SYSTEMS BIOLOGY: ESSENTIAL PRINCIPLES AND OMICs APPROACHES

R. Stoika


DOI: http://dx.doi.org/10.30970/sbi.1401.609

Abstract


The systems biology is a new branch of modern biology aimed at studying various systems existing in nature on the level of their biological components such as biomolecules (ex. DNA, RNA, proteins, carbohydrates, lipids, etc.), cells and organelles, tissues and organs, organisms, or species. It differs from the synthetic biology aimed at the creation and study of the biological systems that do not exist in nature for: a) development and combining different functional modules in one system, b) better understanding of processes taking place in the living organisms, c) development of new methods for influencing functions of the living organisms. These two branches of modern biology have similar methodological approaches based on the achievements of molecular bio­logy and bioinformatics. The behavior of different systems in the living organisms is very compleх, however, it might be defined using specific approaches of the systems bio­logy based on studying qualitative and quantitative characteristics of all components of a specific biological system through the OMICS approaches. In this review, specific OMICS for various biological systems are briefly described. The application of the systems bio­logy and the OMICS approaches demonstrated their great potentials for medicine, pharmaceutics, biotechnology, namely for the development of “smart” biomaterials.


Keywords


systems biology, synthetic biology, OMICS approaches

Full Text:

PDF

References


1. Al-Mufti F., Kim M., Dodson V., Sursal T., Bowers C., Cole C., Scurlock C., Becker C., Gandhi C., Mayer S.A. Machine Learning and Artificial Intelligence in Neurocritical Care: a Specialty-Wide Disruptive Transformation or a Strategy for Success. Curr Neurol Neurosci Rep, 2019; 19(11): 89.
CrossrefPubMedGoogle Scholar

2. Anderson N.G. Proteome and proteomics: new technologies, new concepts, and new words. Electrophoresis, 1998; 19(11): 1853-1861
CrossrefPubMedGoogle Scholar

3. Aslam B., Basit M., Nisar M.A., Khurshid M., Rasool M.H. Proteomics: Technologies and Their Applications. J. Chromatogr. Sci., 2017; 55(2): 182-196.
CrossrefPubMedGoogle Scholar

4. Beard D.A.,Neal M.L., Tabesh-Saleki N., Thompson C.T., Bassingthwaighte J.B., Shimoyama M., Carlson B.E. Multiscale modeling and data integration in the virtual physiological rat project. Ann. Biomed. Eng., 2012; 40(11): 2365-78.
CrossrefPubMedGoogle Scholar

5. Broekema R.V., Bakker O.B., Jonkers I.H. A practical view of fine-mapping and gene prioritization in the post-genome-wide association era. Open Biol., 2020; 10(1): 190221.
CrossrefPubMedGoogle Scholar

6. Costa A., Franco O.L. Impact and influence of "omics" technology on hypertension studies. Int. J. Cardiol., 2017; 228: 1022-1034.
CrossrefPubMedGoogle Scholar

7 Filyak Y.Z., Filyak O.S., Souchelnitskyi S.I., Stoika R.S. Proteomics of activation of murine T lymphocytes with pttg-knockout. Reports of the National Academy of Sciences of Ukraine, 2007, N 5, P. 172-179.
http://nbuv.gov.ua/UJRN/dnanu_2007_5_31

8. Haug K., Salek R.M., Steinbeck C. Global open data management in metabolomics. Curr. Opin. Chem. Biol., 2017; 36: 58-63.
CrossrefPubMedGoogle Scholar

9. Houdebine L.-M. Design of Vectors for optimizing transgene expression. In Chapter 17. Transgenic Animal Technology (Third Edition), 2014: 489-511.
CrossrefGoogle Scholar

10. Joyce A.R. Palsson B.Ø. The model organism as a system: integrating "omics" data sets. Nature Reviews in Molecular Cell Biology, 2006; 7(3): 198-210.
CrossrefPubMedGoogle Scholar

11. Kashchak N., Tsaryk R., Stoika R. Bystander effect of normal fibroblasts for macrophages co-cultured with susceptible transformed target cells. Cell Biol Int, 2005; 29(1): 41-50.
CrossrefPubMedGoogle Scholar

12. Lander E.S., Linton L.M., Birren B. et al. Initial sequencing and analysis of the human genome. Nature, 2001; 409(6822): 860-921.
CrossrefPubMedGoogle Scholar

13. Latorre Amparo and Silva Francisco J. The size of the genome and the complexity of living beings. Metode 32, 2013.

14. Mammalian Gene Collection (MGC) Program Team. Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences. Proc. Nat. Acad. Sci. USA, 2002; 99(26): 16899-16903.
CrossrefPubMedGoogle Scholar

15. Martens L., Vizcaíno J.A. A golden age for working with public proteomics data. Trends Biochem. Sci., 2017; 42(5): 333-341.
CrossrefPubMedGoogle Scholar

16. Menni C, Zierer J, Valdes AM, Spector TD. Mixing omics: combining genetics and metabolomics to study rheumatic diseases. Nat. Rev. Rheumatol., 2017; 13(3): 174-181.
CrossrefPubMedGoogle Scholar

17. Millius A., Ueda H.R. Systems biology-derived discoveries of intrinsic clocks. Front. Neurol., 2017; 8(25): 1-19.
CrossrefPubMedGoogle Scholar

18. Mocellin S. Provenzano M. RNA interference: learning gene knock-down from cell physiology. J. Transl. Med., 2004; 2: 39.
CrossrefPubMedGoogle Scholar

19. Prelich G. Gene Overexpression: Uses, Mechanisms, and Interpretation. Genetics, 2012; 190(3): 841-854.
CrossrefPubMedGoogle Scholar

20. Saccone C., Pesole G. Handbook of Comparative Genomics: Principles and Methodology / John Wiley and Sons. 2003. Chapter 2. Eukaryotes. P. 36-84.
CrossrefGoogle Scholar

21. Schneider М.V., Orchard S. Omics technologies, data and bioinformatics principles. Chapter 1 In: Mayer B. (Ed.) Bioinformatics for OMICs data: Methods and protocols, Methods in molecular biology. Springer Science+Business Media, LLC, 2011; 719: 3-30.
CrossrefPubMedGoogle Scholar

22. Silverman R.B., Holladay M.W. The organic chemistry of drug design and drug action. 3rd Edition. Elsevier - Academic Press, Amsterdam, Boston, Heidelberg et al. 2014: 536 p. In Chapter 3: p.125. eBook ISBN: 9780123820310
Google Scholar


Refbacks

  • There are currently no refbacks.


Copyright (c) 2020 R. Stoika

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.