1
|
Unger Avila P, Padvitski T, Leote AC, Chen H, Saez-Rodriguez J, Kann M, Beyer A. Gene regulatory networks in disease and ageing. Nat Rev Nephrol 2024; 20:616-633. [PMID: 38867109 DOI: 10.1038/s41581-024-00849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
The precise control of gene expression is required for the maintenance of cellular homeostasis and proper cellular function, and the declining control of gene expression with age is considered a major contributor to age-associated changes in cellular physiology and disease. The coordination of gene expression can be represented through models of the molecular interactions that govern gene expression levels, so-called gene regulatory networks. Gene regulatory networks can represent interactions that occur through signal transduction, those that involve regulatory transcription factors, or statistical models of gene-gene relationships based on the premise that certain sets of genes tend to be coexpressed across a range of conditions and cell types. Advances in experimental and computational technologies have enabled the inference of these networks on an unprecedented scale and at unprecedented precision. Here, we delineate different types of gene regulatory networks and their cell-biological interpretation. We describe methods for inferring such networks from large-scale, multi-omics datasets and present applications that have aided our understanding of cellular ageing and disease mechanisms.
Collapse
Affiliation(s)
- Paula Unger Avila
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tsimafei Padvitski
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ana Carolina Leote
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - He Chen
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Martin Kann
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas Beyer
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
2
|
James K, Alsobhe A, Cockell SJ, Wipat A, Pocock M. Integration of probabilistic functional networks without an external Gold Standard. BMC Bioinformatics 2022; 23:302. [PMID: 35879662 PMCID: PMC9316706 DOI: 10.1186/s12859-022-04834-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Probabilistic functional integrated networks (PFINs) are designed to aid our understanding of cellular biology and can be used to generate testable hypotheses about protein function. PFINs are generally created by scoring the quality of interaction datasets against a Gold Standard dataset, usually chosen from a separate high-quality data source, prior to their integration. Use of an external Gold Standard has several drawbacks, including data redundancy, data loss and the need for identifier mapping, which can complicate the network build and impact on PFIN performance. Additionally, there typically are no Gold Standard data for non-model organisms. RESULTS We describe the development of an integration technique, ssNet, that scores and integrates both high-throughput and low-throughout data from a single source database in a consistent manner without the need for an external Gold Standard dataset. Using data from Saccharomyces cerevisiae we show that ssNet is easier and faster, overcoming the challenges of data redundancy, Gold Standard bias and ID mapping. In addition ssNet results in less loss of data and produces a more complete network. CONCLUSIONS The ssNet method allows PFINs to be built successfully from a single database, while producing comparable network performance to networks scored using an external Gold Standard source and with reduced data loss.
Collapse
Affiliation(s)
- Katherine James
- Department of Applied Sciences, Northumbria University, Sandyford Rd, Newcastle upon Tyne, NE1 8ST, UK. .,Interdisciplinary Computing and Complex BioSystems Group, Newcastle University, Science Square, Newcastle upon Tyne, NE4 5TG, UK.
