The current state of methods for gene regulatory networks reconstruction

Author(s) Collection number Pages Download abstract Download full text
Лях І. № 2 (63) 97-111 Image Image

The paper presents the analysis of current methods of gene regulatory networks (GRN) reconstruction on the basis of gene expressions data with the allocation of their advantages, disadvantages and ways of further improvement. The gene regulatory network is represented as both an oriented and non-oriented graph, where the weight of the arc determines the strength of the corresponding connection. Within the framework of this review, the following methods of gene regulatory networks reconstruction are considered: based on the analysis of correlations; based on the analysis of mutual information between genes and/or transcription factors; based on Bayesian networks; based on differential equations and based on regression analysis. The analysis of the relevant methods has allowed one to conclude that the current state of development of this subject area is determined by the hybridization of existing models, methods and algorithms. Currently, there is no effective information technology for gene regulatory network reconstruction, which takes into account the optimization of the network topology by estimating the distribution of topological parameters in synthetic and reconstructed networks, comparative analysis of different methods for gene regulatory networks reconstruction to form the optimal network topology. The use of ensembles of methods to reconstruct the gene regulatory networks helps to increase the adequacy of the reconstructed gene regulatory network relative to biological gene networks, which, in turn, contributes to a better understanding of the nature of the genes and/or transcription factors interaction in the network.

Keywords: gene regulatory network, an algorithm of gene regulatory network reconstruction, transcription factor, activating connection, inhibitory connection, oriented graph, undirected graph.

