1
|
Nourisa J, Passemiers A, Shakeri F, Omidi M, Helmholz H, Raimondi D, Moreau Y, Tomforde S, Schlüter H, Luthringer-Feyerabend B, Cyron CJ, Aydin RC, Willumeit-Römer R, Zeller-Plumhoff B. Gene regulatory network analysis identifies MYL1, MDH2, GLS, and TRIM28 as the principal proteins in the response of mesenchymal stem cells to Mg 2+ ions. Comput Struct Biotechnol J 2024; 23:1773-1785. [PMID: 38689715 PMCID: PMC11058716 DOI: 10.1016/j.csbj.2024.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
Magnesium (Mg)-based implants have emerged as a promising alternative for orthopedic applications, owing to their bioactive properties and biodegradability. As the implants degrade, Mg2+ ions are released, influencing all surrounding cell types, especially mesenchymal stem cells (MSCs). MSCs are vital for bone tissue regeneration, therefore, it is essential to understand their molecular response to Mg2+ ions in order to maximize the potential of Mg-based biomaterials. In this study, we conducted a gene regulatory network (GRN) analysis to examine the molecular responses of MSCs to Mg2+ ions. We used time-series proteomics data collected at 11 time points across a 21-day period for the GRN construction. We studied the impact of Mg2+ ions on the resulting networks and identified the key proteins and protein interactions affected by the application of Mg2+ ions. Our analysis highlights MYL1, MDH2, GLS, and TRIM28 as the primary targets of Mg2+ ions in the response of MSCs during 1-21 days phase. Our results also identify MDH2-MYL1, MDH2-RPS26, TRIM28-AK1, TRIM28-SOD2, and GLS-AK1 as the critical protein relationships affected by Mg2+ ions. By offering a comprehensive understanding of the regulatory role of Mg2+ ions on MSCs, our study contributes valuable insights into the molecular response of MSCs to Mg-based materials, thereby facilitating the development of innovative therapeutic strategies for orthopedic applications.
Collapse
Affiliation(s)
- Jalil Nourisa
- Institute of Material Systems Modeling, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | | | - Farhad Shakeri
- Institute of Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Maryam Omidi
- Institute of Clinical Chemistry/Central Laboratories, University Medical Center Hamburg, Hamburg, Germany
| | - Heike Helmholz
- Institute of Metallic Biomaterials, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | | | | | - Sven Tomforde
- Department of Computer Science, Intelligent Systems, University of Kiel, Kiel, Germany
| | - Hartmuth Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine Diagnostic Center, University of Hamburg, Hamburg, Germany
| | | | - Christian J. Cyron
- Institute of Material Systems Modeling, Helmholtz Zentrum Hereon, Geesthacht, Germany
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany
| | - Roland C. Aydin
- Institute of Material Systems Modeling, Helmholtz Zentrum Hereon, Geesthacht, Germany
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany
| | | | | |
Collapse
|
2
|
Unger Avila P, Padvitski T, Leote AC, Chen H, Saez-Rodriguez J, Kann M, Beyer A. Gene regulatory networks in disease and ageing. Nat Rev Nephrol 2024; 20:616-633. [PMID: 38867109 DOI: 10.1038/s41581-024-00849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
The precise control of gene expression is required for the maintenance of cellular homeostasis and proper cellular function, and the declining control of gene expression with age is considered a major contributor to age-associated changes in cellular physiology and disease. The coordination of gene expression can be represented through models of the molecular interactions that govern gene expression levels, so-called gene regulatory networks. Gene regulatory networks can represent interactions that occur through signal transduction, those that involve regulatory transcription factors, or statistical models of gene-gene relationships based on the premise that certain sets of genes tend to be coexpressed across a range of conditions and cell types. Advances in experimental and computational technologies have enabled the inference of these networks on an unprecedented scale and at unprecedented precision. Here, we delineate different types of gene regulatory networks and their cell-biological interpretation. We describe methods for inferring such networks from large-scale, multi-omics datasets and present applications that have aided our understanding of cellular ageing and disease mechanisms.
Collapse
Affiliation(s)
- Paula Unger Avila
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tsimafei Padvitski
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ana Carolina Leote
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - He Chen
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Martin Kann
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas Beyer
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
3
|
Huo Q, Song R, Ma Z. Recent advances in exploring transcriptional regulatory landscape of crops. FRONTIERS IN PLANT SCIENCE 2024; 15:1421503. [PMID: 38903438 PMCID: PMC11188431 DOI: 10.3389/fpls.2024.1421503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024]
Abstract
Crop breeding entails developing and selecting plant varieties with improved agronomic traits. Modern molecular techniques, such as genome editing, enable more efficient manipulation of plant phenotype by altering the expression of particular regulatory or functional genes. Hence, it is essential to thoroughly comprehend the transcriptional regulatory mechanisms that underpin these traits. In the multi-omics era, a large amount of omics data has been generated for diverse crop species, including genomics, epigenomics, transcriptomics, proteomics, and single-cell omics. The abundant data resources and the emergence of advanced computational tools offer unprecedented opportunities for obtaining a holistic view and profound understanding of the regulatory processes linked to desirable traits. This review focuses on integrated network approaches that utilize multi-omics data to investigate gene expression regulation. Various types of regulatory networks and their inference methods are discussed, focusing on recent advancements in crop plants. The integration of multi-omics data has been proven to be crucial for the construction of high-confidence regulatory networks. With the refinement of these methodologies, they will significantly enhance crop breeding efforts and contribute to global food security.
Collapse
Affiliation(s)
| | | | - Zeyang Ma
- State Key Laboratory of Maize Bio-breeding, Frontiers Science Center for Molecular Design Breeding, Joint International Research Laboratory of Crop Molecular Breeding, National Maize Improvement Center, College of Agronomy and Biotechnology, China Agricultural University, Beijing, China
| |
Collapse
|
4
|
Guo C, Huang Z, Chen J, Yu G, Wang Y, Wang X. Identification of Novel Regulators of Leaf Senescence Using a Deep Learning Model. PLANTS (BASEL, SWITZERLAND) 2024; 13:1276. [PMID: 38732491 PMCID: PMC11085074 DOI: 10.3390/plants13091276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Deep learning has emerged as a powerful tool for investigating intricate biological processes in plants by harnessing the potential of large-scale data. Gene regulation is a complex process that transcription factors (TFs), cooperating with their target genes, participate in through various aspects of biological processes. Despite its significance, the study of gene regulation has primarily focused on a limited number of notable instances, leaving numerous aspects and interactions yet to be explored comprehensively. Here, we developed DEGRN (Deep learning on Expression for Gene Regulatory Network), an innovative deep learning model designed to decipher gene interactions by leveraging high-dimensional expression data obtained from bulk RNA-Seq and scRNA-Seq data in the model plant Arabidopsis. DEGRN exhibited a compared level of predictive power when applied to various datasets. Through the utilization of DEGRN, we successfully identified an extensive set of 3,053,363 high-quality interactions, encompassing 1430 TFs and 13,739 non-TF genes. Notably, DEGRN's predictive capabilities allowed us to uncover novel regulators involved in a range of complex biological processes, including development, metabolism, and stress responses. Using leaf senescence as an example, we revealed a complex network underpinning this process composed of diverse TF families, including bHLH, ERF, and MYB. We also identified a novel TF, named MAF5, whose expression showed a strong linear regression relation during the progression of senescence. The mutant maf5 showed early leaf decay compared to the wild type, indicating a potential role in the regulation of leaf senescence. This hypothesis was further supported by the expression patterns observed across four stages of leaf development, as well as transcriptomics analysis. Overall, the comprehensive coverage provided by DEGRN expands our understanding of gene regulatory networks and paves the way for further investigations into their functional implications.
