151
|
Gysi DM, Nowick K. Construction, comparison and evolution of networks in life sciences and other disciplines. J R Soc Interface 2020; 17:20190610. [PMID: 32370689 PMCID: PMC7276545 DOI: 10.1098/rsif.2019.0610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Network approaches have become pervasive in many research fields. They allow for a more comprehensive understanding of complex relationships between entities as well as their group-level properties and dynamics. Many networks change over time, be it within seconds or millions of years, depending on the nature of the network. Our focus will be on comparative network analyses in life sciences, where deciphering temporal network changes is a core interest of molecular, ecological, neuropsychological and evolutionary biologists. Further, we will take a journey through different disciplines, such as social sciences, finance and computational gastronomy, to present commonalities and differences in how networks change and can be analysed. Finally, we envision how borrowing ideas from these disciplines could enrich the future of life science research.
Collapse
Affiliation(s)
- Deisy Morselli Gysi
- Department of Computer Science, Interdisciplinary Center of Bioinformatics, University of Leipzig, 04109 Leipzig, Germany
- Swarm Intelligence and Complex Systems Group, Faculty of Mathematics and Computer Science, University of Leipzig, 04109 Leipzig, Germany
- Center for Complex Networks Research, Northeastern University, 177 Huntington Avenue, Boston, MA 02115, USA
| | - Katja Nowick
- Human Biology Group, Institute for Biology, Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Königin-Luise-Straβe 1-3, 14195 Berlin, Germany
| |
Collapse
|
152
|
Amanatidou AI, Nastou KC, Tsitsilonis OE, Iconomidou VA. Visualization and analysis of the interaction network of proteins associated with blood-cell targeting autoimmune diseases. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165714. [PMID: 32023482 DOI: 10.1016/j.bbadis.2020.165714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/17/2022]
|
153
|
Abstract
Understanding the individual and joint contribution of multiple protein levels toward a phenotype requires precise and tunable multigene expression control. Here we introduce a pair of mammalian synthetic gene circuits that linearly and orthogonally control the expression of two reporter genes in mammalian cells with low variability in response to chemical inducers introduced into the growth medium. These gene expression systems can be used to simultaneously probe the individual and joint effects of two gene product concentrations on a cellular phenotype in basic research or biomedical applications.
Collapse
Affiliation(s)
- Mariola Szenk
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794, United States
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Terrence Yim
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Gábor Balázsi
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794, United States
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
154
|
Ko K, Lee CW, Nam S, Ahn SV, Bae JH, Ban CY, Yoo J, Park J, Han HW. Epidemiological Characterization of a Directed and Weighted Disease Network Using Data From a Cohort of One Million Patients: Network Analysis. J Med Internet Res 2020; 22:e15196. [PMID: 32271154 PMCID: PMC7180516 DOI: 10.2196/15196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/08/2019] [Accepted: 01/24/2020] [Indexed: 11/25/2022] Open
Abstract
Background In the past 20 years, various methods have been introduced to construct disease networks. However, established disease networks have not been clinically useful to date because of differences among demographic factors, as well as the temporal order and intensity among disease-disease associations. Objective This study sought to investigate the overall patterns of the associations among diseases; network properties, such as clustering, degree, and strength; and the relationship between the structure of disease networks and demographic factors. Methods We used National Health Insurance Service-National Sample Cohort (NHIS-NSC) data from the Republic of Korea, which included the time series insurance information of 1 million out of 50 million Korean (approximately 2%) patients obtained between 2002 and 2013. After setting the observation and outcome periods, we selected only 520 common Korean Classification of Disease, sixth revision codes that were the most prevalent diagnoses, making up approximately 80% of the cases, for statistical validity. Using these data, we constructed a directional and weighted temporal network that considered both demographic factors and network properties. Results Our disease network contained 294 nodes and 3085 edges, a relative risk value of more than 4, and a false discovery rate-adjusted P value of <.001. Interestingly, our network presented four large clusters. Analysis of the network topology revealed a stronger correlation between in-strength and out-strength than between in-degree and out-degree. Further, the mean age of each disease population was related to the position along the regression line of the out/in-strength plot. Conversely, clustering analysis suggested that our network boasted four large clusters with different sex, age, and disease categories. Conclusions We constructed a directional and weighted disease network visualizing demographic factors. Our proposed disease network model is expected to be a valuable tool for use by early clinical researchers seeking to explore the relationships among diseases in the future.
Collapse
Affiliation(s)
- Kyungmin Ko
- Department of Biomedical Informatics, CHA University of Medicine, Seongnam, Republic of Korea.,Department of Pathology, Medstar Georgetown University Hospital, Washington, DC, WA, United States
| | - Chae Won Lee
- Department of Biomedical Informatics, CHA University of Medicine, Seongnam, Republic of Korea.,Institute of Basic Medical Sciences, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Sangmin Nam
- Department of Ophthalmology, CHA Bundang Medical Center, Seongnam, Republic of Korea
| | - Song Vogue Ahn
- Department of Health Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Jung Ho Bae
- Department of Internal Medicine, Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Republic of Korea
| | - Chi Yong Ban
- Department of Biomedical Informatics, CHA University of Medicine, Seongnam, Republic of Korea.,Institute of Basic Medical Sciences, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Jongman Yoo
- Institute of Basic Medical Sciences, School of Medicine, CHA University, Seongnam, Republic of Korea.,Department of Microbiology, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Jungmin Park
- Department of Nursing, School of Nursing, Hanyang University, Seoul, Republic of Korea
| | - Hyun Wook Han
- Department of Biomedical Informatics, CHA University of Medicine, Seongnam, Republic of Korea.,Institute of Basic Medical Sciences, School of Medicine, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
155
|
Ding J, Li Y, Zhang Y, Fan B, Li Q, Zhang J, Zhang J. Identification of key lncRNAs in the tumorigenesis of intraductal pancreatic mucinous neoplasm by coexpression network analysis. Cancer Med 2020; 9:3840-3851. [PMID: 32239802 PMCID: PMC7286472 DOI: 10.1002/cam4.2927] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 01/15/2020] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
Intraductal papillary mucinous neoplasm (IPMN) is an intraepithelial precancerous lesion of pancreatic ductal adenocarcinoma (PDAC) that progresses from adenoma to carcinoma, and long noncoding RNAs (lncRNA) might be involved in the tumorigenesis. In this study, we obtained the expression profiles of more than 4000 lncRNAs by probe reannotation of a microarray dataset. As a correlation network‐based systems biology method, weighted gene coexpression network analysis (WGCNA) was used to find clusters of highly correlated lncRNAs in the tumorigenesis of IPMN, which covered four stepwise stages from normal main pancreatic duct to invasive IPMN. In the most relevant module (R2 = −0.75 and P = 5E‐05), three hub lncRNAs were identified (HAND2‐AS1, CTD‐2033D15.2, and lncRNA‐TFG). HAND2‐AS1 and CTD‐2033D15.2 were negatively correlated with the tumorigenesis (P in one‐way ANOVA test = 1.45E‐07 and 1.39E‐0.5), while lncRNA‐TFG were positively correlated with the tumorigenesis (P = 3.99E‐08). The validation set reached consistent results (P = 2.66E‐03 in HAND2‐AS1, 1.47E‐04 in CTD‐2033D15.2 and 6.23E‐08 in lncRNA‐TFG). In functional enrichment analysis, the target genes of microRNAs targeting also these lncRNAs were overlapped in multiple biological processes, pathways and malignant diseases including pancreatic cancer. In survival analysis, patients with higher expression of HAND2‐AS1‐targeted and CTD‐2033D15.2‐targeted microRNAs showed a significantly poorer prognosis in PDAC, while high expression of lncRNA‐TFG‐targeted microRNAs demonstrated an obviously better prognosis (log‐rank P < .05). In conclusion, by coexpression network analysis of the lncRNA profiles, three key lncRNAs were identified in association with the tumorigenesis of IPMN, and those lncRNAs might act as early diagnostic biomarkers or therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Jun Ding
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| | - Yi Li
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| | - Yong Zhang
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| | - Bin Fan
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| | - Qinghe Li
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| | - Jian Zhang
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| | - Jiayao Zhang
- Department of Hepatobiliary SurgeryThe Central Hospital of Enshi Autonomous PrefectureEnshiChina
| |
Collapse
|
156
|
Hewitt SK, Duangrattanalert K, Burgis T, Zeef LAH, Naseeb S, Delneri D. Plasticity of Mitochondrial DNA Inheritance and its Impact on Nuclear Gene Transcription in Yeast Hybrids. Microorganisms 2020; 8:microorganisms8040494. [PMID: 32244414 PMCID: PMC7232527 DOI: 10.3390/microorganisms8040494] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/25/2020] [Accepted: 03/29/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial DNA (mtDNA) in yeast is biparentally inherited, but colonies rapidly lose one type of parental mtDNA, thus becoming homoplasmic. Therefore, hybrids between the yeast species possess two homologous nuclear genomes, but only one type of mitochondrial DNA. We hypothesise that the choice of mtDNA retention is influenced by its contribution to hybrid fitness in different environments, and the allelic expression of the two nuclear sub-genomes is affected by the presence of different mtDNAs in hybrids. Saccharomyces cerevisiae/S. uvarum hybrids preferentially retained S. uvarum mtDNA when formed on rich media at colder temperatures, while S. cerevisiae mtDNA was primarily retained on non-fermentable carbon source, at any temperature. Transcriptome data for hybrids harbouring different mtDNA showed a strong environmentally dependent allele preference, which was more important in respiratory conditions. Co-expression analysis for specific biological functions revealed a clear pattern of concerted allelic transcription within the same allele type, which supports the notion that the hybrid cell works preferentially with one set of parental alleles (or the other) for different cellular functions. Given that the type of mtDNA retained in hybrids affects both nuclear expression and fitness, it might play a role in driving hybrid genome evolution in terms of gene retention and loss.
Collapse
Affiliation(s)
- Sarah K Hewitt
- Manchester Institute of Biotechnology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M1 7DN, UK
- Division of Evolution and Genomic Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Kobchai Duangrattanalert
- Manchester Institute of Biotechnology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M1 7DN, UK
- Division of Evolution and Genomic Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Tim Burgis
- Division of Evolution and Genomic Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Leo A H Zeef
- Division of Evolution and Genomic Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Samina Naseeb
- Manchester Institute of Biotechnology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M1 7DN, UK
- Division of Evolution and Genomic Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Daniela Delneri
- Manchester Institute of Biotechnology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M1 7DN, UK
- Division of Evolution and Genomic Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
157
|
Kelly J, Moyeed R, Carroll C, Luo S, Li X. Genetic networks in Parkinson's and Alzheimer's disease. Aging (Albany NY) 2020; 12:5221-5243. [PMID: 32205467 PMCID: PMC7138567 DOI: 10.18632/aging.102943] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Parkinson’s disease (PD) and Alzheimer’s disease (AD) are the most common neurodegenerative diseases and there is increasing evidence that they share common physiological and pathological links. Here we have conducted the largest network analysis of PD and AD based on their gene expressions in blood to date. We identified modules that were not preserved between disease and healthy control (HC) networks, and important hub genes and transcription factors (TFs) in these modules. We highlighted that the PD module not preserved in HCs was associated with insulin resistance, and HDAC6 was identified as a hub gene in this module which may have the role of influencing tau phosphorylation and autophagic flux in neurodegenerative disease. The AD module associated with regulation of lipolysis in adipocytes and neuroactive ligand-receptor interaction was not preserved in healthy and mild cognitive impairment networks and the key hubs TRPC5 and BRAP identified as potential targets for therapeutic treatments of AD. Our study demonstrated that PD and AD share common disrupted genetics and identified novel pathways, hub genes and TFs that may be new areas for mechanistic study and important targets in both diseases.
