PRINCIPLES OF MATHEMATICAL MODELING OF microRNA-MEDIATED SIGNALING NETWORKS IN HUMAN DISEASES

Authors

DOI:

https://doi.org/10.11603/mie.1996-1960.2020.3.11610

Abstract

Background. Taking into account the growth of scientific knowledge and the discovery of new miRNAs and their gene targets, approaches to computational modeling of new functional associations between differentiated expression of miRNAs and diseases, as well as phenotypic patterns of miRNAs expression in gene regulatory networks. The goal of the conceptualization was the results of comparatively recent studies in systems biology, which use various kinetic modeling methods to help identify possibilities regarding the regulatory function and therapeutic potential of miRNAs in human diseases.

Materials and methods. Results. Some of the key computational aspects of mathematical modeling are also considered, which include: regulation of miRNAs-mediated network motifs in the regulation of gene expression; models of miRNAs biogenesis and miRNA target interactions; inclusion of such models in complex pathways of disease development, systemic understanding of their pathophysiological context. It is concluded that the efficiency and practicality of using small miRNA-associated network motifs, simplified to several components, to study the prognostic characteristics of the simulated network dynamics in diseases and physiological conditions. It is emphasized that most experimental studies focus on direct interactions of target miRNAs. Thus, the role of miRNAs in systems is revealed and, therefore, a systematic understanding of gene-mediated miRNA gene repression is provided.

Conclusions. However, in addition to the usual interactions of target miRNAs, recent experiments have shown that primary miRNAs or miRNA precursors formed during miRNA biogenesis can also compete with mature miRNAs for binding sites on target miRNAs. It is also important to move from the temporal dynamics of gene regulation by miRNAs, to the analysis and modeling of miRNA spatial information in cells as different subcellular locations.

References

Aushev V. N. (2015). Micro NRC: small molecules with high value. Clinical oncohematology, 8(1), 1-12. [In Ukrainian].

Mintser O. P., Zaliskyi V. M. (2019). Systems interaction micrornas in pathogenesis of cardiovascular diseases. Medical Informatics and Engineering, З, 4-19. doi: https://doi.org/10.11603/mie.1996-1960.2019.3.10428. [In Ukrainian].

Cora D., Re A., Cassele M. (2017). Micro-NRA-mediated regulatory chains: perspectives and prospects. Phys -boil, 14, 045001. [In Ukrainian].

Ha М., Кім V. N. (2014). Regulation of miRNA biogenesis. Nat. Rev. Mol. Cell Biol., 15, 509-524.

Herranz H., Cahen S. M. (2010). microRNA and gene regulatory networks imaging the impact of noise in biological systems. Genes Dev., 24 (1З), 1ЗЗ9-44.

Hwang H. W., Mendell J. T. (2007). MicroRNAs in cell proliferation, cell death, and tumorigenesis. Brit. J. Cancer., 96, suppl., R40-44.

Jolly M., Huang B., Lu M., Mani S., Levine H., Ben-Jacob E. (2014). Towards elucidating the connection between epithelial-mesenchymal transitions and sternness. J. R. Soc. Interface. 11:20140962.

Hye W. K., Crawford M., Muller F., Gerard N. et al. (2013). A Mathematical Model for MicroRNA in Lung Cancer. PLoS One., 8 (1), e53663.

Reena V. K., Subbaya S. (2014). Competing endogenous RNAs (ceRNAs): new entrants to the intricacies of gene regulation. Front. Genet., 5, 8.

Kim Y., Roh S., Lawler S. et al. (2011). MiR451 and AMPK Mutual Antagonism in Glioma Cell Migration and Proliferation: A Mathematical Model. PLoS One, 6 (12), e28293.

Lai X., Wolkenhauer O. et al. (2016). Understanding microRNA-mediated gene regulatory networks through mathematical modeling. Nucleic Acid Res., 44, 60196035.

Lai X., Bhattacharya A., Schmitz U. et al. (2013). A systems' biology approach to study microRNA-mediated gene regulatory networks. Biomed Research International, 17 Nov, 703849.