Collapse
Affiliation(s)
| | | | | | | | | | - Xu Wang
- Shanghai Collaborative Innovation Center of Agri-Seeds, Joint Center for Single Cell Biology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (C.G.); (Z.H.); (J.C.); (G.Y.); (Y.W.)
| |
Collapse
|
5
|
Wang Y, Chen X, Zheng Z, Huang L, Xie W, Wang F, Zhang Z, Wong KC. scGREAT: Transformer-based deep-language model for gene regulatory network inference from single-cell transcriptomics. iScience 2024; 27:109352. [PMID: 38510148 PMCID: PMC10951644 DOI: 10.1016/j.isci.2024.109352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/29/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Gene regulatory networks (GRNs) involve complex and multi-layer regulatory interactions between regulators and their target genes. Precise knowledge of GRNs is important in understanding cellular processes and molecular functions. Recent breakthroughs in single-cell sequencing technology made it possible to infer GRNs at single-cell level. Existing methods, however, are limited by expensive computations, and sometimes simplistic assumptions. To overcome these obstacles, we propose scGREAT, a framework to infer GRN using gene embeddings and transformer from single-cell transcriptomics. scGREAT starts by constructing gene expression and gene biotext dictionaries from scRNA-seq data and gene text information. The representation of TF gene pairs is learned through optimizing embedding space by transformer-based engine. Results illustrated scGREAT outperformed other contemporary methods on benchmarks. Besides, gene representations from scGREAT provide valuable gene regulation insights, and external validation on spatial transcriptomics illuminated the mechanism behind scGREAT annotation. Moreover, scGREAT identified several TF target regulations corroborated in studies.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Xingjian Chen
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zetian Zheng
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Lei Huang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Weidun Xie
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Fuzhou Wang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Zhaolei Zhang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
6
|
Murphy KA, Bassett DS. Information decomposition in complex systems via machine learning. Proc Natl Acad Sci U S A 2024; 121:e2312988121. [PMID: 38498714 PMCID: PMC10990158 DOI: 10.1073/pnas.2312988121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/30/2024] [Indexed: 03/20/2024] Open
Abstract
One of the fundamental steps toward understanding a complex system is identifying variation at the scale of the system's components that is most relevant to behavior on a macroscopic scale. Mutual information provides a natural means of linking variation across scales of a system due to its independence of functional relationship between observables. However, characterizing the manner in which information is distributed across a set of observables is computationally challenging and generally infeasible beyond a handful of measurements. Here, we propose a practical and general methodology that uses machine learning to decompose the information contained in a set of measurements by jointly optimizing a lossy compression of each measurement. Guided by the distributed information bottleneck as a learning objective, the information decomposition identifies the variation in the measurements of the system state most relevant to specified macroscale behavior. We focus our analysis on two paradigmatic complex systems: a Boolean circuit and an amorphous material undergoing plastic deformation. In both examples, the large amount of entropy of the system state is decomposed, bit by bit, in terms of what is most related to macroscale behavior. The identification of meaningful variation in data, with the full generality brought by information theory, is made practical for studying the connection between micro- and macroscale structure in complex systems.
Collapse
Affiliation(s)
- Kieran A. Murphy
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA19104
| | - Dani S. Bassett
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA19104
- Department of Electrical & Systems Engineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA19104
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Physics & Astronomy, College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA19104
- The Santa Fe Institute, Santa Fe, NM87501
| |
Collapse
|
7
|
Kim D, Tran A, Kim HJ, Lin Y, Yang JYH, Yang P. Gene regulatory network reconstruction: harnessing the power of single-cell multi-omic data. NPJ Syst Biol Appl 2023; 9:51. [PMID: 37857632 PMCID: PMC10587078 DOI: 10.1038/s41540-023-00312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023] Open
Abstract
Inferring gene regulatory networks (GRNs) is a fundamental challenge in biology that aims to unravel the complex relationships between genes and their regulators. Deciphering these networks plays a critical role in understanding the underlying regulatory crosstalk that drives many cellular processes and diseases. Recent advances in sequencing technology have led to the development of state-of-the-art GRN inference methods that exploit matched single-cell multi-omic data. By employing diverse mathematical and statistical methodologies, these methods aim to reconstruct more comprehensive and precise gene regulatory networks. In this review, we give a brief overview on the statistical and methodological foundations commonly used in GRN inference methods. We then compare and contrast the latest state-of-the-art GRN inference methods for single-cell matched multi-omics data, and discuss their assumptions, limitations and opportunities. Finally, we discuss the challenges and future directions that hold promise for further advancements in this rapidly developing field.
Collapse
Affiliation(s)
- Daniel Kim
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia
- Computational Systems Biology Unit, Children's Medical Research Institute, University of Sydney, Camperdown, NSW, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, Australia
| | - Andy Tran
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Hani Jieun Kim
- Computational Systems Biology Unit, Children's Medical Research Institute, University of Sydney, Camperdown, NSW, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, Australia
| | - Yingxin Lin
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Jean Yee Hwa Yang
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia.
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, Australia.
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia.
| | - Pengyi Yang
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia.
- Computational Systems Biology Unit, Children's Medical Research Institute, University of Sydney, Camperdown, NSW, Australia.
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, Australia.
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
8
|
Saint-Antoine M, Singh A. Benchmarking Gene Regulatory Network Inference Methods on Simulated and Experimental Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.12.540581. [PMID: 37215029 PMCID: PMC10197678 DOI: 10.1101/2023.05.12.540581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Although the challenge of gene regulatory network inference has been studied for more than a decade, it is still unclear how well network inference methods work when applied to real data. Attempts to benchmark these methods on experimental data have yielded mixed results, in which sometimes even the best methods fail to outperform random guessing, and in other cases they perform reasonably well. So, one of the most valuable contributions one can currently make to the field of network inference is to benchmark methods on experimental data for which the true underlying network is already known, and report the results so that we can get a clearer picture of their efficacy. In this paper, we report results from the first, to our knowledge, benchmarking of network inference methods on single cell E. coli transcriptomic data. We report a moderate level of accuracy for the methods, better than random chance but still far from perfect. We also find that some methods that were quite strong and accurate on microarray and bulk RNA-seq data did not perform as well on the single cell data. Additionally, we benchmark a simple network inference method (Pearson correlation), on data generated through computer simulations in order to draw conclusions about general best practices in network inference studies. We predict that network inference would be more accurate using proteomic data rather than transcriptomic data, which could become relevant if high-throughput proteomic experimental methods are developed in the future. We also show through simulations that using a simplified model of gene expression that skips the mRNA step tends to substantially overestimate the accuracy of network inference methods, and advise against using this model for future in silico benchmarking studies.
Collapse
Affiliation(s)
- Michael Saint-Antoine
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE USA 19716
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, Biomedical Engineering, Mathematical Sciences, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE USA 19716
| |
Collapse
|
9
|
Arend M, Yuan Y, Ruiz-Sola MÁ, Omranian N, Nikoloski Z, Petroutsos D. Widening the landscape of transcriptional regulation of green algal photoprotection. Nat Commun 2023; 14:2687. [PMID: 37164999 PMCID: PMC10172295 DOI: 10.1038/s41467-023-38183-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/17/2023] [Indexed: 05/12/2023] Open
Abstract
Availability of light and CO2, substrates of microalgae photosynthesis, is frequently far from optimal. Microalgae activate photoprotection under strong light, to prevent oxidative damage, and the CO2 Concentrating Mechanism (CCM) under low CO2, to raise intracellular CO2 levels. The two processes are interconnected; yet, the underlying transcriptional regulators remain largely unknown. Employing a large transcriptomic data compendium of Chlamydomonas reinhardtii's responses to different light and carbon supply, we reconstruct a consensus genome-scale gene regulatory network from complementary inference approaches and use it to elucidate transcriptional regulators of photoprotection. We show that the CCM regulator LCR1 also controls photoprotection, and that QER7, a Squamosa Binding Protein, suppresses photoprotection- and CCM-gene expression under the control of the blue light photoreceptor Phototropin. By demonstrating the existence of regulatory hubs that channel light- and CO2-mediated signals into a common response, our study provides an accessible resource to dissect gene expression regulation in this microalga.
Collapse
Affiliation(s)
- Marius Arend
- Bioinformatics Group, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany
- Systems Biology and Mathematical Modeling Group, Max-Planck-Institute of Molecular Plant Physiology, 14476, Potsdam, Germany
- Bioinformatics and Mathematical Modeling Department, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria
| | - Yizhong Yuan
- University of Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, 38000, Grenoble, France
| | - M Águila Ruiz-Sola
- University of Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, 38000, Grenoble, France
- Instituto de Bioquímica Vegetal y Fotosíntesis, Universidad de Sevilla-CSIC, 41092, Sevilla, Spain
| | - Nooshin Omranian
- Bioinformatics Group, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany
- Systems Biology and Mathematical Modeling Group, Max-Planck-Institute of Molecular Plant Physiology, 14476, Potsdam, Germany
- Bioinformatics and Mathematical Modeling Department, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria
| | - Zoran Nikoloski
- Bioinformatics Group, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany.
- Systems Biology and Mathematical Modeling Group, Max-Planck-Institute of Molecular Plant Physiology, 14476, Potsdam, Germany.
- Bioinformatics and Mathematical Modeling Department, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria.
| | - Dimitris Petroutsos
- University of Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, 38000, Grenoble, France.
| |
Collapse
|
10
|
Bornhofen E, Fè D, Nagy I, Lenk I, Greve M, Didion T, Jensen CS, Asp T, Janss L. Genetic architecture of inter-specific and -generic grass hybrids by network analysis on multi-omics data. BMC Genomics 2023; 24:213. [PMID: 37095447 PMCID: PMC10127077 DOI: 10.1186/s12864-023-09292-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/02/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Understanding the mechanisms underlining forage production and its biomass nutritive quality at the omics level is crucial for boosting the output of high-quality dry matter per unit of land. Despite the advent of multiple omics integration for the study of biological systems in major crops, investigations on forage species are still scarce. RESULTS Our results identified substantial changes in gene co-expression and metabolite-metabolite network topologies as a result of genetic perturbation by hybridizing L. perenne with another species within the genus (L. multiflorum) relative to across genera (F. pratensis). However, conserved hub genes and hub metabolomic features were detected between pedigree classes, some of which were highly heritable and displayed one or more significant edges with agronomic traits in a weighted omics-phenotype network. In spite of tagging relevant biological molecules as, for example, the light-induced rice 1 (LIR1), hub features were not necessarily better explanatory variables for omics-assisted prediction than features stochastically sampled and all available regressors. CONCLUSIONS The utilization of computational techniques for the reconstruction of co-expression networks facilitates the identification of key omic features that serve as central nodes and demonstrate correlation with the manifestation of observed traits. Our results also indicate a robust association between early multi-omic traits measured in a greenhouse setting and phenotypic traits evaluated under field conditions.