Collapse
Affiliation(s)
- Jack Kelly
- Faculty of Health: Medicine, Dentistry and Human Sciences, Plymouth University, Plymouth PL6 8BU, UK
| | - Rana Moyeed
- Faculty of Science and Engineering, Plymouth University, Plymouth PL6 8BU, UK
| | - Camille Carroll
- Faculty of Health: Medicine, Dentistry and Human Sciences, Plymouth University, Plymouth PL6 8BU, UK
| | - Shouqing Luo
- Faculty of Health: Medicine, Dentistry and Human Sciences, Plymouth University, Plymouth PL6 8BU, UK
| | - Xinzhong Li
- School of Science, Engineering and Design, Teesside University, Middlesbrough TS1 3BX, UK
| |
Collapse
|
158
|
Park HM, Park N, Myaeng SH, Kang U. PACC: Large scale connected component computation on Hadoop and Spark. PLoS One 2020; 15:e0229936. [PMID: 32187232 PMCID: PMC7080249 DOI: 10.1371/journal.pone.0229936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/17/2020] [Indexed: 11/19/2022] Open
Abstract
A connected component in a graph is a set of nodes linked to each other by paths. The problem of finding connected components has been applied to diverse graph analysis tasks such as graph partitioning, graph compression, and pattern recognition. Several distributed algorithms have been proposed to find connected components in enormous graphs. Ironically, the distributed algorithms do not scale enough due to unnecessary data IO & processing, massive intermediate data, numerous rounds of computations, and load balancing issues. In this paper, we propose a fast and scalable distributed algorithm PACC (Partition-Aware Connected Components) for connected component computation based on three key techniques: two-step processing of partitioning & computation, edge filtering, and sketching. PACC considerably shrinks the size of intermediate data, the size of input graph, and the number of rounds without suffering from load balancing issues. PACC performs 2.9 to 10.7 times faster on real-world graphs compared to the state-of-the-art MapReduce and Spark algorithms.
Collapse
Affiliation(s)
| | - Namyong Park
- Carnegie Mellon University, Pittsburgh, PA, United States of America
| | | | - U Kang
- Seoul National University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
159
|
Brewster JT, Thiabaud GD, Harvey P, Zafar H, Reuther JF, Dell’Acqua S, Johnson RM, Root HD, Metola P, Jasanoff A, Casella L, Sessler JL. Metallotexaphyrins as MRI-Active Catalytic Antioxidants for Neurodegenerative Disease: A Study on Alzheimer's Disease. Chem 2020; 6:703-724. [PMID: 32201749 PMCID: PMC7074011 DOI: 10.1016/j.chempr.2019.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/28/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023]
Abstract
The complex etiology of neurodegeneration continues to stifle efforts to develop effective therapeutics. New agents elucidating key pathways causing neurodegeneration might serve to increase our understanding and potentially lead to improved treatments. Here, we demonstrate that a water-soluble manganese(II) texaphyrin (MMn) is a suitable magnetic resonance imaging (MRI) contrast agent for detecting larger amyloid beta constructs. The imaging potential of MMn was inferred on the basis of in vitro studies and in vivo detection in Alzheimer's disease C. elegans models via MRI and ICP-MS. In vitro antioxidant- and cellular-based assays provide support for the notion that this porphyrin analog shows promise as a therapeutic agent able to mitigate the oxidative and nitrative toxic effects considered causal in neurodegeneration. The present report marks the first elaboration of an MRI-active metalloantioxidant that confers diagnostic and therapeutic benefit in Alzheimer's disease models without conjugation of a radioisotope, targeting moiety, or therapeutic payload.
Collapse
Affiliation(s)
- James T. Brewster
- Department of Chemistry, the University of Texas at Austin, Austin, TX 78712-1224, USA
| | - Gregory D. Thiabaud
- Department of Chemistry, the University of Texas at Austin, Austin, TX 78712-1224, USA
| | - Peter Harvey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Sir Peter Mansfield Imaging Centre, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Hadiqa Zafar
- Department of Chemistry, the University of Texas at Austin, Austin, TX 78712-1224, USA
| | - James F. Reuther
- Department of Chemistry, the University of Texas at Austin, Austin, TX 78712-1224, USA
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Simone Dell’Acqua
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Rachel M. Johnson
- Accelerated Research Initiative, University of Texas at Austin, Austin, TX 78712, USA
| | - Harrison D. Root
- Department of Chemistry, the University of Texas at Austin, Austin, TX 78712-1224, USA
| | - Pedro Metola
- Accelerated Research Initiative, University of Texas at Austin, Austin, TX 78712, USA
| | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Luigi Casella
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Jonathan L. Sessler
- Department of Chemistry, the University of Texas at Austin, Austin, TX 78712-1224, USA
- Center for Supramolecular Chemistry and Catalysis, Shanghai University, Shanghai, China
| |
Collapse
|
160
|
Schwab JD, Kühlwein SD, Ikonomi N, Kühl M, Kestler HA. Concepts in Boolean network modeling: What do they all mean? Comput Struct Biotechnol J 2020; 18:571-582. [PMID: 32257043 PMCID: PMC7096748 DOI: 10.1016/j.csbj.2020.03.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/27/2020] [Accepted: 03/01/2020] [Indexed: 12/02/2022] Open
Abstract
Boolean network models are one of the simplest models to study complex dynamic behavior in biological systems. They can be applied to unravel the mechanisms regulating the properties of the system or to identify promising intervention targets. Since its introduction by Stuart Kauffman in 1969 for describing gene regulatory networks, various biologically based networks and tools for their analysis were developed. Here, we summarize and explain the concepts for Boolean network modeling. We also present application examples and guidelines to work with and analyze Boolean network models.
Collapse
Affiliation(s)
- Julian D Schwab
- Institute of Medical Systems Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Silke D Kühlwein
- Institute of Medical Systems Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Michael Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
161
|
Metzig C, Colijn C. A Maximum Entropy Method for the Prediction of Size Distributions. ENTROPY 2020; 22:e22030312. [PMID: 33286086 PMCID: PMC7516768 DOI: 10.3390/e22030312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/16/2020] [Accepted: 03/05/2020] [Indexed: 12/04/2022]
Abstract
We propose a method to derive the stationary size distributions of a system, and the degree distributions of networks, using maximisation of the Gibbs-Shannon entropy. We apply this to a preferential attachment-type algorithm for systems of constant size, which contains exit of balls and urns (or nodes and edges for the network case). Knowing mean size (degree) and turnover rate, the power law exponent and exponential cutoff can be derived. Our results are confirmed by simulations and by computation of exact probabilities. We also apply this entropy method to reproduce existing results like the Maxwell-Boltzmann distribution for the velocity of gas particles, the Barabasi-Albert model and multiplicative noise systems.
Collapse
Affiliation(s)
- Cornelia Metzig
- Business School, Imperial College London, London SW7 2AZ, UK
- School of Electronic Engineering and Computer Science, Queen Mary University, London E1 7NS, UK
- Correspondence:
| | - Caroline Colijn
- Department of Mathematics, Imperial College London, London SW7 2AZ, UK;
- Department of Mathematics, Simon Fraser University, Surrey, BC V3T0A3, Canada
| |
Collapse
|
162
|
Di Lollo V, Canciello A, Orsini M, Bernabò N, Ancora M, Di Federico M, Curini V, Mattioli M, Russo V, Mauro A, Cammà C, Barboni B. Transcriptomic and computational analysis identified LPA metabolism, KLHL14 and KCNE3 as novel regulators of Epithelial-Mesenchymal Transition. Sci Rep 2020; 10:4180. [PMID: 32144311 PMCID: PMC7060278 DOI: 10.1038/s41598-020-61017-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a complex biological program between physiology and pathology. Here, amniotic epithelial cells (AEC) were used as in vitro model of transiently inducible EMT in order to evaluate the transcriptional insights underlying this process. Therefore, RNA-seq was used to identify the differentially expressed genes and enrichment analyses were carried out to assess the intracellular pathways involved. As a result, molecules exclusively expressed in AEC that experienced EMT (GSTA1-1 and GSTM3) or when this process is inhibited (KLHL14 and KCNE3) were identified. Lastly, the network theory was used to obtain a computational model able to recognize putative controller genes involved in the induction and in the prevention of EMT. The results suggested an opposite role of lysophosphatidic acid (LPA) synthesis and degradation enzymes in the regulation of EMT process. In conclusion, these molecules may represent novel EMT regulators and also targets for developing new therapeutic strategies.
Collapse
Affiliation(s)
- V Di Lollo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy. .,Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy.
| | - A Canciello
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy.
| | - M Orsini
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - N Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - M Ancora
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - M Di Federico
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - V Curini
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - M Mattioli
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - C Cammà
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - B Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
163
|
Verma Y, Yadav A, Katara P. Mining of cancer core-genes and their protein interactome using expression profiling based PPI network approach. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2019.100583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
164
|
Liu C, Ma Y, Zhao J, Nussinov R, Zhang YC, Cheng F, Zhang ZK. Computational network biology: Data, models, and applications. PHYSICS REPORTS 2020; 846:1-66. [DOI: 10.1016/j.physrep.2019.12.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
165
|
Mohamed SK, Nounu A, Nováček V. Biological applications of knowledge graph embedding models. Brief Bioinform 2020; 22:1679-1693. [PMID: 32065227 DOI: 10.1093/bib/bbaa012] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 01/04/2023] Open
Abstract
Complex biological systems are traditionally modelled as graphs of interconnected biological entities. These graphs, i.e. biological knowledge graphs, are then processed using graph exploratory approaches to perform different types of analytical and predictive tasks. Despite the high predictive accuracy of these approaches, they have limited scalability due to their dependency on time-consuming path exploratory procedures. In recent years, owing to the rapid advances of computational technologies, new approaches for modelling graphs and mining them with high accuracy and scalability have emerged. These approaches, i.e. knowledge graph embedding (KGE) models, operate by learning low-rank vector representations of graph nodes and edges that preserve the graph's inherent structure. These approaches were used to analyse knowledge graphs from different domains where they showed superior performance and accuracy compared to previous graph exploratory approaches. In this work, we study this class of models in the context of biological knowledge graphs and their different applications. We then show how KGE models can be a natural fit for representing complex biological knowledge modelled as graphs. We also discuss their predictive and analytical capabilities in different biology applications. In this regard, we present two example case studies that demonstrate the capabilities of KGE models: prediction of drug-target interactions and polypharmacy side effects. Finally, we analyse different practical considerations for KGEs, and we discuss possible opportunities and challenges related to adopting them for modelling biological systems.