| | - Aoesha Alsobhe
- Interdisciplinary Computing and Complex BioSystems Group, Newcastle University, Science Square, Newcastle upon Tyne, NE4 5TG, UK.,Saudi Electronic University, Abi Bakr As Siddiq Branch Rd, Riyadh, 1332, Saudi Arabia
| | - Simon J Cockell
- School of Biomedical, Nutritional and Sports Science, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Anil Wipat
- Interdisciplinary Computing and Complex BioSystems Group, Newcastle University, Science Square, Newcastle upon Tyne, NE4 5TG, UK
| | - Matthew Pocock
- Interdisciplinary Computing and Complex BioSystems Group, Newcastle University, Science Square, Newcastle upon Tyne, NE4 5TG, UK
| |
Collapse
|
3
|
Abstract
Making computing machines mimic living organisms has captured the imagination of many since the dawn of digital computers. However, today’s artificial intelligence technologies fall short of replicating even the basic autopoietic and cognitive behaviors found in primitive biological systems. According to Charles Darwin, the difference in mind between humans and higher animals, great as it is, certainly is one of degree and not of kind. Autopoiesis refers to the behavior of a system that replicates itself and maintains identity and stability while facing fluctuations caused by external influences. Cognitive behaviors model the system’s state, sense internal and external changes, analyze, predict and take action to mitigate any risk to its functional fulfillment. How did intelligence evolve? what is the relationship between the mind and body? Answers to these questions should guide us to infuse autopoietic and cognitive behaviors into digital machines. In this paper, we show how to use the structural machine to build a cognitive reasoning system that integrates the knowledge from various digital symbolic and sub-symbolic computations. This approach is analogous to how the neocortex repurposed the reptilian brain and paves the path for digital machines to mimic living organisms using an integrated knowledge representation from different sources.
Collapse
|
4
|
OUP accepted manuscript. Brief Funct Genomics 2022; 21:243-269. [DOI: 10.1093/bfgp/elac007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/14/2022] Open
|
5
|
Capelle CM, Zeng N, Danileviciute E, Rodrigues SF, Ollert M, Balling R, He FQ. Identification of VIMP as a gene inhibiting cytokine production in human CD4+ effector T cells. iScience 2021; 24:102289. [PMID: 33851102 PMCID: PMC8024663 DOI: 10.1016/j.isci.2021.102289] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/08/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Many players regulating the CD4+ T cell-mediated inflammatory response have already been identified. However, the critical nodes that constitute the regulatory and signaling networks underlying CD4 T cell responses are still missing. Using a correlation-network-guided approach, here we identified VIMP (VCP-interacting membrane protein), one of the 25 genes encoding selenoproteins in humans, as a gene regulating the effector functions of human CD4 T cells, especially production of several cytokines including IL2 and CSF2. We identified VIMP as an endogenous inhibitor of cytokine production in CD4 effector T cells via both the E2F5 transcription regulatory pathway and the Ca2+/NFATC2 signaling pathway. Our work not only indicates that VIMP might be a promising therapeutic target for various inflammation-associated diseases but also shows that our network-guided approach can significantly aid in predicting new functions of the genes of interest.
Collapse
Affiliation(s)
- Christophe M. Capelle
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, 4354 Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2, avenue de Université, 4365 Esch-sur-Alzette, Luxembourg
| | - Ni Zeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, 4354 Esch-sur-Alzette, Luxembourg
| | - Egle Danileviciute
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, 4354 Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, 4367 Belvaux, Luxembourg
| | - Sabrina Freitas Rodrigues
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, 4367 Belvaux, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, 4354 Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis (ORCA), University of Southern Denmark, Odense, 5000 C, Denmark
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, 4367 Belvaux, Luxembourg
| | - Feng Q. He
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, 4354 Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, 4367 Belvaux, Luxembourg
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
6
|
Mita P, Sun X, Fenyö D, Kahler DJ, Li D, Agmon N, Wudzinska A, Keegan S, Bader JS, Yun C, Boeke JD. BRCA1 and S phase DNA repair pathways restrict LINE-1 retrotransposition in human cells. Nat Struct Mol Biol 2020; 27:179-191. [PMID: 32042152 PMCID: PMC7082080 DOI: 10.1038/s41594-020-0374-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 01/02/2020] [Indexed: 12/30/2022]
Abstract
Long interspersed element-1 (LINE-1 or L1) is the only autonomous retrotransposon active in human cells. Different host factors have been shown to influence L1 mobility however, systematic analyses of these factors are limited. Here, we developed a high-throughput microscopy-based retrotransposition assay that identified the Double-Stranded Break (DSB) repair and Fanconi Anemia factors active in the S/G2 phase as potent inhibitors and regulators of L1 activity. In particular BRCA1, an E3 ubiquitin ligase with a key role in several DNA repair pathways, directly affects L1 retrotransposition frequency and structure and also plays a distinct role in controlling L1 ORF2 protein translation through L1 mRNA binding. These results suggest the existence of a “battleground” at the DNA replication fork between HR factors and L1 retrotransposons, and revealing a potential role for L1 in the genotypic evolution of tumors characterized by BRCA1 and HR repair deficiencies.