Collapse
Affiliation(s)
- Elesandro Bornhofen
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark.
| | - Dario Fè
- Research Division, DLF Seeds A/S, Store Heddinge, Denmark
| | - Istvan Nagy
- Center for Quantitative Genetics and Genomics, Aarhus University, Slagelse, Denmark
| | - Ingo Lenk
- Research Division, DLF Seeds A/S, Store Heddinge, Denmark
| | - Morten Greve
- Research Division, DLF Seeds A/S, Store Heddinge, Denmark
| | - Thomas Didion
- Research Division, DLF Seeds A/S, Store Heddinge, Denmark
| | | | - Torben Asp
- Center for Quantitative Genetics and Genomics, Aarhus University, Slagelse, Denmark
| | - Luc Janss
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
11
|
Wang Q, Guo M, Chen J, Duan R. A gene regulatory network inference model based on pseudo-siamese network. BMC Bioinformatics 2023; 24:163. [PMID: 37085776 PMCID: PMC10122305 DOI: 10.1186/s12859-023-05253-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/24/2023] [Indexed: 04/23/2023] Open
Abstract
MOTIVATION Gene regulatory networks (GRNs) arise from the intricate interactions between transcription factors (TFs) and their target genes during the growth and development of organisms. The inference of GRNs can unveil the underlying gene interactions in living systems and facilitate the investigation of the relationship between gene expression patterns and phenotypic traits. Although several machine-learning models have been proposed for inferring GRNs from single-cell RNA sequencing (scRNA-seq) data, some of these models, such as Boolean and tree-based networks, suffer from sensitivity to noise and may encounter difficulties in handling the high noise and dimensionality of actual scRNA-seq data, as well as the sparse nature of gene regulation relationships. Thus, inferring large-scale information from GRNs remains a formidable challenge. RESULTS This study proposes a multilevel, multi-structure framework called a pseudo-Siamese GRN (PSGRN) for inferring large-scale GRNs from time-series expression datasets. Based on the pseudo-Siamese network, we applied a gated recurrent unit to capture the time features of each TF and target matrix and learn the spatial features of the matrices after merging by applying the DenseNet framework. Finally, we applied a sigmoid function to evaluate interactions. We constructed two maize sub-datasets, including gene expression levels and GRNs, using existing open-source maize multi-omics data and compared them to other GRN inference methods, including GENIE3, GRNBoost2, nonlinear ordinary differential equations, CNNC, and DGRNS. Our results show that PSGRN outperforms state-of-the-art methods. This study proposed a new framework: a PSGRN that allows GRNs to be inferred from scRNA-seq data, elucidating the temporal and spatial features of TFs and their target genes. The results show the model's robustness and generalization, laying a theoretical foundation for maize genotype-phenotype associations with implications for breeding work.
Collapse
Affiliation(s)
- Qian Wang
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing, China
| | - Maozu Guo
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing, China.
| | - Jian Chen
- College of Agronomy and Biotechnology, China Agricultural University, Beijing, China
| | - Ran Duan
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing, China
| |
Collapse
|
12
|
Cinaglia P, Cannataro M. A Method Based on Temporal Embedding for the Pairwise Alignment of Dynamic Networks. ENTROPY (BASEL, SWITZERLAND) 2023; 25:e25040665. [PMID: 37190452 PMCID: PMC10138164 DOI: 10.3390/e25040665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023]
Abstract
In network analysis, real-world systems may be represented via graph models, where nodes and edges represent the set of biological objects (e.g., genes, proteins, molecules) and their interactions, respectively. This representative knowledge-graph model may also consider the dynamics involved in the evolution of the network (i.e., dynamic networks), in addition to a classic static representation (i.e., static networks). Bioinformatics solutions for network analysis allow knowledge extraction from the features related to a single network of interest or by comparing networks of different species. For instance, we may align a network related to a well known species to a more complex one in order to find a match able to support new hypotheses or studies. Therefore, the network alignment is crucial for transferring the knowledge between species, usually from simplest (e.g., rat) to more complex (e.g., human). Methods: In this paper, we present Dynamic Network Alignment based on Temporal Embedding (DANTE), a novel method for pairwise alignment of dynamic networks that applies the temporal embedding to investigate the topological similarities between the two input dynamic networks. The main idea of DANTE is to consider the evolution of interactions and the changes in network topology. Briefly, the proposed solution builds a similarity matrix by integrating the tensors computed via the embedding process and, subsequently, it aligns the pairs of nodes by performing its own iterative maximization function. Results: The performed experiments have reported promising results in terms of precision and accuracy, as well as good robustness as the number of nodes and time points increases. The proposed solution showed an optimal trade-off between sensitivity and specificity on the alignments produced on several noisy versions of the dynamic yeast network, by improving by ∼18.8% (with a maximum of 20.6%) the Area Under the Receiver Operating Characteristic (ROC) Curve (i.e., AUC or AUROC), compared to two well known methods: DYNAMAGNA++ and DYNAWAVE. From the point of view of quality, DANTE outperformed these by ∼91% as nodes increase and by ∼75% as the number of time points increases. Furthermore, a ∼23.73% improvement in terms of node correctness was reported with our solution on real dynamic networks.
Collapse
Affiliation(s)
- Pietro Cinaglia
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Mario Cannataro
- Department of Medical and Surgical Sciences, Data Analytics Research Center, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
13
|
Borg JP, Colinge J, Ravel P. Modular response analysis reformulated as a multilinear regression problem. Bioinformatics 2023; 39:btad166. [PMID: 37021935 PMCID: PMC10097436 DOI: 10.1093/bioinformatics/btad166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/29/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
MOTIVATION Modular response analysis (MRA) is a well-established method to infer biological networks from perturbation data. Classically, MRA requires the solution of a linear system, and results are sensitive to noise in the data and perturbation intensities. Due to noise propagation, applications to networks of 10 nodes or more are difficult. RESULTS We propose a new formulation of MRA as a multilinear regression problem. This enables to integrate all the replicates and potential additional perturbations in a larger, over-determined, and more stable system of equations. More relevant confidence intervals on network parameters can be obtained, and we show competitive performance for networks of size up to 1000. Prior knowledge integration in the form of known null edges further improves these results. AVAILABILITY AND IMPLEMENTATION The R code used to obtain the presented results is available from GitHub: https://github.com/J-P-Borg/BioInformatics.
Collapse
Affiliation(s)
- Jean-Pierre Borg
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Montpellier 34298, France
- Institut régional du Cancer Montpellier, Montpellier 34298, France
- Université de Montpellier, Montpellier 34090, France
| | - Jacques Colinge
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Montpellier 34298, France
- Institut régional du Cancer Montpellier, Montpellier 34298, France
- Université de Montpellier, Montpellier 34090, France
- Faculté de Médecine, Montpellier 34090, France
| | - Patrice Ravel
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Montpellier 34298, France
- Institut régional du Cancer Montpellier, Montpellier 34298, France
- Université de Montpellier, Montpellier 34090, France
- Faculté de Pharmacie, Montpellier 34090, France
| |
Collapse
|
14
|
Ventre E, Herbach U, Espinasse T, Benoit G, Gandrillon O. One model fits all: Combining inference and simulation of gene regulatory networks. PLoS Comput Biol 2023; 19:e1010962. [PMID: 36972296 PMCID: PMC10079230 DOI: 10.1371/journal.pcbi.1010962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 04/06/2023] [Accepted: 02/17/2023] [Indexed: 03/29/2023] Open
Abstract
The rise of single-cell data highlights the need for a nondeterministic view of gene expression, while offering new opportunities regarding gene regulatory network inference. We recently introduced two strategies that specifically exploit time-course data, where single-cell profiling is performed after a stimulus: HARISSA, a mechanistic network model with a highly efficient simulation procedure, and CARDAMOM, a scalable inference method seen as model calibration. Here, we combine the two approaches and show that the same model driven by transcriptional bursting can be used simultaneously as an inference tool, to reconstruct biologically relevant networks, and as a simulation tool, to generate realistic transcriptional profiles emerging from gene interactions. We verify that CARDAMOM quantitatively reconstructs causal links when the data is simulated from HARISSA, and demonstrate its performance on experimental data collected on in vitro differentiating mouse embryonic stem cells. Overall, this integrated strategy largely overcomes the limitations of disconnected inference and simulation.