Collapse
Affiliation(s)
| | - Aayah Nounu
- Insight Centre for Data Analytics, NUI Galway, Galway, Ireland
| | - Vít Nováček
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
166
|
Gupta SK, Srivastava M, Osmanoglu Ö, Dandekar T. Genome-wide inference of the Camponotus floridanus protein-protein interaction network using homologous mapping and interacting domain profile pairs. Sci Rep 2020; 10:2334. [PMID: 32047225 PMCID: PMC7012867 DOI: 10.1038/s41598-020-59344-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Apart from some model organisms, the interactome of most organisms is largely unidentified. High-throughput experimental techniques to determine protein-protein interactions (PPIs) are resource intensive and highly susceptible to noise. Computational methods of PPI determination can accelerate biological discovery by identifying the most promising interacting pairs of proteins and by assessing the reliability of identified PPIs. Here we present a first in-depth study describing a global view of the ant Camponotus floridanus interactome. Although several ant genomes have been sequenced in the last eight years, studies exploring and investigating PPIs in ants are lacking. Our study attempts to fill this gap and the presented interactome will also serve as a template for determining PPIs in other ants in future. Our C. floridanus interactome covers 51,866 non-redundant PPIs among 6,274 proteins, including 20,544 interactions supported by domain-domain interactions (DDIs), 13,640 interactions supported by DDIs and subcellular localization, and 10,834 high confidence interactions mediated by 3,289 proteins. These interactions involve and cover 30.6% of the entire C. floridanus proteome.
Collapse
Affiliation(s)
- Shishir K Gupta
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, D-97074, Würzburg, Germany.,Department of Microbiology, Biocenter, Am Hubland, D-97074, Würzburg, Germany
| | - Mugdha Srivastava
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, D-97074, Würzburg, Germany
| | - Özge Osmanoglu
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, D-97074, Würzburg, Germany
| | - Thomas Dandekar
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, D-97074, Würzburg, Germany. .,EMBL Heidelberg, BioComputing Unit, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| |
Collapse
|
167
|
Singh V, Singh G, Singh V. TulsiPIN: An Interologous Protein Interactome of Ocimum tenuiflorum. J Proteome Res 2020; 19:884-899. [PMID: 31789043 DOI: 10.1021/acs.jproteome.9b00683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Ocimum tenuiflorum, commonly known as holy basil or tulsi, is globally recognized for its multitude of medicinal properties. However, a comprehensive study revealing the complex interplay among its constituent proteins at subcellular level is still lacking. To bridge this gap, in this work, a genome-scale interologous protein-protein interaction (PPI) network, TulsiPIN, is developed using 36 template plants, which consists of 13 660 nodes and 327 409 binary interactions. A high confidence network, hc-TulsiPIN, consisting of 7719 nodes having 95 532 interactions is inferred using domain-domain interaction information along with interolog-based statistics, and its reliability is assessed using pathway enrichment, functional homogeneity, and protein colocalization of PPIs. Examination of topological features revealed that hc-TulsiPIN possesses conventional properties, like small-world, scale-free, and modular architecture. A total of 1625 vital proteins are predicted by statistically evaluating hc-TulsiPIN with two ensembles of corresponding random networks, each consisting of 10 000 realizations of Erdoős-Rényi and Barabási-Albert models. Also, numerous regulatory proteins like transcription factors, transcription regulators, and protein kinases are profiled. Using 36 guide genes participating in 9 secondary metabolite biosynthetic pathways, a subnetwork consisting of 171 proteins and 612 interactions was constructed, and 127 of these proteins could be successfully characterized. Detailed information of TulsiPIN is available at https://cuhpcbbtulsipin.shinyapps.io/tulsipin_v0/ .
Collapse
Affiliation(s)
- Vikram Singh
- Centre for Computational Biology and Bioinformatics , Central University of Himahcal Pradesh , Dharamshala 176206 , India
| | - Gagandeep Singh
- Centre for Computational Biology and Bioinformatics , Central University of Himahcal Pradesh , Dharamshala 176206 , India
| | - Vikram Singh
- Centre for Computational Biology and Bioinformatics , Central University of Himahcal Pradesh , Dharamshala 176206 , India
| |
Collapse
|
168
|
Sabir JSM, El Omri A, Banaganapalli B, Aljuaid N, Omar AMS, Altaf A, Hajrah NH, Zrelli H, Arfaoui L, Elango R, Alharbi MG, Alhebshi AM, Jansen RK, Shaik NA, Khan M. Unraveling the role of salt-sensitivity genes in obesity with integrated network biology and co-expression analysis. PLoS One 2020; 15:e0228400. [PMID: 32027667 PMCID: PMC7004317 DOI: 10.1371/journal.pone.0228400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is a multifactorial disease caused by complex interactions between genes and dietary factors. Salt-rich diet is related to the development and progression of several chronic diseases including obesity. However, the molecular basis of how salt sensitivity genes (SSG) contribute to adiposity in obesity patients remains unexplored. In this study, we used the microarray expression data of visceral adipose tissue samples and constructed a complex protein-interaction network of salt sensitivity genes and their co-expressed genes to trace the molecular pathways connected to obesity. The Salt Sensitivity Protein Interaction Network (SSPIN) of 2691 differentially expressed genes and their 15474 interactions has shown that adipose tissues are enriched with the expression of 23 SSGs, 16 hubs and 84 bottlenecks (p = 2.52 x 10-16) involved in diverse molecular pathways connected to adiposity. Fifteen of these 23 SSGs along with 8 other SSGs showed a co-expression with enriched obesity-related genes (r ≥ 0.8). These SSGs and their co-expression partners are involved in diverse metabolic pathways including adipogenesis, adipocytokine signaling pathway, renin-angiotensin system, etc. This study concludes that SSGs could act as molecular signatures for tracing the basis of adipogenesis among obese patients. Integrated network centered methods may accelerate the identification of new molecular targets from the complex obesity genomics data.
Collapse
Affiliation(s)
- Jamal Sabir M. Sabir
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelfatteh El Omri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nada Aljuaid
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulkader M. Shaikh Omar
- Biology, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulmalik Altaf
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nahid H. Hajrah
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Houda Zrelli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Leila Arfaoui
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona G. Alharbi
- Biology, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alawiah M. Alhebshi
- Biology, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Robert K. Jansen
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, United States of America
| | - Noor A. Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhummadh Khan
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail:
| |
Collapse
|
169
|
Cortés MP, Acuña V, Travisany D, Siegel A, Maass A, Latorre M. Integration of Biological Networks for Acidithiobacillus thiooxidans Describes a Modular Gene Regulatory Organization of Bioleaching Pathways. Front Mol Biosci 2020; 6:155. [PMID: 31998751 PMCID: PMC6966769 DOI: 10.3389/fmolb.2019.00155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/13/2019] [Indexed: 11/13/2022] Open
Abstract
Acidithiobacillus thiooxidans is one of the most studied biomining species, highlighting its ability to oxidize reduced inorganic sulfur compounds, coupled with its elevated capacity to live under an elevated concentration of heavy metals. In this work, using an in silico semi-automatic genome scale approach, two biological networks for A. thiooxidans Licanantay were generated: (i) An affinity transcriptional regulatory network composed of 42 regulatory family genes and 1,501 operons (57% genome coverage) linked through 2,646 putative DNA binding sites (arcs), (ii) A metabolic network reconstruction made of 523 genes and 1,203 reactions (22 pathways related to biomining processes). Through the identification of confident connections between both networks (V-shapes), it was possible to identify a sub-network of transcriptional factor (34 regulators) regulating genes (61 operons) encoding for proteins involved in biomining-related pathways. Network analysis suggested that transcriptional regulation of biomining genes is organized into different modules. The topological parameters showed a high hierarchical organization by levels inside this network (14 layers), highlighting transcription factors CysB, LysR, and IHF as complex modules with high degree and number of controlled pathways. In addition, it was possible to identify transcription factor modules named primary regulators (not controlled by other regulators in the sub-network). Inside this group, CysB was the main module involved in gene regulation of several bioleaching processes. In particular, metabolic processes related to energy metabolism (such as sulfur metabolism) showed a complex integrated regulation, where different primary regulators controlled several genes. In contrast, pathways involved in iron homeostasis and oxidative stress damage are mainly regulated by unique primary regulators, conferring Licanantay an efficient, and specific metal resistance response. This work shows new evidence in terms of transcriptional regulation at a systems level and broadens the study of bioleaching in A. thiooxidans species.
Collapse
Affiliation(s)
- María Paz Cortés
- Center for Mathematical Modeling, Universidad de Chile and UMI CNRS 2807, Santiago, Chile.,Center for Genome Regulation, Universidad de Chile, Santiago, Chile
| | - Vicente Acuña
- Center for Mathematical Modeling, Universidad de Chile and UMI CNRS 2807, Santiago, Chile
| | - Dante Travisany
- Center for Mathematical Modeling, Universidad de Chile and UMI CNRS 2807, Santiago, Chile.,Center for Genome Regulation, Universidad de Chile, Santiago, Chile
| | - Anne Siegel
- IRISA, UMR 6074, CNRS, Rennes, France.,INRIA, Dyliss Team, Centre Rennes-Bretagne-Atlantique, Rennes, France
| | - Alejandro Maass
- Center for Mathematical Modeling, Universidad de Chile and UMI CNRS 2807, Santiago, Chile.,Center for Genome Regulation, Universidad de Chile, Santiago, Chile.,Department of Mathematical Engineering, Universidad de Chile, Santiago, Chile
| | - Mauricio Latorre
- Center for Mathematical Modeling, Universidad de Chile and UMI CNRS 2807, Santiago, Chile.,Center for Genome Regulation, Universidad de Chile, Santiago, Chile.,Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile.,Instituto de Ciencias de la Ingeniería, Universidad de O'Higgins, Rancagua, Chile
| |
Collapse
|
170
|
Ahmed SS, Roy S, Kalita J. Assessing the Effectiveness of Causality Inference Methods for Gene Regulatory Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:56-70. [PMID: 29994618 DOI: 10.1109/tcbb.2018.2853728] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Causality inference is the use of computational techniques to predict possible causal relationships for a set of variables, thereby forming a directed network. Causality inference in Gene Regulatory Networks (GRNs) is an important, yet challenging task due to the limits of available data and lack of efficiency in existing causality inference techniques. A number of techniques have been proposed and applied to infer causal relationships in various domains, although they are not specific to regulatory network inference. In this paper, we assess the effectiveness of methods for inferring causal GRNs. We introduce seven different inference methods and apply them to infer directed edges in GRNs. We use time-series expression data from the DREAM challenges to assess the methods in terms of quality of inference and rank them based on performance. The best method is applied to Breast Cancer data to infer a causal network. Experimental results show that Causation Entropy is best, however, highly time-consuming and not feasible to use in a relatively large network. We infer Breast Cancer GRN with the second-best method, Transfer Entropy. The topological analysis of the network reveals that top out-degree genes such as SLC39A5 which are considered central genes, play important role in cancer progression.