Collapse
Affiliation(s)
- Paolo Mita
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA.
| | - Xiaoji Sun
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA.,Cellarity Inc., Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - David J Kahler
- High Throughput Biology Core, NYU Langone Health, New York, NY, USA.,Planet Pharma, Boston, MA, USA
| | - Donghui Li
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA.,Flagship VL58, Inc., Cambridge, MA, USA
| | - Neta Agmon
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Aleksandra Wudzinska
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Sarah Keegan
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Joel S Bader
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chi Yun
- High Throughput Biology Core, NYU Langone Health, New York, NY, USA
| | - Jef D Boeke
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
7
|
Licon K, Shen JP, Munson BP, Michaca M, Fassino C, Fassino L, Kreisberg JF, Ideker T. Ultrahigh-Density Screens for Genome-Wide Yeast EMAPs in a Single Plate. Methods Mol Biol 2019; 2049:73-85. [PMID: 31602605 PMCID: PMC7423300 DOI: 10.1007/978-1-4939-9736-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Systematic measurements of genetic interactions have been used to classify gene functions and to categorize genes into protein complexes, functional pathways and biological processes. This protocol describes how to perform a high-throughput genetic interaction screen in S. cerevisiae using a variant of epistatic miniarray profiles (E-MAP) in which the fitnesses of 6144 colonies are measured simultaneously. We also describe the computational methods to analyze the resulting data.
Collapse
Affiliation(s)
| | | | - Brenton P Munson
- Department of Medicine, UC San Diego, La Jolla, CA, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
| | | | - Cole Fassino
- Department of Medicine, UC San Diego, La Jolla, CA, USA
| | - Luke Fassino
- Department of Medicine, UC San Diego, La Jolla, CA, USA
| | | | - Trey Ideker
- Department of Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Yang R, Watson D, Williams J, Kumar R, Campbell R, Mudunuri U, Hammamieh R, Jett M. PanoromiX: a time-course network medicine platform integrating molecular assays and pathophenotypic data. BMC Bioinformatics 2018; 19:458. [PMID: 30497372 PMCID: PMC6267067 DOI: 10.1186/s12859-018-2494-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 11/13/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Network medicine aims to map molecular perturbations of any given diseases onto complex networks with functional interdependencies that underlie a pathological phenotype. Furthermore, investigating the time dimension of disease progression from a network perspective is key to gaining key insights to the disease process and to identify diagnostic or therapeutic targets. Existing platforms are ineffective to modularize the large complex systems into subgroups and consolidate heterogeneous data to web-based interactive animation. RESULTS We have developed PanoromiX platform, a data-agnostic dynamic interactive visualization web application, enables the visualization of outputs from genome based molecular assays onto modular and interactive networks that are correlated with any pathophenotypic data (MRI, Xray, behavioral, etc.) over a time course all in one pane. As a result, PanoromiX reveals the complex organizing principles that orchestrate a disease-pathology from a gene regulatory network (nodes, edges, hubs, etc.) perspective instead of snap shots of assays. Without extensive programming experience, users can design, share, and interpret their dynamic networks through the PanoromiX platform with rich built-in functionalities. CONCLUSIONS This emergent tool of network medicine is the first to visualize the interconnectedness of tailored genome assays to pathological networks and phenotypes for cells or organisms in a data-agnostic manner. As an advanced network medicine tool, PanoromiX allows monitoring of panel of biomarker perturbations over the progression of diseases, disease classification based on changing network modules that corresponds to specific patho-phenotype as opposed to clinical symptoms, systematic exploration of complex molecular interactions and distinct disease states via regulatory network changes, and the discovery of novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Ruoting Yang
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702-5010 USA
- Integrative Systems Biology Program, US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD 21702-5010 USA
| | - Daniel Watson
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702-5010 USA
| | - Joshua Williams
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702-5010 USA
- Integrative Systems Biology Program, US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD 21702-5010 USA
| | - Raina Kumar
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702-5010 USA