Collapse
Affiliation(s)
- Elias Ventre
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Lyon, France
- Inria Center Grenoble Rhône-Alpes, Équipe Dracula, Villeurbanne, France
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5208, Institut Camille Jordan, Villeurbanne, France
| | - Ulysse Herbach
- Université de Lorraine, CNRS, Inria, IECL, Nancy, France
| | - Thibault Espinasse
- Inria Center Grenoble Rhône-Alpes, Équipe Dracula, Villeurbanne, France
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5208, Institut Camille Jordan, Villeurbanne, France
| | - Gérard Benoit
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Lyon, France
| | - Olivier Gandrillon
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Lyon, France
- Inria Center Grenoble Rhône-Alpes, Équipe Dracula, Villeurbanne, France
| |
Collapse
|
15
|
Upadhyaya P, Zhang K, Li C, Jiang X, Kim Y. Scalable Causal Structure Learning: Scoping Review of Traditional and Deep Learning Algorithms and New Opportunities in Biomedicine. JMIR Med Inform 2023; 11:e38266. [PMID: 36649070 PMCID: PMC9890349 DOI: 10.2196/38266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/30/2022] [Accepted: 09/18/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Causal structure learning refers to a process of identifying causal structures from observational data, and it can have multiple applications in biomedicine and health care. OBJECTIVE This paper provides a practical review and tutorial on scalable causal structure learning models with examples of real-world data to help health care audiences understand and apply them. METHODS We reviewed traditional (combinatorial and score-based) methods for causal structure discovery and machine learning-based schemes. Various traditional approaches have been studied to tackle this problem, the most important among these being the Peter Spirtes and Clark Glymour algorithms. This was followed by analyzing the literature on score-based methods, which are computationally faster. Owing to the continuous constraint on acyclicity, there are new deep learning approaches to the problem in addition to traditional and score-based methods. Such methods can also offer scalability, particularly when there is a large amount of data involving multiple variables. Using our own evaluation metrics and experiments on linear, nonlinear, and benchmark Sachs data, we aimed to highlight the various advantages and disadvantages associated with these methods for the health care community. We also highlighted recent developments in biomedicine where causal structure learning can be applied to discover structures such as gene networks, brain connectivity networks, and those in cancer epidemiology. RESULTS We also compared the performance of traditional and machine learning-based algorithms for causal discovery over some benchmark data sets. Directed Acyclic Graph-Graph Neural Network has the lowest structural hamming distance (19) and false positive rate (0.13) based on the Sachs data set, whereas Greedy Equivalence Search and Max-Min Hill Climbing have the best false discovery rate (0.68) and true positive rate (0.56), respectively. CONCLUSIONS Machine learning-based approaches, including deep learning, have many advantages over traditional approaches, such as scalability, including a greater number of variables, and potentially being applied in a wide range of biomedical applications, such as genetics, if sufficient data are available. Furthermore, these models are more flexible than traditional models and are poised to positively affect many applications in the future.
Collapse
Affiliation(s)
- Pulakesh Upadhyaya
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, HOUSTON, TX, United States
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, United States
| | - Kai Zhang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, HOUSTON, TX, United States
| | - Can Li
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, HOUSTON, TX, United States
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, HOUSTON, TX, United States
| | - Yejin Kim
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, HOUSTON, TX, United States
| |
Collapse
|
16
|
Chen G, Liu ZP. Inferring causal gene regulatory network via GreyNet: From dynamic grey association to causation. Front Bioeng Biotechnol 2022; 10:954610. [PMID: 36237217 PMCID: PMC9551017 DOI: 10.3389/fbioe.2022.954610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
Gene regulatory network (GRN) provides abundant information on gene interactions, which contributes to demonstrating pathology, predicting clinical outcomes, and identifying drug targets. Existing high-throughput experiments provide rich time-series gene expression data to reconstruct the GRN to further gain insights into the mechanism of organisms responding to external stimuli. Numerous machine-learning methods have been proposed to infer gene regulatory networks. Nevertheless, machine learning, especially deep learning, is generally a “black box,” which lacks interpretability. The causality has not been well recognized in GRN inference procedures. In this article, we introduce grey theory integrated with the adaptive sliding window technique to flexibly capture instant gene–gene interactions in the uncertain regulatory system. Then, we incorporate generalized multivariate Granger causality regression methods to transform the dynamic grey association into causation to generate directional regulatory links. We evaluate our model on the DREAM4 in silico benchmark dataset and real-world hepatocellular carcinoma (HCC) time-series data. We achieved competitive results on the DREAM4 compared with other state-of-the-art algorithms and gained meaningful GRN structure on HCC data respectively.
Collapse
Affiliation(s)
- Guangyi Chen
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong, China
| | - Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong, China
- Center for Intelligent Medicine, Shandong University, Jinan, Shandong, China
- *Correspondence: Zhi-Ping Liu,
| |
Collapse
|
17
|
Amini Farsani Z, Schmid VJ. Maximum Entropy Technique and Regularization Functional for Determining the Pharmacokinetic Parameters in DCE-MRI. J Digit Imaging 2022; 35:1176-1188. [PMID: 35618849 PMCID: PMC9582183 DOI: 10.1007/s10278-022-00646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/15/2022] [Accepted: 04/21/2022] [Indexed: 10/31/2022] Open
Abstract
This paper aims to solve the arterial input function (AIF) determination in dynamic contrast-enhanced MRI (DCE-MRI), an important linear ill-posed inverse problem, using the maximum entropy technique (MET) and regularization functionals. In addition, estimating the pharmacokinetic parameters from a DCE-MR image investigations is an urgent need to obtain the precise information about the AIF-the concentration of the contrast agent on the left ventricular blood pool measured over time. For this reason, the main idea is to show how to find a unique solution of linear system of equations generally in the form of [Formula: see text] named an ill-conditioned linear system of equations after discretization of the integral equations, which appear in different tomographic image restoration and reconstruction issues. Here, a new algorithm is described to estimate an appropriate probability distribution function for AIF according to the MET and regularization functionals for the contrast agent concentration when applying Bayesian estimation approach to estimate two different pharmacokinetic parameters. Moreover, by using the proposed approach when analyzing simulated and real datasets of the breast tumors according to pharmacokinetic factors, it indicates that using Bayesian inference-that infer the uncertainties of the computed solutions, and specific knowledge of the noise and errors-combined with the regularization functional of the maximum entropy problem, improved the convergence behavior and led to more consistent morphological and functional statistics and results. Finally, in comparison to the proposed exponential distribution based on MET and Newton's method, or Weibull distribution via the MET and teaching-learning-based optimization (MET/TLBO) in the previous studies, the family of Gamma and Erlang distributions estimated by the new algorithm are more appropriate and robust AIFs.
Collapse
Affiliation(s)
- Zahra Amini Farsani
- Bayesian Imaging and Spatial Statistics Group, Institute of Statistics, Ludwig-Maximilian-Universität München, Ludwigstraße 33, 80539, Munich, Germany. .,Statistics Department, School of Science, Lorestan University, 68151-44316, Khorramabad, Iran.
| | - Volker J Schmid
- Bayesian Imaging and Spatial Statistics Group, Institute of Statistics, Ludwig-Maximilian-Universität München, Ludwigstraße 33, 80539, Munich, Germany
| |
Collapse
|
18
|
Liu W, Sun X, Yang L, Li K, Yang Y, Fu X. NSCGRN: a network structure control method for gene regulatory network inference. Brief Bioinform 2022; 23:6585392. [PMID: 35554485 DOI: 10.1093/bib/bbac156] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 01/18/2023] Open
Abstract
Accurate inference of gene regulatory networks (GRNs) is an essential premise for understanding pathogenesis and curing diseases. Various computational methods have been developed for GRN inference, but the identification of redundant regulation remains a challenge faced by researchers. Although combining global and local topology can identify and reduce redundant regulations, the topologies' specific forms and cooperation modes are unclear and real regulations may be sacrificed. Here, we propose a network structure control method [network-structure-controlling-based GRN inference method (NSCGRN)] that stipulates the global and local topology's specific forms and cooperation mode. The method is carried out in a cooperative mode of 'global topology dominates and local topology refines'. Global topology requires layering and sparseness of the network, and local topology requires consistency of the subgraph association pattern with the network motifs (fan-in, fan-out, cascade and feedforward loop). Specifically, an ordered gene list is obtained by network topology centrality sorting. A Bernaola-Galvan mutation detection algorithm applied to the list gives the hierarchy of GRNs to control the upstream and downstream regulations within the global scope. Finally, four network motifs are integrated into the hierarchy to optimize local complex regulations and form a cooperative mode where global and local topologies play the dominant and refined roles, respectively. NSCGRN is compared with state-of-the-art methods on three different datasets (six networks in total), and it achieves the highest F1 and Matthews correlation coefficient. Experimental results show its unique advantages in GRN inference.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China.,School of Computer Science, Xiangtan University, Xiangtan, 411105, China
| | - Xingen Sun
- School of Computer Science, Xiangtan University, Xiangtan, 411105, China.,Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Li Yang
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Kaiwen Li
- Artificial Intelligence Research Institute, China University of Mining and Technology, Xuzhou, 221116, China
| | - Yu Yang
- School of Computer Science, Xiangtan University, Xiangtan, 411105, China.,Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Xiangzheng Fu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410000, China
| |
Collapse
|
19
|
Inference of Molecular Regulatory Systems Using Statistical Path-Consistency Algorithm. ENTROPY 2022; 24:e24050693. [PMID: 35626576 PMCID: PMC9142129 DOI: 10.3390/e24050693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/16/2022]
Abstract
One of the key challenges in systems biology and molecular sciences is how to infer regulatory relationships between genes and proteins using high-throughout omics datasets. Although a wide range of methods have been designed to reverse engineer the regulatory networks, recent studies show that the inferred network may depend on the variable order in the dataset. In this work, we develop a new algorithm, called the statistical path-consistency algorithm (SPCA), to solve the problem of the dependence of variable order. This method generates a number of different variable orders using random samples, and then infers a network by using the path-consistent algorithm based on each variable order. We propose measures to determine the edge weights using the corresponding edge weights in the inferred networks, and choose the edges with the largest weights as the putative regulations between genes or proteins. The developed method is rigorously assessed by the six benchmark networks in DREAM challenges, the mitogen-activated protein (MAP) kinase pathway, and a cancer-specific gene regulatory network. The inferred networks are compared with those obtained by using two up-to-date inference methods. The accuracy of the inferred networks shows that the developed method is effective for discovering molecular regulatory systems.