Collapse
|
171
|
Rahbar Saadat Y, Pourseif MM, Zununi Vahed S, Barzegari A, Omidi Y, Barar J. Modulatory Role of Vaginal-Isolated Lactococcus lactis on the Expression of miR-21, miR-200b, and TLR-4 in CAOV-4 Cells and In Silico Revalidation. Probiotics Antimicrob Proteins 2019; 12:1083-1096. [DOI: 10.1007/s12602-019-09596-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
172
|
Sabir JSM, El Omri A, Banaganapalli B, Al-Shaeri MA, Alkenani NA, Sabir MJ, Hajrah NH, Zrelli H, Ciesla L, Nasser KK, Elango R, Shaik NA, Khan M. Dissecting the Role of NF-κb Protein Family and Its Regulators in Rheumatoid Arthritis Using Weighted Gene Co-Expression Network. Front Genet 2019; 10:1163. [PMID: 31824568 PMCID: PMC6879671 DOI: 10.3389/fgene.2019.01163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic synovial autoinflammatory disease that destructs the cartilage and bone, leading to disability. The functional regulation of major immunity-related pathways like nuclear factor kappa B (NF-κB), which is involved in the chronic inflammatory reactions underlying the development of RA, remains to be explored. Therefore, this study has adopted statistical and knowledge-based systemic investigations (like gene correlation, semantic similarity, and topological parameters based on graph theory) to study the gene expression status of NF-κB protein family (NKPF) and its regulators in synovial tissues to trace the molecular pathways through which these regulators contribute to RA. A complex protein–protein interaction map (PPIM) of 2,742 genes and 37,032 interactions was constructed from differentially expressed genes (p ≤ 0.05). PPIM was further decomposed into a Regulator Allied Protein Interaction Network (RAPIN) based on the interaction between genes (5 NKPF, 31 seeds, 131 hubs, and 652 bottlenecks). Pathway network analysis has shown the RA-specific disturbances in the functional connectivity between seed genes (RIPK1, ATG7, TLR4, TNFRSF1A, KPNA1, CFLAR, SNW1, FOSB, PARVA, CX3CL1, and TRPC6) and NKPF members (RELA, RELB, NFKB2, and REL). Interestingly, these genes are known for their involvement in inflammation and immune system (signaling by interleukins, cytokine signaling in immune system, NOD-like receptor signaling, MAPK signaling, Toll-like receptor signaling, and TNF signaling) pathways connected to RA. This study, for the first time, reports that SNW1, along with other NK regulatory genes, plays an important role in RA pathogenesis and might act as potential biomarker for RA. Additionally, these genes might play important roles in RA pathogenesis, as well as facilitate the development of effective targeted therapies. Our integrative data analysis and network-based methods could accelerate the identification of novel drug targets for RA from high-throughput genomic data.
Collapse
Affiliation(s)
- Jamal S M Sabir
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelfatteh El Omri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majed A Al-Shaeri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Naser A Alkenani
- Biology-Zoology Division, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mumdooh J Sabir
- Department of Computer Sciences, Faculty of Computers and Information Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nahid H Hajrah
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Houda Zrelli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Lukasz Ciesla
- Department of Biological Sciences, Science and Engineering Complex, The University of Alabama, Tuscaloosa, AL, United States
| | - Khalidah K Nasser
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor Ahmad Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhummadh Khan
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
173
|
Fraunberger E, Esser MJ. Neuro-Inflammation in Pediatric Traumatic Brain Injury-from Mechanisms to Inflammatory Networks. Brain Sci 2019; 9:E319. [PMID: 31717597 PMCID: PMC6895990 DOI: 10.3390/brainsci9110319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Compared to traumatic brain injury (TBI) in the adult population, pediatric TBI has received less research attention, despite its potential long-term impact on the lives of many children around the world. After numerous clinical trials and preclinical research studies examining various secondary mechanisms of injury, no definitive treatment has been found for pediatric TBIs of any severity. With the advent of high-throughput and high-resolution molecular biology and imaging techniques, inflammation has become an appealing target, due to its mixed effects on outcome, depending on the time point examined. In this review, we outline key mechanisms of inflammation, the contribution and interactions of the peripheral and CNS-based immune cells, and highlight knowledge gaps pertaining to inflammation in pediatric TBI. We also introduce the application of network analysis to leverage growing multivariate and non-linear inflammation data sets with the goal to gain a more comprehensive view of inflammation and develop prognostic and treatment tools in pediatric TBI.
Collapse
Affiliation(s)
- Erik Fraunberger
- Alberta Children’s Hospital Research Institute, Calgary, AB T3B 6A8, Canada;
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Michael J. Esser
- Alberta Children’s Hospital Research Institute, Calgary, AB T3B 6A8, Canada;
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Pediatrics, Cumming School Of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
174
|
Bordeu I, Amarteifio S, Garcia-Millan R, Walter B, Wei N, Pruessner G. Volume explored by a branching random walk on general graphs. Sci Rep 2019; 9:15590. [PMID: 31666539 PMCID: PMC6821755 DOI: 10.1038/s41598-019-51225-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/25/2019] [Indexed: 11/09/2022] Open
Abstract
Branching processes are used to model diverse social and physical scenarios, from extinction of family names to nuclear fission. However, for a better description of natural phenomena, such as viral epidemics in cellular tissues, animal populations and social networks, a spatial embedding-the branching random walk (BRW)-is required. Despite its wide range of applications, the properties of the volume explored by the BRW so far remained elusive, with exact results limited to one dimension. Here we present analytical results, supported by numerical simulations, on the scaling of the volume explored by a BRW in the critical regime, the onset of epidemics, in general environments. Our results characterise the spreading dynamics on regular lattices and general graphs, such as fractals, random trees and scale-free networks, revealing the direct relation between the graphs' dimensionality and the rate of propagation of the viral process. Furthermore, we use the BRW to determine the spectral properties of real social and metabolic networks, where we observe that a lack of information of the network structure can lead to differences in the observed behaviour of the spreading process. Our results provide observables of broad interest for the characterisation of real world lattices, tissues, and networks.
Collapse
Affiliation(s)
- Ignacio Bordeu
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK. .,Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK. .,DAMTP, Centre for Mathematical Sciences, University of Cambridge, Cambridge, CB3 0WA, UK. .,The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK.
| | - Saoirse Amarteifio
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.,Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Rosalba Garcia-Millan
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.,Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Benjamin Walter
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.,Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Nanxin Wei
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.,Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Gunnar Pruessner
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK. .,Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
175
|
He X, Xu H, Zhao W, Zhan M, Li Y, Liu H, Tan L, Lu L. POPDC3 is a potential biomarker for prognosis and radioresistance in patients with head and neck squamous cell carcinoma. Oncol Lett 2019; 18:5468-5480. [PMID: 31612055 PMCID: PMC6781657 DOI: 10.3892/ol.2019.10888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 08/20/2019] [Indexed: 12/01/2022] Open
Abstract
Radiotherapy is the primary means of treatment for patients with head and neck squamous cell carcinoma (HNSCC); however, radioresistance-induced recurrence is the primary cause of HNSCC treatment failure. Therefore, identifying specific predictive biomarkers of the response to radiotherapy may improve prognosis. In the present study, to identify the potential candidate genes associated with radioresistance in patients with HNSCC, the microarray datasets GSE9716, GSE61772 and GSE20549 were downloaded from the Gene Expression Omnibus database. The original CEL files were preprocessed using the Affymetrix package and quantile normalization and background correction were conducted using the Core package in Bioconductor. The GSE9716 dataset, consisting of 18 irradiated and 16 non-irradiated samples, was divided into two groups according to their exposure to irradiation: i) Non-irradiation group, which included 8 radioresistant samples and 8 radiosensitive samples; and ii) post-irradiation group, which included 9 radioresistant samples and 9 radiosensitive samples. The two groups were treated as separate datasets and screened. A total of 86 differentially expressed genes (DEGs) were identified in the non-irradiation group and 405 DEGs in the post-irradiation group. Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analysis detected several significant functions associated with the DEGs. In the co-expression analysis, 76 hub genes in the light green module and 917 hub genes with a high connectivity were selected for further analysis. Finally, overlapping the DEGs and hub genes from the two groups yielded a map of 13 shared differentially expressed genes. The 13 genes showed significantly different expression in radioresistant samples compared with the radiosensitive samples before and after irradiation. Out of these genes, popeye domain-containing protein 3 (POPDC3) was highly expressed in the post-irradiation group compared with the non-irradiation group. In survival analysis, high POPDC3 expression correlated with poor a prognosis for patients with HNSCC. The independent prognostic factors were identified using univariate and multivariate Cox analyses based on The Cancer Genome Atlas database. These were incorporated into a nomogram to predict 3- and 5-year overall survival. Receiver operating characteristic curves were used to estimate the accuracy of the nomogram. Together these studies suggest that POPDC3 may serve as a potential predictive biomarker for prognosis and radioresistance of patients with HNSCC as well as clinical diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Xu He
- Department of Interventional Oncology, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China.,Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hongfa Xu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Wei Zhao
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Meixiao Zhan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Yong Li
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hongyi Liu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Li Tan
- Center of Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Ligong Lu
- Department of Interventional Oncology, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China.,Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
176
|
Selvarajoo K. Large‐scale‐free network organisation is likely key for biofilm phase transition. ENGINEERING BIOLOGY 2019. [DOI: 10.1049/enb.2019.0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Kumar Selvarajoo
- Computational and Systems Biology, Biotransformation Innovation Platform (BioTrans), Agency for Science Technology & Research (A*STAR) 61 Biopolis Drive Proteos 13873 Singapore
| |
Collapse
|
177
|
Enciso J, Pelayo R, Villarreal C. From Discrete to Continuous Modeling of Lymphocyte Development and Plasticity in Chronic Diseases. Front Immunol 2019; 10:1927. [PMID: 31481957 PMCID: PMC6710364 DOI: 10.3389/fimmu.2019.01927] [Citation(s) in RCA: 6] [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: 12/12/2018] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
The molecular events leading to differentiation, development, and plasticity of lymphoid cells have been subject of intense research due to their key roles in multiple pathologies, such as lymphoproliferative disorders, tumor growth maintenance and chronic diseases. The emergent roles of lymphoid cells and the use of high-throughput technologies have led to an extensive accumulation of experimental data allowing the reconstruction of gene regulatory networks (GRN) by integrating biochemical signals provided by the microenvironment with transcriptional modules of lineage-specific genes. Computational modeling of GRN has been useful for the identification of molecular switches involved in lymphoid specification, prediction of microenvironment-dependent cell plasticity, and analyses of signaling events occurring downstream the activation of antigen recognition receptors. Among most common modeling strategies to analyze the dynamical behavior of GRN, discrete dynamic models are widely used for their capacity to capture molecular interactions when a limited knowledge of kinetic parameters is present. However, they are less powerful when modeling complex systems sensitive to biochemical gradients. To compensate it, discrete models may be transformed into regulatory networks that includes state variables and parameters varying within a continuous range. This approach is based on a system of differential equations dynamics with regulatory interactions described by fuzzy logic propositions. Here, we discuss the applicability of this method on modeling of development and plasticity processes of adaptive lymphocytes, and its potential implications in the study of pathological landscapes associated to chronic diseases.