- Integrative Systems Biology Program, US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD 21702-5010 USA
| | - Ross Campbell
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702-5010 USA
- Integrative Systems Biology Program, US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD 21702-5010 USA
| | - Uma Mudunuri
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702-5010 USA
| | - Rasha Hammamieh
- Integrative Systems Biology Program, US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD 21702-5010 USA
| | - Marti Jett
- Integrative Systems Biology Program, US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD 21702-5010 USA
| |
Collapse
|
9
|
Feng C, Song C, Ning Z, Ai B, Wang Q, Xu Y, Li M, Bai X, Zhao J, Liu Y, Li X, Zhang J, Li C. ce-Subpathway: Identification of ceRNA-mediated subpathways via joint power of ceRNAs and pathway topologies. J Cell Mol Med 2018; 23:967-984. [PMID: 30421585 PMCID: PMC6349186 DOI: 10.1111/jcmm.13997] [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: 06/12/2018] [Revised: 08/28/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022] Open
Abstract
Competing endogenous RNAs (ceRNAs) represent a novel mechanism of gene regulation that may mediate key subpathway regions and contribute to the altered activities of pathways. However, the classical methods used to identify pathways fail to specifically consider ceRNAs within the pathways and key regions impacted by them. We proposed a powerful strategy named ce-Subpathway for the identification of ceRNA-mediated functional subpathways. It provided an effective level of pathway analysis via integrating ceRNAs, differentially expressed (DE) genes and their key regions within the given pathways. We respectively analysed one pulmonary arterial hypertension (PAH) and one myocardial infarction (MI) data sets and demonstrated that ce-Subpathway could identify many subpathways whose corresponding entire pathways were ignored by those non-ceRNA-mediated pathway identification methods. And these pathways have been well reported to be associated with PAH/MI-related cardiovascular diseases. Further evidence showed reliability of ceRNA interactions and robustness/reproducibility of the ce-Subpathway strategy by several data sets of different cancers, including breast cancer, oesophageal cancer and colon cancer. Survival analysis was finally applied to illustrate the clinical application value of the ceRNA-mediated functional subpathways using another data sets of pancreatic cancer. Comprehensive analyses have shown the power of a joint ceRNAs/DE genes and subpathway strategy based on their topologies.
Collapse
Affiliation(s)
- Chenchen Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Chao Song
- Department of Pharmacology, Daqing Campus, Harbin Medical University, Daqing, China
| | - Ziyu Ning
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Bo Ai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Qiuyu Wang
- School of Nursing, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yong Xu
- The fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Meng Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xuefeng Bai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jianmei Zhao
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yuejuan Liu
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xuecang Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jian Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| |
Collapse
|
10
|
Martínez-Cano DJ, Bor G, Moya A, Delaye L. Testing the Domino Theory of Gene Loss in Buchnera aphidicola: The Relevance of Epistatic Interactions. Life (Basel) 2018; 8:life8020017. [PMID: 29843462 PMCID: PMC6027505 DOI: 10.3390/life8020017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023] Open
Abstract
The domino theory of gene loss states that when some particular gene loses its function and cripples a cellular function, selection will relax in all functionally related genes, which may allow for the non-functionalization and loss of these genes. Here we study the role of epistasis in determining the pattern of gene losses in a set of genes participating in cell envelope biogenesis in the endosymbiotic bacteria Buchnera aphidicola. We provide statistical evidence indicating pairs of genes in B. aphidicola showing correlated gene loss tend to have orthologs in Escherichia coli known to have alleviating epistasis. In contrast, pairs of genes in B. aphidicola not showing correlated gene loss tend to have orthologs in E. coli known to have aggravating epistasis. These results suggest that during the process of genome reduction in B. aphidicola by gene loss, positive or alleviating epistasis facilitates correlated gene losses while negative or aggravating epistasis impairs correlated gene losses. We interpret this as evidence that the reduced proteome of B. aphidicola contains less pathway redundancy and more compensatory interactions, mimicking the situation of E. coli when grown under environmental constrains.