Collapse
|
20
|
Inference on the structure of gene regulatory networks. J Theor Biol 2022; 539:111055. [PMID: 35150721 DOI: 10.1016/j.jtbi.2022.111055] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 11/20/2022]
Abstract
In this paper, we conduct theoretical analyses on inferring the structure of gene regulatory networks. Depending on the experimental method and data type, the inference problem is classified into 20 different scenarios. For each scenario, we discuss the problem that with enough data, under what assumptions, what can be inferred about the structure. For scenarios that have been covered in the literature, we provide a brief review. For scenarios that have not been covered in literature, if the structure can be inferred, we propose new mathematical inference methods and evaluate them on simulated data. Otherwise, we prove that the structure cannot be inferred.
Collapse
|
21
|
Tao W, Radstake TRDJ, Pandit A. RegEnrich gene regulator enrichment analysis reveals a key role of the ETS transcription factor family in interferon signaling. Commun Biol 2022; 5:31. [PMID: 35017649 PMCID: PMC8752721 DOI: 10.1038/s42003-021-02991-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Changes in a few key transcriptional regulators can lead to different biological states. Extracting the key gene regulators governing a biological state allows us to gain mechanistic insights. Most current tools perform pathway/GO enrichment analysis to identify key genes and regulators but tend to overlook the gene/protein regulatory interactions. Here we present RegEnrich, an open-source Bioconductor R package, which combines differential expression analysis, data-driven gene regulatory network inference, enrichment analysis, and gene regulator ranking to identify key regulators using gene/protein expression profiling data. By benchmarking using multiple gene expression datasets of gene silencing studies, we found that RegEnrich using the GSEA method to rank the regulators performed the best. Further, RegEnrich was applied to 21 publicly available datasets on in vitro interferon-stimulation of different cell types. Collectively, RegEnrich can accurately identify key gene regulators from the cells under different biological states, which can be valuable in mechanistically studying cell differentiation, cell response to drug stimulation, disease development, and ultimately drug development.
Collapse
Affiliation(s)
- Weiyang Tao
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Timothy R D J Radstake
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Aridaman Pandit
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
22
|
Emerging Machine Learning Techniques for Modelling Cellular Complex Systems in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1338:199-208. [PMID: 34973026 DOI: 10.1007/978-3-030-78775-2_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
We live in the big data era in the biomedical field, where machine learning has a very important contribution to the interpretation of complex biological processes and diseases, since it has the potential to create predictive models from multidimensional data sets. Part of the application of machine learning in biomedical science is to study and model complex cellular systems such as biological networks. In this context, the study of complex diseases, such as Alzheimer's diseases (AD), benefits from established methodologies of network science and machine learning as they offer algorithmic tools and techniques that can address the limitations and challenges of modeling and studying cellular AD-related networks. In this paper we analyze the opportunities and challenges at the intersection of machine learning and network biology and whether this can affect the biological interpretation and clarification of diseases. Specifically, we focus on GRN techniques which through omics data and the use of machine learning techniques can construct a network that captures all the information at the molecular level for the disease under study. We record the emerging machine learning techniques that are focus on ensemble tree-based techniques in the area of classification and regression. Their potential for unraveling the complexity of model cellular systems in complex diseases, such as AD, offers the opportunity for novel machine learning methodologies to decipher the mechanisms of the various AD processes.
Collapse
|
23
|
Gong H, Zhu S, Zhu X, Fang Q, Zhang XY, Wu R. A Multilayer Interactome Network Constructed in a Forest Poplar Population Mediates the Pleiotropic Control of Complex Traits. Front Genet 2021; 12:769688. [PMID: 34868256 PMCID: PMC8633413 DOI: 10.3389/fgene.2021.769688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
The effects of genes on physiological and biochemical processes are interrelated and interdependent; it is common for genes to express pleiotropic control of complex traits. However, the study of gene expression and participating pathways in vivo at the whole-genome level is challenging. Here, we develop a coupled regulatory interaction differential equation to assess overall and independent genetic effects on trait growth. Based on evolutionary game theory and developmental modularity theory, we constructed multilayer, omnigenic networks of bidirectional, weighted, and positive or negative epistatic interactions using a forest poplar tree mapping population, which were organized into metagalactic, intergalactic, and local interstellar networks that describe layers of structure between modules, submodules, and individual single nucleotide polymorphisms, respectively. These multilayer interactomes enable the exploration of complex interactions between genes, and the analysis of not only differential expression of quantitative trait loci but also previously uncharacterized determinant SNPs, which are negatively regulated by other SNPs, based on the deconstruction of genetic effects to their component parts. Our research framework provides a tool to comprehend the pleiotropic control of complex traits and explores the inherent directional connections between genes in the structure of omnigenic networks.
Collapse
Affiliation(s)
- Huiying Gong
- College of Science, Beijing Forestry University, Beijing, China
| | - Sheng Zhu
- College of Biology and the Environment, Nanjing Forestry University, Nanjing, China
| | - Xuli Zhu
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Qing Fang
- Faculty of Science, Yamagata University, Yamagata, Japan
| | - Xiao-Yu Zhang
- College of Science, Beijing Forestry University, Beijing, China
| | - Rongling Wu
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
- Center for Statistical Genetics, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
24
|
Bernal V, Bischoff R, Horvatovich P, Guryev V, Grzegorczyk M. The 'un-shrunk' partial correlation in Gaussian graphical models. BMC Bioinformatics 2021; 22:424. [PMID: 34493207 PMCID: PMC8424921 DOI: 10.1186/s12859-021-04313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 08/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In systems biology, it is important to reconstruct regulatory networks from quantitative molecular profiles. Gaussian graphical models (GGMs) are one of the most popular methods to this end. A GGM consists of nodes (representing the transcripts, metabolites or proteins) inter-connected by edges (reflecting their partial correlations). Learning the edges from quantitative molecular profiles is statistically challenging, as there are usually fewer samples than nodes ('high dimensional problem'). Shrinkage methods address this issue by learning a regularized GGM. However, it remains open to study how the shrinkage affects the final result and its interpretation. RESULTS We show that the shrinkage biases the partial correlation in a non-linear way. This bias does not only change the magnitudes of the partial correlations but also affects their order. Furthermore, it makes networks obtained from different experiments incomparable and hinders their biological interpretation. We propose a method, referred to as 'un-shrinking' the partial correlation, which corrects for this non-linear bias. Unlike traditional methods, which use a fixed shrinkage value, the new approach provides partial correlations that are closer to the actual (population) values and that are easier to interpret. This is demonstrated on two gene expression datasets from Escherichia coli and Mus musculus. CONCLUSIONS GGMs are popular undirected graphical models based on partial correlations. The application of GGMs to reconstruct regulatory networks is commonly performed using shrinkage to overcome the 'high-dimensional problem'. Besides it advantages, we have identified that the shrinkage introduces a non-linear bias in the partial correlations. Ignoring this type of effects caused by the shrinkage can obscure the interpretation of the network, and impede the validation of earlier reported results.
Collapse
Affiliation(s)
- Victor Bernal
- Bernoulli Institute, University of Groningen, Groningen, 9747 AG, The Netherlands.,Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands.