Collapse
Affiliation(s)
- Jennifer Enciso
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Carlos Villarreal
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Física Cuántica y Fotónica, Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
178
|
Velázquez-Lizárraga AE, Juárez-Morales JL, Racotta IS, Villarreal-Colmenares H, Valdes-Lopez O, Luna-González A, Rodríguez-Jaramillo C, Estrada N, Ascencio F. Transcriptomic analysis of Pacific white shrimp (Litopenaeus vannamei, Boone 1931) in response to acute hepatopancreatic necrosis disease caused by Vibrio parahaemolyticus. PLoS One 2019; 14:e0220993. [PMID: 31408485 PMCID: PMC6692014 DOI: 10.1371/journal.pone.0220993] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022] Open
Abstract
Acute hepatopancreatic necrosis disease (AHPND), caused by marine bacteria Vibrio Parahaemolyticus, is a huge problem in shrimp farms. The V. parahaemolyticus infecting material is contained in a plasmid which encodes for the lethal toxins PirABVp, whose primary target tissue is the hepatopancreas, causing sloughing of epithelial cells, necrosis, and massive hemocyte infiltration. To get a better understanding of the hepatopancreas response during AHPND, juvenile shrimp Litopenaeus vannamei were infected by immersion with V. parahaemolyticus. We performed transcriptomic mRNA sequencing of infected shrimp hepatopancreas, at 24 hours post-infection, to identify novel differentially expressed genes a total of 174,098 transcripts were examined of which 915 transcripts were found differentially expressed after comparative transcriptomic analysis: 442 up-regulated and 473 down-regulated transcripts. Gene Ontology term enrichment analysis for up-regulated transcripts includes metabolic process, regulation of programmed cell death, carbohydrate metabolic process, and biological adhesion, whereas for down-regulated transcripts include, microtubule-based process, cell activation, and chitin metabolic process. The analysis of protein- protein network between up and down-regulated genes indicates that the first gene interactions are connected to oxidation-processes and sarcomere organization. Additionally, protein-protein networks analysis identified 20-top highly connected hub nodes. Based on their immunological or metabolic function, ten candidate transcripts were selected to measure their mRNA relative expression levels in AHPND infected shrimp hepatopancreas by RT-qPCR. Our results indicate a close connection between the immune and metabolism systems during AHPND infection. Our RNA-Seq and RT-qPCR data provide the possible immunological and physiological scenario as well as the molecular pathways that take place in the shrimp hepatopancreas in response to an infectious disease.
Collapse
Affiliation(s)
- Adrián E. Velázquez-Lizárraga
- Laboratorio de Patogénesis Microbiana, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| | - José Luis Juárez-Morales
- Programa de Cátedras CONACyT, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| | - Ilie S. Racotta
- Laboratorio de Metabolismo Energético, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| | - Humberto Villarreal-Colmenares
- Parque de Innovación Tecnológica, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| | - Oswaldo Valdes-Lopez
- Departamento de Bioquímica, Facultad de Estudios Superiores – Universidad Autónoma de México, Tlalnepantla de Baz, Estado de México, México
| | - Antonio Luna-González
- Departamento de Acuacultura. Instituto Politécnico Nacional-Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional, Unidad Sinaloa (IPN-CIIDIR Sinaloa), Guasave, Sinaloa, México
| | - Carmen Rodríguez-Jaramillo
- Laboratorio de Histología, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| | - Norma Estrada
- Programa de Cátedras CONACyT, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| | - Felipe Ascencio
- Laboratorio de Patogénesis Microbiana, Centro de Investigaciones Biológicas del Noroeste, S. C. (CIBNOR), La Paz, Baja California Sur, México
| |
Collapse
|
179
|
Ito EA, Katahira I, Vicente FFDR, Pereira LFP, Lopes FM. BASiNET-BiologicAl Sequences NETwork: a case study on coding and non-coding RNAs identification. Nucleic Acids Res 2019; 46:e96. [PMID: 29873784 PMCID: PMC6144827 DOI: 10.1093/nar/gky462] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/22/2018] [Indexed: 01/23/2023] Open
Abstract
With the emergence of Next Generation Sequencing (NGS) technologies, a large volume of sequence data in particular de novo sequencing was rapidly produced at relatively low costs. In this context, computational tools are increasingly important to assist in the identification of relevant information to understand the functioning of organisms. This work introduces BASiNET, an alignment-free tool for classifying biological sequences based on the feature extraction from complex network measurements. The method initially transform the sequences and represents them as complex networks. Then it extracts topological measures and constructs a feature vector that is used to classify the sequences. The method was evaluated in the classification of coding and non-coding RNAs of 13 species and compared to the CNCI, PLEK and CPC2 methods. BASiNET outperformed all compared methods in all adopted organisms and datasets. BASiNET have classified sequences in all organisms with high accuracy and low standard deviation, showing that the method is robust and non-biased by the organism. The proposed methodology is implemented in open source in R language and freely available for download at https://cran.r-project.org/package=BASiNET.
Collapse
Affiliation(s)
- Eric Augusto Ito
- Department of Computer Science, Bioinformatics Graduate Program, Federal University of Technology - Paraná, Cornélio Procópio, PR 86300-000, Brazil
| | - Isaque Katahira
- Department of Computer Science, Bioinformatics Graduate Program, Federal University of Technology - Paraná, Cornélio Procópio, PR 86300-000, Brazil
| | - Fábio Fernandes da Rocha Vicente
- Department of Computer Science, Bioinformatics Graduate Program, Federal University of Technology - Paraná, Cornélio Procópio, PR 86300-000, Brazil
| | - Luiz Filipe Protasio Pereira
- Department of Computer Science, Bioinformatics Graduate Program, Federal University of Technology - Paraná, Cornélio Procópio, PR 86300-000, Brazil.,Empresa Brasileira de Pesquisa Agropecuária, Embrapa Café, Brasília, DF 70770-901, Brazil
| | - Fabrício Martins Lopes
- Department of Computer Science, Bioinformatics Graduate Program, Federal University of Technology - Paraná, Cornélio Procópio, PR 86300-000, Brazil
| |
Collapse
|
180
|
Sosa EJ, Burguener G, Lanzarotti E, Defelipe L, Radusky L, Pardo AM, Marti M, Turjanski AG, Fernández Do Porto D. Target-Pathogen: a structural bioinformatic approach to prioritize drug targets in pathogens. Nucleic Acids Res 2019; 46:D413-D418. [PMID: 29106651 PMCID: PMC5753371 DOI: 10.1093/nar/gkx1015] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/16/2017] [Indexed: 12/20/2022] Open
Abstract
Available genomic data for pathogens has created new opportunities for drug discovery and development to fight them, including new resistant and multiresistant strains. In particular structural data must be integrated with both, gene information and experimental results. In this sense, there is a lack of an online resource that allows genome wide-based data consolidation from diverse sources together with thorough bioinformatic analysis that allows easy filtering and scoring for fast target selection for drug discovery. Here, we present Target-Pathogen database (http://target.sbg.qb.fcen.uba.ar/patho), designed and developed as an online resource that allows the integration and weighting of protein information such as: function, metabolic role, off-targeting, structural properties including druggability, essentiality and omic experiments, to facilitate the identification and prioritization of candidate drug targets in pathogens. We include in the database 10 genomes of some of the most relevant microorganisms for human health (Mycobacterium tuberculosis, Mycobacterium leprae, Klebsiella pneumoniae, Plasmodium vivax, Toxoplasma gondii, Leishmania major, Wolbachia bancrofti, Trypanosoma brucei, Shigella dysenteriae and Schistosoma Smanosoni) and show its applicability. New genomes can be uploaded upon request.
Collapse
Affiliation(s)
- Ezequiel J Sosa
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Germán Burguener
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Esteban Lanzarotti
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Lucas Defelipe
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Leandro Radusky
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Agustín M Pardo
- Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Marcelo Marti
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Adrián G Turjanski
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| | - Darío Fernández Do Porto
- IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA Ciudad de Buenos Aires, Argentina
| |
Collapse
|
181
|
Gene Modules Co-regulated with Biosynthetic Gene Clusters for Allelopathy between Rice and Barnyardgrass. Int J Mol Sci 2019; 20:ijms20163846. [PMID: 31394718 PMCID: PMC6719971 DOI: 10.3390/ijms20163846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
Allelopathy is a central process in crop–weed interactions and is mediated by the release of allelochemicals that result in adverse growth effects on one or the other plant in the interaction. The genomic mechanism for the biosynthesis of many critical allelochemicals is unknown but may involve the clustering of non-homologous biosynthetic genes involved in their formation and regulatory gene modules involved in controlling the coordinated expression within these gene clusters. In this study, we used the transcriptomes from mono- or co-cultured rice and barnyardgrass to investigate the nature of the gene clusters and their regulatory gene modules involved in the allelopathic interactions of these two plants. In addition to the already known biosynthetic gene clusters in barnyardgrass we identified three potential new clusters including one for quercetin biosynthesis and potentially involved in allelopathic interaction with rice. Based on the construction of gene networks, we identified one gene regulatory module containing hub transcription factors, significantly positively co-regulated with both the momilactone A and phytocassane clusters in rice. In barnyardgrass, gene modules and hub genes co-expressed with the gene clusters responsible for 2,4-dihydroxy-7-methoxy-1,4-benzoxazin-3-one (DIMBOA) biosynthesis were also identified. In addition, we found three genes in barnyardgrass encoding indole-3-glycerolphosphate synthase that regulate the expression of the DIMBOA cluster. Our findings offer new insights into the regulatory mechanisms of biosynthetic gene clusters involved in allelopathic interactions between rice and barnyardgrass, and have potential implications in controlling weeds for crop protection.
Collapse
|
182
|
Generalised thresholding of hidden variable network models with scale-free property. Sci Rep 2019; 9:11273. [PMID: 31375716 PMCID: PMC6677767 DOI: 10.1038/s41598-019-47628-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/19/2019] [Indexed: 11/09/2022] Open
Abstract
The hidden variable formalism (based on the assumption of some intrinsic node parameters) turned out to be a remarkably efficient and powerful approach in describing and analyzing the topology of complex networks. Owing to one of its most advantageous property - namely proven to be able to reproduce a wide range of different degree distribution forms - it has become a standard tool for generating networks having the scale-free property. One of the most intensively studied version of this model is based on a thresholding mechanism of the exponentially distributed hidden variables associated to the nodes (intrinsic vertex weights), which give rise to the emergence of a scale-free network where the degree distribution p(k) ~ k−γ is decaying with an exponent of γ = 2. Here we propose a generalization and modification of this model by extending the set of connection probabilities and hidden variable distributions that lead to the aforementioned degree distribution, and analyze the conditions leading to the above behavior analytically. In addition, we propose a relaxation of the hard threshold in the connection probabilities, which opens up the possibility for obtaining sparse scale free networks with arbitrary scaling exponent.