Collapse
Affiliation(s)
- David J Martínez-Cano
- Departamento de Ingeniería Genética, CINVESTAV Irapuato, Km. 9.6 Libramiento Norte Carretera Irapuato-León, 36821 Irapuato, Guanajuato, Mexico.
| | - Gil Bor
- CIMAT, A.P. 402, Guanajuato 36000, Gto., Mexico.
| | - Andrés Moya
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO)-Salud Pública, Avenida de Catalunya 21, 46020 València, Spain.
- Institute for Integrative Systems Biology, Universitat de València, Calle Catedrático José Beltrán 2, 46980 Paterna, València, Spain.
| | - Luis Delaye
- Departamento de Ingeniería Genética, CINVESTAV Irapuato, Km. 9.6 Libramiento Norte Carretera Irapuato-León, 36821 Irapuato, Guanajuato, Mexico.
| |
Collapse
|
11
|
Ashraf N, Basu S, Narula K, Ghosh S, Tayal R, Gangisetty N, Biswas S, Aggarwal PR, Chakraborty N, Chakraborty S. Integrative network analyses of wilt transcriptome in chickpea reveal genotype dependent regulatory hubs in immunity and susceptibility. Sci Rep 2018; 8:6528. [PMID: 29695764 PMCID: PMC5916944 DOI: 10.1038/s41598-018-19919-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022] Open
Abstract
Host specific resistance and non-host resistance are two plant immune responses to counter pathogen invasion. Gene network organizing principles leading to quantitative differences in resistant and susceptible host during host specific resistance are poorly understood. Vascular wilt caused by root pathogen Fusarium species is complex and governed by host specific resistance in crop plants, including chickpea. Here, we temporally profiled two contrasting chickpea genotypes in disease and immune state to better understand gene expression switches in host specific resistance. Integrative gene-regulatory network elucidated tangible insight into interaction coordinators leading to pathway determination governing distinct (disease or immune) phenotypes. Global network analysis identified five major hubs with 389 co-regulated genes. Functional enrichment revealed immunome containing three subnetworks involving CTI, PTI and ETI and wilt diseasome encompassing four subnetworks highlighting pathogen perception, penetration, colonization and disease establishment. These subnetworks likely represent key components that coordinate various biological processes favouring defence or disease. Furthermore, we identified core 76 disease/immunity related genes through subcellular analysis. Our regularized network with robust statistical assessment captured known and unexpected gene interaction, candidate novel regulators as future biomarkers and first time showed system-wide quantitative architecture corresponding to genotypic characteristics in wilt landscape.