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands.
| | - Marco Grzegorczyk
- Bernoulli Institute, University of Groningen, Groningen, 9747 AG, The Netherlands.
| |
Collapse
|
25
|
Masoomy H, Askari B, Tajik S, Rizi AK, Jafari GR. Topological analysis of interaction patterns in cancer-specific gene regulatory network: persistent homology approach. Sci Rep 2021; 11:16414. [PMID: 34385492 PMCID: PMC8361050 DOI: 10.1038/s41598-021-94847-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/16/2021] [Indexed: 01/01/2023] Open
Abstract
In this study, we investigated cancer cellular networks in the context of gene interactions and their associated patterns in order to recognize the structural features underlying this disease. We aim to propose that the quest of understanding cancer takes us beyond pairwise interactions between genes to a higher-order construction. We characterize the most prominent network deviations in the gene interaction patterns between cancer and normal samples that contribute to the complexity of this disease. What we hope is that through understanding these interaction patterns we will notice a deeper structure in the cancer network. This study uncovers the significant deviations that topological features in cancerous cells show from the healthy one, where the last stage of filtration confirms the importance of one-dimensional holes (topological loops) in cancerous cells and two-dimensional holes (topological voids) in healthy cells. In the small threshold region, the drop in the number of connected components of the cancer network, along with the rise in the number of loops and voids, all occurring at some smaller weight values compared to the normal case, reveals the cancerous network tendency to certain pathways.
Collapse
Affiliation(s)
- Hosein Masoomy
- Physics Department, Shahid Beheshti University, Tehran, Iran
| | - Behrouz Askari
- Physics Department, Shahid Beheshti University, Tehran, Iran
| | - Samin Tajik
- Physics Department, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Abbas K Rizi
- Department of Computer Science, School of Science, Aalto University, 0007, Espoo, Finland
| | - G Reza Jafari
- Physics Department, Shahid Beheshti University, Tehran, Iran.
- Department of Network and Data Science, Central European University, Budapest, 1051, Hungary.
| |
Collapse
|
26
|
Vatsa D, Agarwal S. PEPN-GRN: A Petri net-based approach for the inference of gene regulatory networks from noisy gene expression data. PLoS One 2021; 16:e0251666. [PMID: 33989333 PMCID: PMC8121333 DOI: 10.1371/journal.pone.0251666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/30/2021] [Indexed: 11/22/2022] Open
Abstract
The inference of gene regulatory networks (GRNs) from expression data is a challenging problem in systems biology. The stochasticity or fluctuations in the biochemical processes that regulate the transcription process poses as one of the major challenges. In this paper, we propose a novel GRN inference approach, named the Probabilistic Extended Petri Net for Gene Regulatory Network (PEPN-GRN), for the inference of gene regulatory networks from noisy expression data. The proposed inference approach makes use of transition of discrete gene expression levels across adjacent time points as different evidence types that relate to the production or decay of genes. The paper examines three variants of the PEPN-GRN method, which mainly differ by the way the scores of network edges are computed using evidence types. The proposed method is evaluated on the benchmark DREAM4 in silico data sets and a real time series data set of E. coli from the DREAM5 challenge. The PEPN-GRN_v3 variant (the third variant of the PEPN-GRN approach) sought to learn the weights of evidence types in accordance with their contribution to the activation and inhibition gene regulation process. The learned weights help understand the time-shifted and inverted time-shifted relationship between regulator and target gene. Thus, PEPN-GRN_v3, along with the inference of network edges, also provides a functional understanding of the gene regulation process.
Collapse
Affiliation(s)
- Deepika Vatsa
- Department of Electrical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Sumeet Agarwal
- Department of Electrical Engineering, Indian Institute of Technology Delhi, New Delhi, India
- * E-mail: ,
| |
Collapse
|
27
|
Zhao M, He W, Tang J, Zou Q, Guo F. A comprehensive overview and critical evaluation of gene regulatory network inference technologies. Brief Bioinform 2021; 22:6128842. [PMID: 33539514 DOI: 10.1093/bib/bbab009] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/11/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Gene regulatory network (GRN) is the important mechanism of maintaining life process, controlling biochemical reaction and regulating compound level, which plays an important role in various organisms and systems. Reconstructing GRN can help us to understand the molecular mechanism of organisms and to reveal the essential rules of a large number of biological processes and reactions in organisms. Various outstanding network reconstruction algorithms use specific assumptions that affect prediction accuracy, in order to deal with the uncertainty of processing. In order to study why a certain method is more suitable for specific research problem or experimental data, we conduct research from model-based, information-based and machine learning-based method classifications. There are obviously different types of computational tools that can be generated to distinguish GRNs. Furthermore, we discuss several classical, representative and latest methods in each category to analyze core ideas, general steps, characteristics, etc. We compare the performance of state-of-the-art GRN reconstruction technologies on simulated networks and real networks under different scaling conditions. Through standardized performance metrics and common benchmarks, we quantitatively evaluate the stability of various methods and the sensitivity of the same algorithm applying to different scaling networks. The aim of this study is to explore the most appropriate method for a specific GRN, which helps biologists and medical scientists in discovering potential drug targets and identifying cancer biomarkers.
Collapse
Affiliation(s)
- Mengyuan Zhao
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Wenying He
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Jijun Tang
- University of South Carolina, Tianjin, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Fei Guo
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| |
Collapse
|
28
|
Zanin M, Aitya NA, Basilio J, Baumbach J, Benis A, Behera CK, Bucholc M, Castiglione F, Chouvarda I, Comte B, Dao TT, Ding X, Pujos-Guillot E, Filipovic N, Finn DP, Glass DH, Harel N, Iesmantas T, Ivanoska I, Joshi A, Boudjeltia KZ, Kaoui B, Kaur D, Maguire LP, McClean PL, McCombe N, de Miranda JL, Moisescu MA, Pappalardo F, Polster A, Prasad G, Rozman D, Sacala I, Sanchez-Bornot JM, Schmid JA, Sharp T, Solé-Casals J, Spiwok V, Spyrou GM, Stalidzans E, Stres B, Sustersic T, Symeonidis I, Tieri P, Todd S, Van Steen K, Veneva M, Wang DH, Wang H, Wang H, Watterson S, Wong-Lin K, Yang S, Zou X, Schmidt HH. An Early Stage Researcher's Primer on Systems Medicine Terminology. NETWORK AND SYSTEMS MEDICINE 2021; 4:2-50. [PMID: 33659919 PMCID: PMC7919422 DOI: 10.1089/nsm.2020.0003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Systems Medicine is a novel approach to medicine, that is, an interdisciplinary field that considers the human body as a system, composed of multiple parts and of complex relationships at multiple levels, and further integrated into an environment. Exploring Systems Medicine implies understanding and combining concepts coming from diametral different fields, including medicine, biology, statistics, modeling and simulation, and data science. Such heterogeneity leads to semantic issues, which may slow down implementation and fruitful interaction between these highly diverse fields. Methods: In this review, we collect and explain more than100 terms related to Systems Medicine. These include both modeling and data science terms and basic systems medicine terms, along with some synthetic definitions, examples of applications, and lists of relevant references. Results: This glossary aims at being a first aid kit for the Systems Medicine researcher facing an unfamiliar term, where he/she can get a first understanding of them, and, more importantly, examples and references for digging into the topic.