Collapse
|
183
|
Li X, Wang H, Tong W, Feng L, Wang L, Rahman SU, Wei G, Tao S. Exploring the evolutionary dynamics of Rhizobium plasmids through bipartite network analysis. Environ Microbiol 2019; 22:934-951. [PMID: 31361937 DOI: 10.1111/1462-2920.14762] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/24/2019] [Accepted: 07/25/2019] [Indexed: 10/26/2022]
Abstract
The genus Rhizobium usually has a multipartite genome architecture with a chromosome and several plasmids, making these bacteria a perfect candidate for plasmid biology studies. As there are no universally shared genes among typical plasmids, network analyses can complement traditional phylogenetics in a broad-scale study of plasmid evolution. Here, we present an exhaustive analysis of 216 plasmids from 49 complete genomes of Rhizobium by constructing a bipartite network that consists of two classes of nodes, the plasmids and homologous protein families that connect them. Dissection of the network using a hierarchical clustering strategy reveals extensive variety, with 34 homologous plasmid clusters. Four large clusters including one cluster of symbiotic plasmids and two clusters of chromids carrying some truly essential genes are widely distributed among Rhizobium. In contrast, the other clusters are quite small and rare. Symbiotic clusters and rare accessory clusters are exogenetic and do not appear to have co-evolved with the common accessory clusters; the latter ones have a large coding potential and functional complementarity for different lifestyles in Rhizobium. The bipartite network also provides preliminary evidence of Rhizobium plasmid variation and formation including genetic exchange, plasmid fusion and fission, exogenetic plasmid transfer, host plant selection, and environmental adaptation.
Collapse
Affiliation(s)
- Xiangchen Li
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hao Wang
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Wenjun Tong
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Li Feng
- College of Enology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lina Wang
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Siddiq Ur Rahman
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak, Khyber Pakhtunkhwa, 27200, Pakistan
| | - Gehong Wei
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shiheng Tao
- State Key Laboratory of Crop Stress Biology in Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
184
|
Farquhar KS, Charlebois DA, Szenk M, Cohen J, Nevozhay D, Balázsi G. Role of network-mediated stochasticity in mammalian drug resistance. Nat Commun 2019; 10:2766. [PMID: 31235692 PMCID: PMC6591227 DOI: 10.1038/s41467-019-10330-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 05/03/2019] [Indexed: 11/11/2022] Open
Abstract
A major challenge in biology is that genetically identical cells in the same environment can display gene expression stochasticity (noise), which contributes to bet-hedging, drug tolerance, and cell-fate switching. The magnitude and timescales of stochastic fluctuations can depend on the gene regulatory network. Currently, it is unclear how gene expression noise of specific networks impacts the evolution of drug resistance in mammalian cells. Answering this question requires adjusting network noise independently from mean expression. Here, we develop positive and negative feedback-based synthetic gene circuits to decouple noise from the mean for Puromycin resistance gene expression in Chinese Hamster Ovary cells. In low Puromycin concentrations, the high-noise, positive-feedback network delays long-term adaptation, whereas it facilitates adaptation under high Puromycin concentration. Accordingly, the low-noise, negative-feedback circuit can maintain resistance by acquiring mutations while the positive-feedback circuit remains mutation-free and regains drug sensitivity. These findings may have profound implications for chemotherapeutic inefficiency and cancer relapse. The role of gene expression noise in the evolution of drug resistance in mammalian cells is unclear. Here, by uncoupling noise from mean expression of a drug resistance gene in CHO cells the authors show that noisy expression aids adaptation to high drug levels, but delays it at low drug levels.
Collapse
Affiliation(s)
- Kevin S Farquhar
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center, UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Daniel A Charlebois
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Physics, University of Alberta, Edmonton, AB, 4-181 CCIS, T6G-2E1, Canada
| | - Mariola Szenk
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Joseph Cohen
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Dmitry Nevozhay
- School of Biomedicine, Far Eastern Federal University, 8 Sukhanova Street, Vladivostok, 690950, Russia.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gábor Balázsi
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA. .,Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center, UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA. .,Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA. .,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
185
|
Kizhakkethil Youseph AS, Chetty M, Karmakar G. Reverse engineering genetic networks using nonlinear saturation kinetics. Biosystems 2019; 182:30-41. [PMID: 31185246 DOI: 10.1016/j.biosystems.2019.103977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/25/2019] [Accepted: 05/27/2019] [Indexed: 01/01/2023]
Abstract
A gene regulatory network (GRN) represents a set of genes along with their regulatory interactions. Cellular behavior is driven by genetic level interactions. Dynamics of such systems show nonlinear saturation kinetics which can be best modeled by Michaelis-Menten (MM) and Hill equations. Although MM equation is being widely used for modeling biochemical processes, it has been applied rarely for reverse engineering GRNs. In this paper, we develop a complete framework for a novel model for GRN inference using MM kinetics. A set of coupled equations is first proposed for modeling GRNs. In the coupled model, Michaelis-Menten constant associated with regulation by a gene is made invariant irrespective of the gene being regulated. The parameter estimation of the proposed model is carried out using an evolutionary optimization method, namely, trigonometric differential evolution (TDE). Subsequently, the model is further improved and the regulations of different genes by a given gene are made distinct by allowing varying values of Michaelis-Menten constants for each regulation. Apart from making the model more relevant biologically, the improvement results in a decoupled GRN model with fast estimation of model parameters. Further, to enhance exploitation of the search, we propose a local search algorithm based on hill climbing heuristics. A novel mutation operation is also proposed to avoid population stagnation and premature convergence. Real life benchmark data sets generated in vivo are used for validating the proposed model. Further, we also analyze realistic in silico datasets generated using GeneNetweaver. The comparison of the performance of proposed model with other existing methods shows the potential of the proposed model.
Collapse
Affiliation(s)
| | - Madhu Chetty
- School of Science, Engineering and Information Technology, Federation University Australia, Gippsland 3842, Australia
| | - Gour Karmakar
- School of Science, Engineering and Information Technology, Federation University Australia, Gippsland 3842, Australia
| |
Collapse
|
186
|
Iacono G, Massoni-Badosa R, Heyn H. Single-cell transcriptomics unveils gene regulatory network plasticity. Genome Biol 2019; 20:110. [PMID: 31159854 PMCID: PMC6547541 DOI: 10.1186/s13059-019-1713-4] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Single-cell RNA sequencing (scRNA-seq) plays a pivotal role in our understanding of cellular heterogeneity. Current analytical workflows are driven by categorizing principles that consider cells as individual entities and classify them into complex taxonomies. RESULTS We devise a conceptually different computational framework based on a holistic view, where single-cell datasets are used to infer global, large-scale regulatory networks. We develop correlation metrics that are specifically tailored to single-cell data, and then generate, validate, and interpret single-cell-derived regulatory networks from organs and perturbed systems, such as diabetes and Alzheimer's disease. Using tools from graph theory, we compute an unbiased quantification of a gene's biological relevance and accurately pinpoint key players in organ function and drivers of diseases. CONCLUSIONS Our approach detects multiple latent regulatory changes that are invisible to single-cell workflows based on clustering or differential expression analysis, significantly broadening the biological insights that can be obtained with this leading technology.
Collapse
Affiliation(s)
- Giovanni Iacono
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, 08028, Barcelona, Spain.
| | - Ramon Massoni-Badosa
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, 08028, Barcelona, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, 08028, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
187
|
Dozmorov MG. Disease classification: from phenotypic similarity to integrative genomics and beyond. Brief Bioinform 2019; 20:1769-1780. [DOI: 10.1093/bib/bby049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
Abstract
A fundamental challenge of modern biomedical research is understanding how diseases that are similar on the phenotypic level are similar on the molecular level. Integration of various genomic data sets with the traditionally used phenotypic disease similarity revealed novel genetic and molecular mechanisms and blurred the distinction between monogenic (Mendelian) and complex diseases. Network-based medicine has emerged as a complementary approach for identifying disease-causing genes, genetic mediators, disruptions in the underlying cellular functions and for drug repositioning. The recent development of machine and deep learning methods allow for leveraging real-life information about diseases to refine genetic and phenotypic disease relationships. This review describes the historical development and recent methodological advancements for studying disease classification (nosology).
Collapse
Affiliation(s)
- Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, 830 East Main Street, Richmond, VA, USA
| |
Collapse
|
188
|
Thiel D, Conrad ND, Ntini E, Peschutter RX, Siebert H, Marsico A. Identifying lncRNA-mediated regulatory modules via ChIA-PET network analysis. BMC Bioinformatics 2019; 20:292. [PMID: 31142264 PMCID: PMC6540383 DOI: 10.1186/s12859-019-2900-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
Background Although several studies have provided insights into the role of long non-coding RNAs (lncRNAs), the majority of them have unknown function. Recent evidence has shown the importance of both lncRNAs and chromatin interactions in transcriptional regulation. Although network-based methods, mainly exploiting gene-lncRNA co-expression, have been applied to characterize lncRNA of unknown function by means of ’guilt-by-association’, no strategy exists so far which identifies mRNA-lncRNA functional modules based on the 3D chromatin interaction graph. Results To better understand the function of chromatin interactions in the context of lncRNA-mediated gene regulation, we have developed a multi-step graph analysis approach to examine the RNA polymerase II ChIA-PET chromatin interaction network in the K562 human cell line. We have annotated the network with gene and lncRNA coordinates, and chromatin states from the ENCODE project. We used centrality measures, as well as an adaptation of our previously developed Markov State Models (MSM) clustering method, to gain a better understanding of lncRNAs in transcriptional regulation. The novelty of our approach resides in the detection of fuzzy regulatory modules based on network properties and their optimization based on co-expression analysis between genes and gene-lncRNA pairs. This results in our method returning more bona fide regulatory modules than other state-of-the art approaches for clustering on graphs. Conclusions Interestingly, we find that lncRNA network hubs tend to be significantly enriched in evolutionary conserved lncRNAs and enhancer-like functions. We validated regulatory functions for well known lncRNAs, such as MALAT1 and the enhancer-like lncRNA FALEC. In addition, by investigating the modular structure of bigger components we mine putative regulatory functions for uncharacterized lncRNAs. Electronic supplementary material The online version of this article (10.1186/s12859-019-2900-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Denise Thiel
- Max Planck Institute for Molecular Genetics, Berlin, Ihnestraße 63-73, Berlin, 14195, Germany
| | | | - Evgenia Ntini
- Max Planck Institute for Molecular Genetics, Berlin, Ihnestraße 63-73, Berlin, 14195, Germany.,Department of Mathematics and Informatics, Freie Universität, Berlin, Arnimallee 7, Berlin, 14195, Germany
| | - Ria X Peschutter
- Max Planck Institute for Molecular Genetics, Berlin, Ihnestraße 63-73, Berlin, 14195, Germany
| | - Heike Siebert
- Department of Mathematics and Informatics, Freie Universität, Berlin, Arnimallee 7, Berlin, 14195, Germany
| | - Annalisa Marsico
- Max Planck Institute for Molecular Genetics, Berlin, Ihnestraße 63-73, Berlin, 14195, Germany. .,Department of Mathematics and Informatics, Freie Universität, Berlin, Arnimallee 7, Berlin, 14195, Germany. .,Institute of Computational Biology (ICB), Helmholtz Zentrum München, Ingolstädter Landstraße 1, Oberschleißheim, 85764, Germany.