Collapse
Affiliation(s)
- Nasheeman Ashraf
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Swaraj Basu
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Kanika Narula
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sudip Ghosh
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Rajul Tayal
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Nagaraju Gangisetty
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sushmita Biswas
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Pooja R Aggarwal
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Niranjan Chakraborty
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Subhra Chakraborty
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
12
|
Lancour D, Naj A, Mayeux R, Haines JL, Pericak-Vance MA, Schellenberg GD, Crovella M, Farrer LA, Kasif S. One for all and all for One: Improving replication of genetic studies through network diffusion. PLoS Genet 2018; 14:e1007306. [PMID: 29684019 PMCID: PMC5933817 DOI: 10.1371/journal.pgen.1007306] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 05/03/2018] [Accepted: 03/11/2018] [Indexed: 12/31/2022] Open
Abstract
Improving accuracy in genetic studies would greatly accelerate understanding the genetic basis of complex diseases. One approach to achieve such an improvement for risk variants identified by the genome wide association study (GWAS) approach is to incorporate previously known biology when screening variants across the genome. We developed a simple approach for improving the prioritization of candidate disease genes that incorporates a network diffusion of scores from known disease genes using a protein network and a novel integration with GWAS risk scores, and tested this approach on a large Alzheimer disease (AD) GWAS dataset. Using a statistical bootstrap approach, we cross-validated the method and for the first time showed that a network approach improves the expected replication rates in GWAS studies. Several novel AD genes were predicted including CR2, SHARPIN, and PTPN2. Our re-prioritized results are enriched for established known AD-associated biological pathways including inflammation, immune response, and metabolism, whereas standard non-prioritized results were not. Our findings support a strategy of considering network information when investigating genetic risk factors. Integrating multiple types of -omics data is a rapidly growing research area due in part to the increasing amount of diverse and publicly accessible data. In this study, we demonstrated that integration of genetic association and protein interaction data using a network diffusion approach measurably improves reproducibility of top candidate genes. Application of this approach to Alzheimer disease (AD) using a large dataset assembled by the Alzheimer’s Disease Genetics Consortium identified several novel candidate AD genes that are supported by pre-existing knowledge of AD pathobiology. Our findings support a strategy of considering network information when investigating genetic risk factors. Finally, we developed a transparent and easy-to-use R package that can facilitate the extension of our methodology to other phenotypes for which genetic data are available.
Collapse
Affiliation(s)
- Daniel Lancour
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Adam Naj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Richard Mayeux
- Department of Neurology and Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Margaret A. Pericak-Vance
- Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mark Crovella
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Computer Science, Boston University, Boston, Massachusetts, United States of America
| | - Lindsay A. Farrer
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States of America
- * E-mail:
| | - Simon Kasif
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
13
|
Sharma S, Petsalaki E. Application of CRISPR-Cas9 Based Genome-Wide Screening Approaches to Study Cellular Signalling Mechanisms. Int J Mol Sci 2018; 19:E933. [PMID: 29561791 PMCID: PMC5979383 DOI: 10.3390/ijms19040933] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/26/2022] Open
Abstract
The cellular signalling process is a highly complex mechanism, involving multiple players, which together orchestrate the cell's response to environmental changes and perturbations. Given the multitude of genes that participate in the process of cellular signalling, its study in a genome-wide manner has proven challenging. Recent advances in gene editing technologies, including clustered regularly-interspaced short palindromic repeats/Cas9 (CRISPR/Cas9) approaches, have opened new opportunities to investigate global regulatory signalling programs of cells in an unbiased manner. In this review, we focus on how the application of pooled genetic screening approaches using the CRISPR/Cas9 system has contributed to a systematic understanding of cellular signalling processes in normal and disease contexts.