Collapse
Affiliation(s)
- Massimiliano Zanin
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Nadim A.A. Aitya
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - José Basilio
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Jan Baumbach
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Arriel Benis
- Faculty of Technology Management, Holon Institute of Technology (HIT), Holon, Israel
| | - Chandan K. Behera
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Magda Bucholc
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Filippo Castiglione
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | - Ioanna Chouvarda
- Lab of Computing, Medical Informatics, and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Blandine Comte
- Université Clermont Auvergne, INRAE, UNH, Plateforme d'Exploration du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Tien-Tuan Dao
- Biomechanics and Bioengineering Laboratory (UMR CNRS 7338), Université de Technologie de Compiègne, Compiègne, France
- Labex MS2T “Control of Technological Systems-of-Systems,” CNRS and Université de Technologie de Compiègne, Compiègne, France
| | - Xuemei Ding
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Estelle Pujos-Guillot
- Université Clermont Auvergne, INRAE, UNH, Plateforme d'Exploration du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Nenad Filipovic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia
- Bioengineering Research and Development Center (BioIRC), Kragujevac, Serbia
- Steinbeis Advanced Risk Technologies Institute doo Kragujevac, Kragujevac, Serbia
| | - David P. Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, National University of Ireland, Galway, Republic of Ireland
| | - David H. Glass
- School of Computing, Ulster University, Ulster, United Kingdom
| | - Nissim Harel
- Faculty of Sciences, Holon Institute of Technology (HIT), Holon, Israel
| | - Tomas Iesmantas
- Department of Mathematics and Natural Sciences, Kaunas University of Technology, Kaunas, Lithuania
| | - Ilinka Ivanoska
- Faculty of Computer Science and Engineering, Ss. Cyril and Methodius University, Skopje, Macedonia
| | - Alok Joshi
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Karim Zouaoui Boudjeltia
- Laboratory of Experimental Medicine (ULB 222), Medicine Faculty, Université libre de Bruxelles, CHU de Charleroi, Charleroi, Belgium
| | - Badr Kaoui
- Biomechanics and Bioengineering Laboratory (UMR CNRS 7338), Université de Technologie de Compiègne, Compiègne, France
- Labex MS2T “Control of Technological Systems-of-Systems,” CNRS and Université de Technologie de Compiègne, Compiègne, France
| | - Daman Kaur
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Ulster, United Kingdom
| | - Liam P. Maguire
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Paula L. McClean
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Ulster, United Kingdom
| | - Niamh McCombe
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - João Luís de Miranda
- Escola Superior de Tecnologia e Gestão, Instituto Politécnico de Portalegre, Portalegre, Portugal
- Centro de Recursos Naturais e Ambiente (CERENA), Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Annikka Polster
- Centre for Molecular Medicine Norway (NCMM), Forskningparken, Oslo, Norway
| | - Girijesh Prasad
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Damjana Rozman
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ioan Sacala
- Faculty of Automatic Control and Computers, University Politehnica of Bucharest, Bucharest, Romania
| | - Jose M. Sanchez-Bornot
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Johannes A. Schmid
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Trevor Sharp
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Jordi Solé-Casals
- Data and Signal Processing Research Group, University of Vic–Central University of Catalonia, Vic, Spain
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Vojtěch Spiwok
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic
| | - George M. Spyrou
- The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Egils Stalidzans
- Computational Systems Biology Group, Institute of Microbiology and Biotechnology, University of Latvia, Riga, Latvia
| | - Blaž Stres
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Civil and Geodetic Engineering, University of Ljubljana, Ljubljana, Slovenia
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Tijana Sustersic
- Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia
- Bioengineering Research and Development Center (BioIRC), Kragujevac, Serbia
- Steinbeis Advanced Risk Technologies Institute doo Kragujevac, Kragujevac, Serbia
| | - Ioannis Symeonidis
- Center for Research and Technology Hellas, Hellenic Institute of Transport, Thessaloniki, Greece
| | - Paolo Tieri
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | - Stephen Todd
- Altnagelvin Area Hospital, Western Health and Social Care Trust, Altnagelvin, United Kingdom
| | - Kristel Van Steen
- BIO3-Systems Genetics, GIGA-R, University of Liege, Liege, Belgium
- BIO3-Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Da-Hui Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, and School of Systems Science, Beijing Normal University, Beijing, China
| | - Haiying Wang
- School of Computing, Ulster University, Ulster, United Kingdom
| | - Hui Wang
- School of Computing, Ulster University, Ulster, United Kingdom
| | - Steven Watterson
- Northern Ireland Centre for Stratified Medicine, Ulster University, Londonderry, United Kingdom
| | - KongFatt Wong-Lin
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Su Yang
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Ulster, United Kingdom
| | - Xin Zou
- Shanghai Centre for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Harald H.H.W. Schmidt
- Faculty of Health, Medicine & Life Science, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
29
|
Rizi AK, Zamani M, Shirazi A, Jafari GR, Kertész J. Stability of Imbalanced Triangles in Gene Regulatory Networks of Cancerous and Normal Cells. Front Physiol 2021; 11:573732. [PMID: 33551827 PMCID: PMC7854919 DOI: 10.3389/fphys.2020.573732] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Genes communicate with each other through different regulatory effects, which lead to the emergence of complex network structures in cells, and such structures are expected to be different for normal and cancerous cells. To study these differences, we have investigated the Gene Regulatory Network (GRN) of cells as inferred from RNA-sequencing data. The GRN is a signed weighted network corresponding to the inductive or inhibitory interactions. Here we focus on a particular of motifs in the GRN, the triangles, which are imbalanced if the number of negative interactions is odd. By studying the stability of imbalanced triangles in the GRN, we show that the network of cancerous cells has fewer imbalanced triangles compared to normal cells. Moreover, in the normal cells, imbalanced triangles are isolated from the main part of the network, while such motifs are part of the network's giant component in cancerous cells. Our result demonstrates that due to genes' collective behavior the structure of the complex networks is different in cancerous cells from those in normal ones.
Collapse
Affiliation(s)
| | - Mina Zamani
- Physics Department, Shahid Beheshti University, Tehran, Iran
| | | | - G Reza Jafari
- Physics Department, Shahid Beheshti University, Tehran, Iran.,Department of Network and Data Science, Central European University, Budapest, Hungary
| | - János Kertész
- Physics Department, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
30
|
Musilova J, Sedlar K. Tools for time-course simulation in systems biology: a brief overview. Brief Bioinform 2021; 22:6076933. [PMID: 33423059 DOI: 10.1093/bib/bbaa392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 11/13/2022] Open
Abstract
Dynamic modeling of biological systems is essential for understanding all properties of a given organism as it allows us to look not only at the static picture of an organism but also at its behavior under various conditions. With the increasing amount of experimental data, the number of tools that enable dynamic analysis also grows. However, various tools are based on different approaches, use different types of data and offer different functions for analyses; so it can be difficult to choose the most suitable tool for a selected type of model. Here, we bring a brief overview containing descriptions of 50 tools for the reconstruction of biological models, their time-course simulation and dynamic analysis. We examined each tool using test data and divided them based on the qualitative and quantitative nature of the mathematical apparatus they use.
Collapse
Affiliation(s)
- Jana Musilova
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czechia
| | - Karel Sedlar
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czechia
| |
Collapse
|
31
|
A review of methods for the reconstruction and analysis of integrated genome-scale models of metabolism and regulation. Biochem Soc Trans 2020; 48:1889-1903. [DOI: 10.1042/bst20190840] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/16/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
The current survey aims to describe the main methodologies for extending the reconstruction and analysis of genome-scale metabolic models and phenotype simulation with Flux Balance Analysis mathematical frameworks, via the integration of Transcriptional Regulatory Networks and/or gene expression data. Although the surveyed methods are aimed at improving phenotype simulations obtained from these models, the perspective of reconstructing integrated genome-scale models of metabolism and gene expression for diverse prokaryotes is still an open challenge.
Collapse
|
32
|
Randhawa V, Pathania S. Advancing from protein interactomes and gene co-expression networks towards multi-omics-based composite networks: approaches for predicting and extracting biological knowledge. Brief Funct Genomics 2020; 19:364-376. [PMID: 32678894 DOI: 10.1093/bfgp/elaa015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/31/2020] [Accepted: 06/15/2020] [Indexed: 01/17/2023] Open
Abstract
Prediction of biological interaction networks from single-omics data has been extensively implemented to understand various aspects of biological systems. However, more recently, there is a growing interest in integrating multi-omics datasets for the prediction of interactomes that provide a global view of biological systems with higher descriptive capability, as compared to single omics. In this review, we have discussed various computational approaches implemented to infer and analyze two of the most important and well studied interactomes: protein-protein interaction networks and gene co-expression networks. We have explicitly focused on recent methods and pipelines implemented to infer and extract biologically important information from these interactomes, starting from utilizing single-omics data and then progressing towards multi-omics data. Accordingly, recent examples and case studies are also briefly discussed. Overall, this review will provide a proper understanding of the latest developments in protein and gene network modelling and will also help in extracting practical knowledge from them.
Collapse
Affiliation(s)
- Vinay Randhawa
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Shivalika Pathania
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
33
|
Saint-Antoine MM, Singh A. Network inference in systems biology: recent developments, challenges, and applications. Curr Opin Biotechnol 2020; 63:89-98. [PMID: 31927423 PMCID: PMC7308210 DOI: 10.1016/j.copbio.2019.12.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
One of the most interesting, difficult, and potentially useful topics in computational biology is the inference of gene regulatory networks (GRNs) from expression data. Although researchers have been working on this topic for more than a decade and much progress has been made, it remains an unsolved problem and even the most sophisticated inference algorithms are far from perfect. In this paper, we review the latest developments in network inference, including state-of-the-art algorithms like PIDC, Phixer, and more. We also discuss unsolved computational challenges, including the optimal combination of algorithms, integration of multiple data sources, and pseudo-temporal ordering of static expression data. Lastly, we discuss some exciting applications of network inference in cancer research, and provide a list of useful software tools for researchers hoping to conduct their own network inference analyses.
Collapse
Affiliation(s)
- Michael M Saint-Antoine
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware 19716, USA
| | - Abhyudai Singh
- Electrical and Computer Engineering, University of Delaware, Newark, Delaware 19716, USA.
| |
Collapse
|
34
|
Chen YC, Desplan C. Gene regulatory networks during the development of the Drosophila visual system. Curr Top Dev Biol 2020; 139:89-125. [PMID: 32450970 DOI: 10.1016/bs.ctdb.2020.02.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Drosophila visual system integrates input from 800 ommatidia and extracts different features in stereotypically connected optic ganglia. The development of the Drosophila visual system is controlled by gene regulatory networks that control the number of precursor cells, generate neuronal diversity by integrating spatial and temporal information, coordinate the timing of retinal and optic lobe cell differentiation, and determine distinct synaptic targets of each cell type. In this chapter, we describe the known gene regulatory networks involved in the development of the different parts of the visual system and explore general components in these gene networks. Finally, we discuss the advantages of the fly visual system as a model for gene regulatory network discovery in the era of single-cell transcriptomics.