| |
Collapse
|
189
|
Omony J, de Jong A, Kok J, van Hijum SAFT. Reconstruction and inference of the Lactococcus lactis MG1363 gene co-expression network. PLoS One 2019; 14:e0214868. [PMID: 31116749 PMCID: PMC6530827 DOI: 10.1371/journal.pone.0214868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/21/2019] [Indexed: 01/30/2023] Open
Abstract
Lactic acid bacteria are Gram-positive bacteria used throughout the world in many industrial applications for their acidification, flavor and texture formation attributes. One of the species, Lactococcus lactis, is employed for the production of fermented milk products like cheese, buttermilk and quark. It ferments lactose to lactic acid and, thus, helps improve the shelf life of the products. Many physiological and transcriptome studies have been performed in L. lactis in order to comprehend and improve its biotechnological assets. Using large amounts of transcriptome data to understand and predict the behavior of biological processes in bacterial or other cell types is a complex task. Gene networks enable predicting gene behavior and function in the context of transcriptionally linked processes. We reconstruct and present the gene co-expression network (GCN) for the most widely studied L. lactis strain, MG1363, using publicly available transcriptome data. Several methods exist to generate and judge the quality of GCNs. Different reconstruction methods lead to networks with varying structural properties, consequently altering gene clusters. We compared the structural properties of the MG1363 GCNs generated by five methods, namely Pearson correlation, Spearman correlation, GeneNet, Weighted Gene Co-expression Network Analysis (WGCNA), and Sparse PArtial Correlation Estimation (SPACE). Using SPACE, we generated an L. lactis MG1363 GCN and assessed its quality using modularity and structural and biological criteria. The L. lactis MG1363 GCN has structural properties similar to those of the gold-standard networks of Escherichia coli K-12 and Bacillus subtilis 168. We showcase that the network can be used to mine for genes with similar expression profiles that are also generally linked to the same biological process.
Collapse
Affiliation(s)
- Jimmy Omony
- Top Institute Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Anne de Jong
- Top Institute Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Jan Kok
- Top Institute Food and Nutrition (TIFN), Wageningen, The Netherlands
- * E-mail:
| | | |
Collapse
|
190
|
Chen J, Qian X, He Y, Han X, Pan Y. Novel key genes in triple‐negative breast cancer identified by weighted gene co‐expression network analysis. J Cell Biochem 2019; 120:16900-16912. [PMID: 31081967 DOI: 10.1002/jcb.28948] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Jian Chen
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Xiaojun Qian
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Yifu He
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Xinghua Han
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Yueyin Pan
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| |
Collapse
|
191
|
Latorre M, Burkhead JL, Hodar C, Arredondo M, González M, Araya M. Chronic copper treatment prevents the liver critical balance transcription response induced by acetaminophen. J Trace Elem Med Biol 2019; 53:113-119. [PMID: 30910193 DOI: 10.1016/j.jtemb.2019.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/31/2019] [Accepted: 02/17/2019] [Indexed: 02/01/2023]
Abstract
The independent toxic effects of copper and acetaminophen are among the most studied topics in liver toxicity. Here, in an animal model of Cebus capucinus chronically exposed to high dietary copper, we assessed clinical and global transcriptional adaptations of the liver induced by a single high dose of acetaminophen. The experiment conditions were chosen to resemble a close to human real-life situation of exposure to both toxic stimuli. The clinical parameters and histological analyses indicated that chronic copper administration does not induce liver damage and may have a protective effect in acetaminophen challenge. Acetaminophen administration in previously non-exposed animals induced down-regulation of a complex network of gene regulators, highlighting the putative participation of the families of gene regulators HNF, FOX, PPAR and NRF controlling this process. This gene response was not observed in animals that previously received chronic oral copper, suggesting that this metal induces a transcriptional adaptation that may protect against acetaminophen toxicity, a classical adaptation response termed preconditioning of the liver.
Collapse
Affiliation(s)
- Mauricio Latorre
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, El Líbano 5524, Macul, Santiago, Chile; Center for Genome Regulation (Fondap 15090007), Universidad de Chile, Blanco Encalada 2085, Santiago, Chile; Mathomics, Center for Mathematical Modeling, Universidad de Chile, Beauchef 851, 7th Floor, Santiago, Chile; Instituto de Ciencias de la Ingeniería, Universidad de O'Higgins, Av. Viel 1497, Rancagua, Chile.
| | - Jason L Burkhead
- Department of Biological Sciences Anchorage, University of Alaska Anchorage, Anchorage, Alaska, United States
| | - Christian Hodar
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, El Líbano 5524, Macul, Santiago, Chile; Center for Genome Regulation (Fondap 15090007), Universidad de Chile, Blanco Encalada 2085, Santiago, Chile
| | - Miguel Arredondo
- Micronutrients Laboratory, INTA, Universidad de Chile, El Líbano 5524, Macul, Santiago, Chile
| | - Mauricio González
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, El Líbano 5524, Macul, Santiago, Chile; Center for Genome Regulation (Fondap 15090007), Universidad de Chile, Blanco Encalada 2085, Santiago, Chile
| | - Magdalena Araya
- Gastroenterología y Nutrición, INTA, Universidad de Chile, El Líbano 5524, Macul, Santiago, Chile.
| |
Collapse
|
192
|
Orellana E, Davies-Sala C, Guerrero LD, Vardé I, Altina M, Lorenzo MC, Figuerola EL, Pontiggia RM, Erijman L. Microbiome network analysis of co-occurrence patterns in anaerobic co-digestion of sewage sludge and food waste. WATER SCIENCE AND TECHNOLOGY : A JOURNAL OF THE INTERNATIONAL ASSOCIATION ON WATER POLLUTION RESEARCH 2019; 79:1956-1965. [PMID: 31294712 DOI: 10.2166/wst.2019.194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Addition of food waste (FW) as a co-substrate in anaerobic digesters of wastewater treatment plants is a desirable strategy towards achievement of the potential of wastewater treatment plants to become energy-neutral, diverting at the same time organic waste from landfills. Because substrate type is a driver of variations in phylogenetic structure of digester microbiomes, it is critical to understand how microbial communities respond to changes in substrate composition and concentration. In this work, high throughput sequencing was used to monitor the dynamics of microbiome changes in four parallel laboratory-scale anaerobic digesters treating sewage sludge during acclimation to an increasing amount of food waste. A co-occurrence network was constructed using data from 49 metagenomes sampled over the 161 days of the digesters' operation. More than half of the nodes in the network were clustered in two major modules, i.e. groups of highly interconnected taxa that had much fewer connections with taxa outside the group. The dynamics of co-occurrence networks evidenced shifts that occurred within microbial communities due to the addition of food waste in the co-digestion process. A diverse and reproducible group of hydrolytic and fermentative bacteria, syntrophic bacteria and methanogenic archaea appeared to grow in a concerted fashion to allow stable performance of anaerobic co-digestion at high FW.
Collapse
Affiliation(s)
- Esteban Orellana
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular 'Dr Héctor N. Torres' (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina E-mail: ; † These authors contributed equally to this work
| | - Carol Davies-Sala
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular 'Dr Héctor N. Torres' (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina E-mail: ; † These authors contributed equally to this work
| | - Leandro D Guerrero
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular 'Dr Héctor N. Torres' (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina E-mail:
| | - Ignacio Vardé
- Investigación, Desarrollo e Innovación, Benito Roggio Ambiental, Buenos Aires, Argentina
| | - Melisa Altina
- Investigación, Desarrollo e Innovación, Benito Roggio Ambiental, Buenos Aires, Argentina
| | - María Cielo Lorenzo
- Investigación, Desarrollo e Innovación, Benito Roggio Ambiental, Buenos Aires, Argentina
| | - Eva L Figuerola
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular 'Dr Héctor N. Torres' (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina E-mail: ; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA Buenos Aires, Argentina
| | - Rodrigo M Pontiggia
- Investigación, Desarrollo e Innovación, Benito Roggio Ambiental, Buenos Aires, Argentina
| | - Leonardo Erijman
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular 'Dr Héctor N. Torres' (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina E-mail: ; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA Buenos Aires, Argentina
| |
Collapse
|
193
|
Pomeroy LW, Moritz M, Garabed R. Network analyses of transhumance movements and simulations of foot-and-mouth disease virus transmission among mobile livestock in Cameroon. Epidemics 2019; 28:100334. [PMID: 31387783 DOI: 10.1016/j.epidem.2019.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/22/2019] [Accepted: 02/27/2019] [Indexed: 10/27/2022] Open
Abstract
Foot-and-mouth disease (FMD) affects cloven-hoofed livestock and agricultural economies worldwide. Analyses of the 2001 FMD outbreak in the United Kingdom informed how livestock movement contributed to disease spread. However, livestock reared in other locations use different production systems that might also influence disease dynamics. Here, we investigate a livestock production system known as transhumance, which is the practice of moving livestock between seasonal grazing areas. We built mechanistic models using livestock movement data from the Far North Region of Cameroon. We represented these data as a dynamic network over which we simulated disease transmission and examined three questions. First, we asked what were characteristics of simulated FMDV transmission across a transhumant pastoralist system. Second, we asked how simulated FMDV transmission across a transhumant pastoralist system differed from transmission across this same population held artificially stationary, thereby revealing the effect of movement on disease dynamics. Third, we asked if disease simulations on well-studied theoretical networks are similar to disease simulations on this empirical dynamic network. The results show that the empirical dynamic network was sparsely connected except for an eight-week period in September and October when pastoralists move from rainy season to dry season grazing areas. The mean epidemic size across all 3,744 simulations was 99.9% and the mean epidemic duration was 1.45 years. Disease simulations across the static network showed a smaller mean epidemic size (27.6%) and a similar epidemic duration (1.5 years). Epidemics simulated on theoretical networks showed similar final epidemic sizes (100%) and different mean durations. Our simulations indicate that transhumant livestock systems have the potential to host FMDV outbreaks that affect almost all livestock and last longer than a year. Furthermore, our comparison of empirical and theoretical networks underscores the importance of using empirical data to understand the role of mobility in the transmission of infectious diseases.