Collapse
Affiliation(s)
- Sumana Sharma
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - Evangelia Petsalaki
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| |
Collapse
|
14
|
Ignatius Pang CN, Goel A, Wilkins MR. Investigating the Network Basis of Negative Genetic Interactions in Saccharomyces cerevisiae with Integrated Biological Networks and Triplet Motif Analysis. J Proteome Res 2018; 17:1014-1030. [DOI: 10.1021/acs.jproteome.7b00649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Chi Nam Ignatius Pang
- Systems
Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Apurv Goel
- Systems
Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Marc R. Wilkins
- Systems
Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
15
|
Fang H, Li X, Zan X, Shen L, Ma R, Liu W. Signaling pathway impact analysis by incorporating the importance and specificity of genes (SPIA-IS). Comput Biol Chem 2017; 71:236-244. [DOI: 10.1016/j.compbiolchem.2017.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/25/2017] [Indexed: 01/28/2023]
|
16
|
Qi J, Ma L, Wang X, Li Y, Wang K. Observation of significant biomarkers in osteosarcoma via integrating module- identification method with attract. Cancer Biomark 2017; 20:87-93. [PMID: 28759958 DOI: 10.3233/cbm-170144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Osteosarcoma (OS) is the most frequent type of bone malignancy, and this disease has a poor prognosis. We aimed to identify the significant genes related with OS by integrating module-identification method and attract approach. METHODS OS-related microarray data E-GEOD-36001 were obtained from ArrayExpress database, and then protein-protein interaction (PPI) networks of normal and OS were re-weighted by means of spearman correlation coefficient (SCC). Next, maximal cliques were detected from the re-weighted PPI networks using clusteringbased on maximal cliques approach. Afterwards, highly overlapped cliques were merged according to the interconnectivity, following by candidate modules and seed modules identification. Attract proposed by Mar et al. who have suggested that this approach can extract and annotate the gene-sets which can distinguish between disease and control samples, and obtained differences of these gene-sets among the expression profile of samples were defined as attractors. Thus, we applied attract method to extract differential modules from the seed modules, and these obtained differential modules were defined as attractors. The genes in attractors were determined as attractor genes. RESULTS After eliminating the maximal cliques with nodes less than 4, there were 1,884 and 528 maximal cliques in normal and OS PPI networks, which were used to conduct module analysis. A total of 60 and 19 candidate modules were obtained in control and OS PPI networks, respectively. By comparing with normal group, 2 seed module pairs with similar gene composition were found. Significantly, based on attract method, we found that these 2 modules were differential. These 2 modules had the same gene size with 4 genes. Of note, genes CCNB1 and KIF11 simultaneously appeared in these two attractors. CONCLUSIONS We successfully identified two attractors via integrating module-identification method and attract approach, and attractor genes, for example, CCNB1 and KIF11 might play pathophysiological roles in OS development and progression.
Collapse
Affiliation(s)
- Jie Qi
- Department of Orthopaedics, Shaanxi Provicial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Liang Ma
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, China
| | - Xiaogang Wang
- Out-patient Department, Affiliated Tumor Hospital of Xinjiang Medical University, Wuluumuqi 830011, Xinjiang, China
| | - Ying Li
- Beijing Spirallink Medical Research Institute, Beijing 100054, China
| | - Kejun Wang
- Department of Orthopaedics, Jingzhou Central Hospital, Jingzhou 434020, Hubei, China
| |
Collapse
|
17
|
Garland J. Unravelling the complexity of signalling networks in cancer: A review of the increasing role for computational modelling. Crit Rev Oncol Hematol 2017; 117:73-113. [PMID: 28807238 DOI: 10.1016/j.critrevonc.2017.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 06/01/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
Cancer induction is a highly complex process involving hundreds of different inducers but whose eventual outcome is the same. Clearly, it is essential to understand how signalling pathways and networks generated by these inducers interact to regulate cell behaviour and create the cancer phenotype. While enormous strides have been made in identifying key networking profiles, the amount of data generated far exceeds our ability to understand how it all "fits together". The number of potential interactions is astronomically large and requires novel approaches and extreme computation methods to dissect them out. However, such methodologies have high intrinsic mathematical and conceptual content which is difficult to follow. This review explains how computation modelling is progressively finding solutions and also revealing unexpected and unpredictable nano-scale molecular behaviours extremely relevant to how signalling and networking are coherently integrated. It is divided into linked sections illustrated by numerous figures from the literature describing different approaches and offering visual portrayals of networking and major conceptual advances in the field. First, the problem of signalling complexity and data collection is illustrated for only a small selection of known oncogenes. Next, new concepts from biophysics, molecular behaviours, kinetics, organisation at the nano level and predictive models are presented. These areas include: visual representations of networking, Energy Landscapes and energy transfer/dissemination (entropy); diffusion, percolation; molecular crowding; protein allostery; quinary structure and fractal distributions; energy management, metabolism and re-examination of the Warburg effect. The importance of unravelling complex network interactions is then illustrated for some widely-used drugs in cancer therapy whose interactions are very extensive. Finally, use of computational modelling to develop micro- and nano- functional models ("bottom-up" research) is highlighted. The review concludes that computational modelling is an essential part of cancer research and is vital to understanding network formation and molecular behaviours that are associated with it. Its role is increasingly essential because it is unravelling the huge complexity of cancer induction otherwise unattainable by any other approach.
