. Michalak, M., Groenendyk, J., Szabo, E., Gold, L. I. & Opas, M. Calreticulin, a multi-process calcium-buffering chaperone of the endoplasmic reticulum. Biochem. J. https://doi.org/10.1042/BJ20081847 (2009).

  30. Sun, S. et al. Inhibition of prolyl 4-hydroxylase, beta polypeptide (P4HB) attenuates temozolomide resistance in malignant glioma via the endoplasmic reticulum stress response (ERSR) pathways. Neuro. Oncol. https://doi.org/10.1093/neuonc/not005 (2013).

  31. Vargas, A., Roux-Dalvai, F., Droit, A. & Lavoie, J. P. Neutrophil-derived exosomes: a new mechanism contributing to airway smooth muscle remodeling. Am. J. Resp. Cell Mol. Biol. https://doi.org/10.1165/rcmb.2016-0033OC (2016).

  32. Winterbourn, C. C., Kettle, A. J. & Hampton, M. B. Reactive oxygen species and neutrophil function. Annu. Rev. Biochem. https://doi.org/10.1146/annurev-biochem-060815-014442 (2016).

  33. Scapini, P. et al. CXCL1/macrophage inflammatory protein-2-induced angiogenesis in vivo is mediated by neutrophil-derived vascular endothelial growth factor-A. J. Immunol. https://doi.org/10.4049/jimmunol.172.8.5034 (2004).

  34. Gaudry, M. et al. Intracellular pool of vascular endothelial growth factor in human neutrophils. Blood https://doi.org/10.1182/blood.v90.10.4153 (1997).

  35. Scapini, P., Calzetti, F. & Cassatella, M. A. On the detection of neutrophil-derived vascular endothelial growth factor (VEGF). J. Immunol. Methods https://doi.org/10.1016/S0022-1759(99)00170-2 (1999).

  36. Jacob, C. O. et al. Lupus-associated causal mutation in neutrophil cytosolic factor 2 (NCF2) brings unique insights to the structure and function of NADPH oxidase. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.1113251108 (2012).

  37. Nauseef, W. M. Assembly of the phagocyte NADPH oxidase. Histochem. Cell Biol. https://doi.org/10.1007/s00418-004-0679-8 (2004).

  38. Groemping, Y. & Rittinger, K. Activation and assembly of the NADPH oxidase: a structural perspective. Biochem. J. https://doi.org/10.1042/BJ20041835 (2005).

  39. Liu, X. et al. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation. Nat. Cell Biol. https://doi.org/10.1038/ncb3527 (2017).

  40. Szentirmay, M. N. Survey and summary: spatial organization of RNA polymerase II transcription in the nucleus. Nucleic Acids Res. https://doi.org/10.1093/nar/28.10.2019 (2000).

  41. Wherry, E. J. T-cell exhaustion. Nat. Immunol. https://doi.org/10.1038/ni.2035 (2011).

  42. Hecht, I. et al. ILDR2 is a novel B7-like protein that negatively regulates T-cell responses. J. Immunol. https://doi.org/10.4049/jimmunol.1700325 (2018).

  43. Long, A. H. et al. 4-1BB costimulation ameliorates T-cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. https://doi.org/10.1038/nm.3838 (2015).

  44. Krishna, S. et al. Chronic activation of the kinase IKKβ impairs T-cell function and survival. J. Immunol. https://doi.org/10.4049/jimmunol.1102429 (2012).

  45. Peled, M. et al. EF hand domain family member D2 is required for T-cell cytotoxicity. J. Immunol. https://doi.org/10.4049/jimmunol.1800839 (2018).

  46. Lando, D.et al. FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev. https://doi.org/10.1101/gad.991402 (2002).

  47. Kim, J. W., Tchernyshyov, I., Semenza, G. L. & Dang, C. V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. https://doi.org/10.1016/j.cmet.2006.02.002 (2006).

  48. Papandreou, I., Cairns, R. A., Fontana, L., Lim, A. L. & Denko, N. C. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. https://doi.org/10.1016/j.cmet.2006.01.012 (2006).

  49. Doedens, A. L. et al. Hypoxia-inducible factors enhance the effector responses of CD8+ T cells to persistent antigen. Nat. Immunol. https://doi.org/10.1038/ni.2714 (2013).

  50. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. https://doi.org/10.21105/joss.00861 (2018).

  51. Klopfenstein, D. V. et al. GOATOOLS: a Python library for gene ontology analyses. Sci. Rep. https://doi.org/10.1038/s41598-018-28948-z (2018).

  52. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. https://doi.org/10.1038/ncomms14049 (2017).

  53. Grønning, A. G. B. Scellnetor_standalone_scripts_data (2021); https://doi.org/10.5281/ZENODO.4419550

Download references

Acknowledgements

J.B. and A.G.B.G. received funding from J.B.’s VILLUM Young Investigator grant no. 13154. The work of J.B. and T.K. was further funded by H2020 project RepoTrial (no. 777111). The work of R.R. and J.B. was partially funded by H2020 project FeatureCloud (no. 826078). J.B. and T.K. are grateful for financial support from BMBF project Sys_Care. M.O. is grateful for financial support from the Collaborative Research Center SFB924.

Author information

Authors and Affiliations

Authors

Contributions

A.G.B.G. developed and implemented the clustering algorithm of the Scellnetor tool. A.G.B.G. developed and implemented all basic backend functionalities of the webtool. A.G.B.G., J.L. and M.O. further developed the webtool. K.K., T.K. and D.Z. tested the webtool, provided critical feedback and, together with A.G.B.G., used Scellnetor to generate the biomedical results presented in the manuscript. All authors contributed equally to writing and improving the paper. A.G.B.G., J.B. and R.R. conceived the idea of the Scellnetor pipeline.

Corresponding authors

Correspondence to Alexander G. B. Grønning or Jan Baumbach.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Computational Science thanks Florian Klimm and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Yann Sweeney was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grønning, A.G.B., Oubounyt, M., Kanev, K. et al. Enabling single-cell trajectory network enrichment. Nat Comput Sci 1, 153–163 (2021). https://doi.org/10.1038/s43588-021-00025-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43588-021-00025-y

This article is cited by

Search

Quick links

Nature Briefing AI and Robotics

Sign up for the Nature Briefing: AI and Robotics newsletter — what matters in AI and robotics research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: AI and Robotics