Landskroner-Eiger S., Moneke I., Sessa W. C. (2013). MiRNAs as modulators of angiogenesis. Cold Spring Harb Perspect Med., Feb 1; 3 (2), a006643. doi: 10.1101/cshperspect.a006643.

Lai X., Wolkenhauer O., Vera J. (2012). Modeling miRNA Regulation in Cancer Signaling Systems: miR-34a Regulation of the p53/Sirt1 Signaling Module. Methods in molecular biology (Clifton, N. J.), 880, 87-108.

Li J., Pandey V., Kessler T. (2012). Modeling of miRNA and Drug Action in the EGFR. Signaling Pathway January 11. https://doi.org/10.1371/journal. pone.0030140.

Lou W., Liu J., Gao Y. et al. (2017). MicroRNAs in cancer metastasis and angiogenesis. Oncotarget., Dec 29; 8(70), 115787-115802.

Lu M., Jolly M. K., Levine H. (2013). MicroRNA-based regulation of epithelial-hybrid-mesenchymal fate determination. Proc Natl Acad Sci USA,, Nov 5; 110 (45), 18144-9.

Paladini S., Fabris L., Bottai G. (2016). Targeting microRNAs as key modulators of tumor immune response. J Exp Clin Cancer Res., 35, 103.

Proctor C. J., Goljanek-Whysall K. (2017). Using computer simulation models to investigate the most promising microRNAs to improve muscle regeneration during ageing. Sci Rep., 7, 12314.

Proctor C. J., Smith G. R. (2017). Computer simulation models as a tool to investigate the role of microRNAs in osteoarthritis. PlOSONE, 12, ev187568.

Rupaimoole R., Slack F. G. (2017). MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nature Reviews Drug Discovery volume 16, 203-222.

Schuetz T. A., Becker S., Mang A. et al. (2013). Modelling of glioblastoma growth by linking a molecular interaction network with an agent-based model. 19, 417- 433.

Schmitz U., Meshkin H. N., Shailendra K. G. (2016). The RNA world in the 21st century-a systems approach to finding non-coding keys to clinical questions. Brief Bioinform., 17 (3), 380-92.

Shoval O., Alon U. (2010). SnapShot: network motifs. 143 (2), 326-e 1.

Suzuki H., Maruyama R., Yamamoto E. (2013). Epigenetic alteration and microRNA dysregulation in cancer.

Treiber T., Treiber N., Meister G. (2018). Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Rev Mol Cell Biol., 20, 5-20.

Vera J., Schmitz U., Lai X. (2013). Kinetic Modeling-Based Detection of Genetic Signatures That Provide Chemoresistance via the E2F1-p73/DNp73-miR-205 Network. Cancer Research, 73 (12).

Wang X., Li Y., Wang Y. (2010). Toward a system-level understanding of microRNA pathway via mathematical modeling. Biosystems, 100, 1.31-38.

Zhang H. M., Kuang S., Xiong X. (2015). Transcription factor and microRNA co-regulatory loops: important regulatory motifs in biological processes and diseases. Brief bioinform., 16, 1.45-58.

Zhao C., Isenberg J. S, Popel A. S. (2018). Human expression patterns: Qualitative and quantitative analysis of thrombospondin-1 under physiological and pathological conditions. Journal of Cellular and Molecular Medicine, 22 (Pt 3).

Zhao C., Isenberg J. S, Popel A. S. (2017). Transcriptional and Post-Transcriptional Regulation of Thrombospondin-1 Expression: A Computational Model. PLoS Comput Biol., 13 (1), e1005272.

Zhao C., Zhang Y., Popel A. S. (2019). Mechanistic Computational Models of MicroRNA-Mediated Signaling Networks in Human Diseases. J. Mol. Sci., 20 (2), 424.

Published

2021-08-11

How to Cite

Mintser, O. P., & Zaliskyi, V. M. . (2021). PRINCIPLES OF MATHEMATICAL MODELING OF microRNA-MEDIATED SIGNALING NETWORKS IN HUMAN DISEASES. Medical Informatics and Engineering, (3), 70–77. https://doi.org/10.11603/mie.1996-1960.2020.3.11610

Issue

Section

Articles