Collapse
Affiliation(s)
- Laura W Pomeroy
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA.
| | - Mark Moritz
- Department of Anthropology, The Ohio State University, Columbus, OH, USA
| | - Rebecca Garabed
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
194
|
Sabir JSM, El Omri A, Shaik NA, Banaganapalli B, Al-Shaeri MA, Alkenani NA, Hajrah NH, Awan ZA, Zrelli H, Elango R, Khan M. Identification of key regulatory genes connected to NF-κB family of proteins in visceral adipose tissues using gene expression and weighted protein interaction network. PLoS One 2019; 14:e0214337. [PMID: 31013288 PMCID: PMC6478283 DOI: 10.1371/journal.pone.0214337] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity is connected to the activation of chronic inflammatory pathways in both adipocytes and macrophages located in adipose tissues. The nuclear factor (NF)-κB is a central molecule involved in inflammatory pathways linked to the pathology of different complex metabolic disorders. Investigating the gene expression data in the adipose tissue would potentially unravel disease relevant gene interactions. The present study is aimed at creating a signature molecular network and at prioritizing the potential biomarkers interacting with NF-κB family of proteins in obesity using system biology approaches. The dataset GSE88837 associated with obesity was downloaded from Gene Expression Omnibus (GEO) database. Statistical analysis represented the differential expression of a total of 2650 genes in adipose tissues (p = <0.05). Using concepts like correlation, semantic similarity, and theoretical graph parameters we narrowed down genes to a network of 23 genes strongly connected with NF-κB family with higher significance. Functional enrichment analysis revealed 21 of 23 target genes of NF-κB were found to have a critical role in the pathophysiology of obesity. Interestingly, GEM and PPP1R13L were predicted as novel genes which may act as potential target or biomarkers of obesity as they occur with other 21 target genes with known obesity relationship. Our study concludes that NF-κB and prioritized target genes regulate the inflammation in adipose tissues through several molecular signaling pathways like NF-κB, PI3K-Akt, glucocorticoid receptor regulatory network, angiogenesis and cytokine pathways. This integrated system biology approaches can be applied for elucidating functional protein interaction networks of NF-κB protein family in different complex diseases. Our integrative and network-based approach for finding therapeutic targets in genomic data could accelerate the identification of novel drug targets for obesity.
Collapse
Affiliation(s)
- Jamal S. M. Sabir
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelfatteh El Omri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (MK); (AEO)
| | - Noor A. Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majed A. Al-Shaeri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Naser A. Alkenani
- Biology- Zoology Division, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nahid H. Hajrah
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King abdulaziz University, Jeddah, Saudi Arabia
| | - Zuhier A. Awan
- Department of Clinical Biochemistry. Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Houda Zrelli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhummadh Khan
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (MK); (AEO)
| |
Collapse
|
195
|
Millán AP, Torres JJ, Marro J. How Memory Conforms to Brain Development. Front Comput Neurosci 2019; 13:22. [PMID: 31057385 PMCID: PMC6477510 DOI: 10.3389/fncom.2019.00022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Nature exhibits countless examples of adaptive networks, whose topology evolves constantly coupled with the activity due to its function. The brain is an illustrative example of a system in which a dynamic complex network develops by the generation and pruning of synaptic contacts between neurons while memories are acquired and consolidated. Here, we consider a recently proposed brain developing model to study how mechanisms responsible for the evolution of brain structure affect and are affected by memory storage processes. Following recent experimental observations, we assume that the basic rules for adding and removing synapses depend on local synaptic currents at the respective neurons in addition to global mechanisms depending on the mean connectivity. In this way a feedback loop between "form" and "function" spontaneously emerges that influences the ability of the system to optimally store and retrieve sensory information in patterns of brain activity or memories. In particular, we report here that, as a consequence of such a feedback-loop, oscillations in the activity of the system among the memorized patterns can occur, depending on parameters, reminding mind dynamical processes. Such oscillations have their origin in the destabilization of memory attractors due to the pruning dynamics, which induces a kind of structural disorder or noise in the system at a long-term scale. This constantly modifies the synaptic disorder induced by the interference among the many patterns of activity memorized in the system. Such new intriguing oscillatory behavior is to be associated only to long-term synaptic mechanisms during the network evolution dynamics, and it does not depend on short-term synaptic processes, as assumed in other studies, that are not present in our model.
Collapse
Affiliation(s)
| | - Joaquín J. Torres
- Institute “Carlos I” for Theoretical and Computational Physics, University of Granada, Granada, Spain
| | | |
Collapse
|
196
|
Rudolph JD, Cox J. A Network Module for the Perseus Software for Computational Proteomics Facilitates Proteome Interaction Graph Analysis. J Proteome Res 2019; 18:2052-2064. [PMID: 30931570 PMCID: PMC6578358 DOI: 10.1021/acs.jproteome.8b00927] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Proteomics data analysis strongly benefits from not studying single proteins in isolation but taking their multivariate interdependence into account. We introduce PerseusNet, the new Perseus network module for the biological analysis of proteomics data. Proteomics is commonly used to generate networks, e.g., with affinity purification experiments, but networks are also used to explore proteomics data. PerseusNet supports the biomedical researcher for both modes of data analysis with a multitude of activities. For affinity purification, a volcano-plot-based statistical analysis method for network generation is featured which is scalable to large numbers of baits. For posttranslational modifications of proteins, such as phosphorylation, a collection of dedicated network analysis tools helps in elucidating cellular signaling events. Co-expression network analysis of proteomics data adopts established tools from transcriptome co-expression analysis. PerseusNet is extensible through a plugin architecture in a multi-lingual way, integrating analyses in C#, Python, and R, and is freely available at http://www.perseus-framework.org .
Collapse
Affiliation(s)
- Jan Daniel Rudolph
- Computational Systems Biochemistry , Max-Planck Institute of Biochemistry , Am Klopferspitz 18 , 82152 Martinsried , Germany
| | - Jürgen Cox
- Computational Systems Biochemistry , Max-Planck Institute of Biochemistry , Am Klopferspitz 18 , 82152 Martinsried , Germany.,Department of Biological and Medical Psychology , University of Bergen , Jonas Liesvei 91 , 5009 Bergen , Norway
| |
Collapse
|
197
|
Network Walking charts transcriptional dynamics of nitrogen signaling by integrating validated and predicted genome-wide interactions. Nat Commun 2019; 10:1569. [PMID: 30952851 PMCID: PMC6451032 DOI: 10.1038/s41467-019-09522-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 03/15/2019] [Indexed: 12/21/2022] Open
Abstract
Charting a temporal path in gene networks requires linking early transcription factor (TF)-triggered events to downstream effects. We scale-up a cell-based TF-perturbation assay to identify direct regulated targets of 33 nitrogen (N)-early response TFs encompassing 88% of N-responsive Arabidopsis genes. We uncover a duality where each TF is an inducer and repressor, and in vitro cis-motifs are typically specific to regulation directionality. Validated TF-targets (71,836) are used to refine precision of a time-inferred root network, connecting 145 N-responsive TFs and 311 targets. These data are used to chart network paths from direct TF1-regulated targets identified in cells to indirect targets responding only in planta via Network Walking. We uncover network paths from TGA1 and CRF4 to direct TF2 targets, which in turn regulate 76% and 87% of TF1 indirect targets in planta, respectively. These results have implications for N-use and the approach can reveal temporal networks for any biological system. Temporal control of transcriptional networks enables organisms to adapt to changing environment. Here, the authors use a scaled-up cell-based assay to identify direct targets of nitrogen-early responsive transcription factors and validate a network path mediating dynamic nitrogen signaling in Arabidopsis.
Collapse
|
198
|
Piras V, Chiow A, Selvarajoo K. Long‐range order and short‐range disorder in
Saccharomyces cerevisiae
biofilm. ENGINEERING BIOLOGY 2019. [DOI: 10.1049/enb.2018.5008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Vincent Piras
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS Université Paris‐Sud, Université Paris‐Saclay avenue de la Terrasse 91198 Gif‐sur‐Yvette Cedex France
| | - Adam Chiow
- Department of Pharmaceutical Engineering Singapore Institute of Technology 10 Dover Drive Singapore 138683 Singapore
| | - Kumar Selvarajoo
- Biotransformation Innovation Platform (BioTrans) Agency for Science, Technology & Research A∗STAR 61 Biopolis Drive, Proteos Singapore 138673 Singapore
| |
Collapse
|
199
|
Tight Regulation of Extracellular Superoxide Points to Its Vital Role in the Physiology of the Globally Relevant Roseobacter Clade. mBio 2019; 10:mBio.02668-18. [PMID: 30862752 PMCID: PMC6414704 DOI: 10.1128/mbio.02668-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There is a growing appreciation within animal and plant physiology that the reactive oxygen species (ROS) superoxide is not only detrimental but also essential for life. Yet, despite widespread production of extracellular superoxide by healthy bacteria and phytoplankton, this molecule remains associated with stress and death. Here, we quantify extracellular superoxide production by seven ecologically diverse bacteria within the Roseobacter clade and specifically target the link between extracellular superoxide and physiology for two species. We reveal for all species a strong inverse relationship between cell-normalized superoxide production rates and cell number. For exponentially growing cells of Ruegeria pomeroyi DSS-3 and Roseobacter sp. strain AzwK-3b, we show that superoxide levels are regulated in response to cell density through rapid modulation of gross production and not decay. Over a life cycle of batch cultures, extracellular superoxide levels are tightly regulated through a balance of both production and decay processes allowing for nearly constant levels of superoxide during active growth and minimal levels upon entering stationary phase. Further, removal of superoxide through the addition of exogenous superoxide dismutase during growth leads to significant growth inhibition. Overall, these results point to tight regulation of extracellular superoxide in representative members of the Roseobacter clade, consistent with a role for superoxide in growth regulation as widely acknowledged in fungal, animal, and plant physiology.IMPORTANCE Formation of reactive oxygen species (ROS) through partial reduction of molecular oxygen is widely associated with stress within microbial and marine systems. Nevertheless, widespread observations of the production of the ROS superoxide by healthy and actively growing marine bacteria and phytoplankton call into question the role of superoxide in the health and physiology of marine microbes. Here, we show that superoxide is produced by several marine bacteria within the widespread and abundant Roseobacter clade. Superoxide levels outside the cell are controlled via a tightly regulated balance of production and decay processes in response to cell density and life stage in batch culture. Removal of extracellular superoxide leads to substantial growth inhibition. These findings point to an essential role for superoxide in the health and growth of this ubiquitous group of microbes, and likely beyond.
Collapse
|
200
|
Zhu G, Li S, Wu J, Li F, Zhao XM. Identification of Functional Gene Modules Associated With STAT-Mediated Antiviral Responses to White Spot Syndrome Virus in Shrimp. Front Physiol 2019; 10:212. [PMID: 30914969 PMCID: PMC6421301 DOI: 10.3389/fphys.2019.00212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/19/2019] [Indexed: 11/13/2022] Open
Abstract
White spot syndrome virus (WSSV) is one of the major threats to shrimp aquaculture. It has been found that the signal transducer and activator of transcription (STAT) protein plays an important role in the antiviral immunity of shrimp with a WSSV infection. However, the mechanism that underlies the STAT-mediated antiviral responses in shrimp, against WSSV infection, remains unclear. In this work, based on the gene expression profiles of shrimp with an injection of WSSV and STAT double strand RNA (dsRNA), we constructed a gene co-expression network for shrimp and identified the gene modules that are possibly responsible for STAT-mediated antiviral responses. These gene modules are found enriched in the regulation of the viral process, JAK-STAT cascade and the regulation of immune effector process pathways. The gene modules identified here provide insights into the molecular mechanism that underlies the STAT-mediated antiviral response of shrimp, against WSSV.
Collapse
Affiliation(s)
- Guanghui Zhu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Department of Computer Science and Technology, Tongji University, Shanghai, China
| | - Shihao Li
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Jun Wu
- Department of Computer Science and Technology, Tongji University, Shanghai, China
| | - Fuhua Li
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Xing-Ming Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| |
Collapse
|