doi: 10.32403/1998-6912-2021-2-63-97-111


  • 1. Akers, K., & Murali, T. M. (2021). Gene regulatory network inference in single-cell biology. Current Opinion in Systems Biology, 26, 87–97 (in English).
  • 2. Liu, E., Li, L., & Cheng, L. (2019). Gene regulatory network revive. Encyclopedia of Bioinformatics and Computational Biology, 2, 155–164 (in English).
  • 3. Van den Broeck, L., Gordon, M., Inzé, D., Williams, C., & Sozzani, R. (2020). Gene Regulatory Network Inference: Connecting Plant Biology and Mathematical Modeling. Frontiers in Genetics, 11, art no 457 (in English).
  • 4. Wagner, A., Regev, A., & Yosef, N. (2016). Revealing the vectors of cellular identity with single-cell genomics. Natural Biotechnology,  34, 1145–1160 (in English).
  • 5. Marbach, D., Costello, J., & Küffner, R. et al. (2012). Wisdom of crowds for robust gene network inference. Natural Methods, 9, 796–804 (in English).
  • 6. Tang, F., Barbacioru, C., & Wang, Y. et al. (2009). mRNA-Seq whole-transcriptome analysis of a single cell. Natural Methods, 6, 377–382 (in English).
  • 7. Akers, K., & Murali, T. M. (2021). Gene regulatory network inference in single-cell biology. Current Opinion in Systems Biology, 26, 87–97 (in English).
  • 8. Trapnell, C., Cacchiarelli, D., & Grimsby, J. et al. (2014). The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Natural Biotechnology, 32, 381–386 (in English).
  • 9. Kim, S. (2015). ppcor: An R Package for a Fast Calculation to Semi-partial Correlation Coef­fi­cients. Communication for the Statistical Applications and Methods, 30, 665–674 (in English).
  • 10. Specht, A. T., & Jun, Li. (2017). LEAP: constructing gene co-expression networks for sing­le-cell RNA-sequencing data using pseudotime ordering. Bioinformatics, 33 (5), 764–766 (in English).
  • 11. Thomas, M. C., & Joy, A. T. (2006). Elements of Information Theory. Wiley, 2nd Edition (in English).
  • 12. Shannon, C. E. (1948). А mathematical theory of communication. Bell System Technical Jour­nal, 27, 379–423, 623–656 (in English).
  • 13. Chumak, O. V. (2011). Jentropii i fraktaky v analize dannyh. Moskva-Izhevsk, NIC «Reguljar­naja i haoticheskaja dinamika» (in Russian).
  • 14. Margolin, A. A., Nemenman, I., & Basso, K. et al. (2006). ARACNE: an algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinformatics, 7, 1–15 (in English).
  • 15. Qiu, X., Rahimzamani, A., & Wang, L. et al. (2020). Inferring causal gene regulatory networks from coupled single cell expression dynamics using Scribe. Cell Systems, 10, 265–274 (in English).
  • 16. Huynh-Thu, V. A., Irrthum, A., Wehenkel, L., & Geurts, P. (2010). Inferring Regulatory Networks from Expression Data Using Tree-Based Methods. PLoS ONE, 5 (9), art no e12776 (in English).
  • 17. Moerman, T., Aibar Santos, S., & Bravo González-Blas, C. et al. (2019). GRNBoost2 and Arboreto: efficient and scalable inference of gene regulatory networks. Bioinformatics, 35 (12), 2159–2161 (in English).
  • 18. Papili Gao, N., Minhaz Ud-Dean, S. M., Gandrillon, O., & Gunawan, R. (2018). SINCERITIES: inferring gene regulatory networks from time-stamped single cell transcriptional expression profiles. Bioinformatics, 34 (2), 258–266 (in English).
  • 19. Sanchez-Castillo, M., Blanco, D., & Tienda-Luna, I. M. et al. (2018). A Bayesian framework for the inference of gene regulatory networks from time and pseudo-time series data. Bioinformatics, 34 (6), 964–970 (in English).
  • 20. Sekula, M., Gaskins, J., & Datta, S. (2020). A sparse Bayesian factor model for the construction of gene co-expression networks from single-cell RNA sequencing count data. BMC Bioinformatics, 21, art no 361 (in English).
  • 21. Clarke, B. (2007). Information optimality and Bayesian modelling. Journal of Econometrics, 138 (2), 405–429 (in English).
  • 22. Friedman, N., Linial, M., Nachman, I., & Pe’er, D. (2000). Journal of Computational Biology, 7 (3–4), 601–620 (in English).
  • 23. Mukherjee, S., & Speed, T. P. (2008). Network inference using informative priors. Procee­dings of the National Academy of Sciences, 105 (38), 14313–14318 (in English).
  • 24. Chickering, D. M. (2002). Optimal structure identification with greedy search. Journal of Ma­chi­ne Learning Research, 3, 507–554 (in English).
  • 25. Liu, Z., Malone, B., & Yuan, C. (2012). Empirical evaluation of scoring functions for Baye­sian network model selection. BMC Bioinformatics, 13, art no S14 (in English).
  • 26. Hill, S. M., Heiser, L. M., & Cokelaer, T. et al. (2016). Inferring causal molecular networks: Empirical assessment through a community-based effort. Nat. Methods, 13 (4), art no 310 (in English).
  • 27. Yu, L., & Liu, H. (2004). Efficient feature selection via analysis of relevance and redundancy. Journal of Machine Learning Research, 5, 1205–1224 (in English).
  • 28. Murphy, K. P., & Russell, S. (2002). Dynamic Bayesian networks: Representation, inference and learning. Thesis of PhD dissertation (in English).
  • 29. Chen, T., He, H. L., & Church, G. M. (1999). Modeling gene expression with differential equations. Pacific Symposium on Biocomputing, 4, 29–40 (in English).
  • 30. D’Haeseleer, P., Wen, X., Fuhrman, S., & Somogyi, R. (1999). Linear modeling of mRNA expression levels during CNS development and injury. Pacific Symposium on Biocomputing, 4, 41–52 (in English).
  • 31. Matsumoto, H., Kiryu, H., & Furusawa, C. et al. (2017). SCODE: an efficient regulatory network inference algorithm from single-cell RNA-Seq during differentiation. Bioinformatics, 33, 2314–2321 (in English).
  • 32. Aubin-Frankowski, P. C., & Vert, J. P. (2020). Gene regulation inference from single-cell RNA-seq data with linear differential equations and velocity inference. Bioinformatics, 36, 4774–4780 (in English).
  • 33. Huynh-Thu, V. A., & Geurts, P. (2018). dynGENIE3: dynamical GENIE3 for the inference of gene networks from time series expression data. Science Report, 8, art no 3384 (in English).
  • 34. Aibar, S., González-Blas, C. B., & Moerman, T. et al. (2017). SCENIC: single-cell regulatory network inference and clustering. Natural Methods, 14, 1083–1086 (in English).
  • 35. Van de Sande, B., Flerin, C., & Davie, K. et al. (2020). A scalable SCENIC workflow for single-cell gene regulatory network analysis. Natural Protocols, 15, 2247–2276 (in English).