Collapse
Affiliation(s)
- Yen-Chung Chen
- Department of Biology, New York University, New York, NY, United States
| | - Claude Desplan
- Department of Biology, New York University, New York, NY, United States.
| |
Collapse
|
35
|
Dieckmann L, Cole S, Kumsta R. Stress genomics revisited: gene co-expression analysis identifies molecular signatures associated with childhood adversity. Transl Psychiatry 2020; 10:34. [PMID: 32066736 PMCID: PMC7026041 DOI: 10.1038/s41398-020-0730-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
Childhood adversity is related to an increased risk for psychopathology in adulthood. Altered regulation of stress response systems, as well as the changes in stress-immune interplay have been suggested as potential mechanisms underlying these long-term effects. We have previously shown altered transcriptional responses to acute psychosocial stress in adults reporting the experience of childhood adversity. Here, we extend these analyses using a network approach. We performed a co-expression network analysis of genome-wide mRNA data derived from isolated monocytes, sampled 3 h after stress exposure from healthy adults, who experienced childhood adversity and a matched control group without adverse childhood experiences. Thirteen co-expression modules were identified, of which four modules were enriched for genes related to immune system function. Gene set enrichment analysis showed differential module activity between the early adversity and control group. In line with previous findings reporting a pro-inflammatory bias following childhood adversity, one module included genes associated with pro-inflammatory function (hub genes: IL6, TM4SF1, ADAMTS4, CYR61, CCDC3), more strongly expressed in the early adversity group. Another module downregulated in the early adversity group was related to platelet activation and wound healing (hub genes: GP9, CMTM5, TUBB1, GNG11, PF4), and resembled a co-expression module previously found over-expressed in post-traumatic stress disorder resilient soldiers. These discovery analysis results provide a system wide and more holistic understanding of gene expression programs associated with childhood adversity. Furthermore, identified hub genes can be used in directed hypothesis testing in future studies.
Collapse
Affiliation(s)
- Linda Dieckmann
- grid.5570.70000 0004 0490 981XDepartment of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Steve Cole
- grid.19006.3e0000 0000 9632 6718Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA USA
| | - Robert Kumsta
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
36
|
Chen X, Li M, Zheng R, Wu FX, Wang J. D3GRN: a data driven dynamic network construction method to infer gene regulatory networks. BMC Genomics 2019; 20:929. [PMID: 31881937 PMCID: PMC6933629 DOI: 10.1186/s12864-019-6298-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND To infer gene regulatory networks (GRNs) from gene-expression data is still a fundamental and challenging problem in systems biology. Several existing algorithms formulate GRNs inference as a regression problem and obtain the network with an ensemble strategy. Recent studies on data driven dynamic network construction provide us a new perspective to solve the regression problem. RESULTS In this study, we propose a data driven dynamic network construction method to infer gene regulatory network (D3GRN), which transforms the regulatory relationship of each target gene into functional decomposition problem and solves each sub problem by using the Algorithm for Revealing Network Interactions (ARNI). To remedy the limitation of ARNI in constructing networks solely from the unit level, a bootstrapping and area based scoring method is taken to infer the final network. On DREAM4 and DREAM5 benchmark datasets, D3GRN performs competitively with the state-of-the-art algorithms in terms of AUPR. CONCLUSIONS We have proposed a novel data driven dynamic network construction method by combining ARNI with bootstrapping and area based scoring strategy. The proposed method performs well on the benchmark datasets, contributing as a competitive method to infer gene regulatory networks in a new perspective.
Collapse
Affiliation(s)
- Xiang Chen
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha, China.
| | - Ruiqing Zheng
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Fang-Xiang Wu
- Department of Mechanical Engineering and Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, China
| |
Collapse
|
37
|
Chen C, Jiang L, Fu G, Wang M, Wang Y, Shen B, Liu Z, Wang Z, Hou W, Berceli SA, Wu R. An omnidirectional visualization model of personalized gene regulatory networks. NPJ Syst Biol Appl 2019; 5:38. [PMID: 31632690 PMCID: PMC6789114 DOI: 10.1038/s41540-019-0116-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/18/2019] [Indexed: 01/09/2023] Open
Abstract
Gene regulatory networks (GRNs) have been widely used as a fundamental tool to reveal the genomic mechanisms that underlie the individual's response to environmental and developmental cues. Standard approaches infer GRNs as holistic graphs of gene co-expression, but such graphs cannot quantify how gene-gene interactions vary among individuals and how they alter structurally across spatiotemporal gradients. Here, we develop a general framework for inferring informative, dynamic, omnidirectional, and personalized networks (idopNetworks) from routine transcriptional experiments. This framework is constructed by a system of quasi-dynamic ordinary differential equations (qdODEs) derived from the combination of ecological and evolutionary theories. We reconstruct idopNetworks using genomic data from a surgical experiment and illustrate how network structure is associated with surgical response to infrainguinal vein bypass grafting and the outcome of grafting. idopNetworks may shed light on genotype-phenotype relationships and provide valuable information for personalized medicine.
Collapse
Affiliation(s)
- Chixiang Chen
- Center for Statistical Genetics, Departments of Public Health Sciences and Statistics, Pennsylvania State University, Hershey, PA 17033 USA
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Libo Jiang
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083 China
| | - Guifang Fu
- Department of Mathematical Sciences, SUNY Binghamton University, Binghamton, NY 13902 USA
| | - Ming Wang
- Center for Statistical Genetics, Departments of Public Health Sciences and Statistics, Pennsylvania State University, Hershey, PA 17033 USA
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Yaqun Wang
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ 08854 USA
| | - Biyi Shen
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Zhenqiu Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Heaven, CT 06520 USA
| | - Wei Hou
- Department of Family, Population & Preventive Medicine, Stony Brook School of Medicine, Stony Brook, NY 11794 USA
| | - Scott A. Berceli
- Malcom Randall VA Medical Center, Gainesville, FL 32610 USA
- Department of Surgery, University of Florida, Box 100128, Gainesville, FL 32610 USA
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32610 USA
| | - Rongling Wu
- Center for Statistical Genetics, Departments of Public Health Sciences and Statistics, Pennsylvania State University, Hershey, PA 17033 USA
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033 USA
| |
Collapse
|
38
|
Park S, Hwang D, Yeo YS, Kim H, Kang J. CONFIGURE: A pipeline for identifying context specific regulatory modules from gene expression data and its application to breast cancer. BMC Med Genomics 2019; 12:97. [PMID: 31296219 PMCID: PMC6624175 DOI: 10.1186/s12920-019-0515-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Gene expression data is widely used for identifying subtypes of diseases such as cancer. Differentially expressed gene analysis and gene set enrichment analysis are widely used for identifying biological mechanisms at the gene level and gene set level, respectively. However, the results of differentially expressed gene analysis are difficult to interpret and gene set enrichment analysis does not consider the interactions among genes in a gene set. RESULTS We present CONFIGURE, a pipeline that identifies context specific regulatory modules from gene expression data. First, CONFIGURE takes gene expression data and context label information as inputs and constructs regulatory modules. Then, CONFIGURE makes a regulatory module enrichment score (RMES) matrix of enrichment scores of the regulatory modules on samples using the single-sample GSEA method. CONFIGURE calculates the importance scores of the regulatory modules on each context to rank the regulatory modules. We evaluated CONFIGURE on the Cancer Genome Atlas (TCGA) breast cancer RNA-seq dataset to determine whether it can produce biologically meaningful regulatory modules for breast cancer subtypes. We first evaluated whether RMESs are useful for differentiating breast cancer subtypes using a multi-class classifier and one-vs-rest binary SVM classifiers. The multi-class and one-vs-rest binary classifiers were trained using the RMESs as features and outperformed baseline classifiers. Furthermore, we conducted literature surveys on the basal-like type specific regulatory modules obtained by CONFIGURE and showed that highly ranked modules were associated with the phenotypes of basal-like type breast cancers. CONCLUSIONS We showed that enrichment scores of regulatory modules are useful for differentiating breast cancer subtypes and validated the basal-like type specific regulatory modules by literature surveys. In doing so, we found regulatory module candidates that have not been reported in previous literature. This demonstrates that CONFIGURE can be used to predict novel regulatory markers which can be validated by downstream wet lab experiments. We validated CONFIGURE on the breast cancer RNA-seq dataset in this work but CONFIGURE can be applied to any gene expression dataset containing context information.
Collapse
Affiliation(s)
- Sungjoon Park
- Department of Computer Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Doyeong Hwang
- Department of Computer Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Yoon Sun Yeo
- Department of Computer Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jaewoo Kang
- Department of Computer Science and Engineering, Korea University, Seoul, Republic of Korea. .,Interdisciplinary Graduate Program in Bioinformatics, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Gupta P, Singh SK. Gene Regulatory Networks: Current Updates and Applications in Plant Biology. ENERGY, ENVIRONMENT, AND SUSTAINABILITY 2019. [DOI: 10.1007/978-981-15-0690-1_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|