Collapse
Affiliation(s)
- John Garland
- Manchester Interdisciplinary Biocentre, Manchester University, Manchester, UK.
| |
Collapse
|
18
|
Christodoulou EG, Yang H, Lademann F, Pilarsky C, Beyer A, Schroeder M. Detection of COPB2 as a KRAS synthetic lethal partner through integration of functional genomics screens. Oncotarget 2017; 8:34283-34297. [PMID: 28415695 PMCID: PMC5470967 DOI: 10.18632/oncotarget.16079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 03/01/2017] [Indexed: 12/30/2022] Open
Abstract
Mutated KRAS plays an important role in many cancers. Although targeting KRAS directly is difficult, indirect inactivation via synthetic lethal partners (SLPs) is promising. Yet to date, there are no SLPs from high-throughput RNAi screening, which are supported by multiple screens. Here, we address this problem by aggregating and ranking data over three independent high-throughput screens. We integrate rankings by minimizing the displacement and by considering established methods such as RIGER and RSA.Our meta analysis reveals COPB2 as a potential SLP of KRAS with good support from all three screens. COPB2 is a coatomer subunit and its knock down has already been linked to disabled autophagy and reduced tumor growth. We confirm COPB2 as SLP in knock down experiments on pancreas and colorectal cancer cell lines.Overall, consistent integration of high throughput data can generate candidate synthetic lethal partners, which individual screens do not uncover. Concretely, we reveal and confirm that COPB2 is a synthetic lethal partner of KRAS and hence a promising cancer target. Ligands inhibiting COPB2 may, therefore, be promising new cancer drugs.
Collapse
Affiliation(s)
- Eleni G. Christodoulou
- Biotechnology Center, TU Dresden, Dresden, Germany
- Department of Medical Oncology, National Cancer Center of Singapore, Singapore
| | - Hai Yang
- Chirurgische Klinik, Translational Research Center, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Christian Pilarsky
- Chirurgische Klinik, Translational Research Center, Universitätsklinikum Erlangen, Erlangen, Germany
- Medizinische Fakultät Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Beyer
- Biotechnology Center, TU Dresden, Dresden, Germany
- Cellular Networks and Systems Biology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
19
|
Calhoun VD. Predicting schizophrenia by fusing networks from SNPs, DNA methylation and fMRI data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2016:1447-1450. [PMID: 28268598 DOI: 10.1109/embc.2016.7590981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In order to comprehensively utilize complementary information from multiple types of data for better disease diagnosis, in this study, we applied a network fusion based approach to integrating three types of data including genetic, epigenetic and neuroimaging data from a study of schizophrenia patients (SCZ). A network is a map of interactions, which contributes to investigating the connectivity of components or links between sub-units. We exploited the potential of using networks as features for discriminating SCZ from healthy controls. We first constructed a single network from each type of data. Then we built four fused networks by the network fusion method: three fused networks for each combination of two types of data and one fused network for all three data types. Based on the local consistency of network, we can predict the group of the unlabeled SCZ subjects. The group prediction method was applied to test the power of network-based features and the performance was evaluated by a 10-fold cross validation. The results show that the prediction accuracy is the highest when applying our prediction method to the fused network derived from three data types among 7 tested networks. As a conclusion, integrative approaches that can comprehensively utilize multiple types of data are more useful for diagnosis and prediction.
Collapse
|
20
|
Ihsan MZ, Ahmad SJN, Shah ZH, Rehman HM, Aslam Z, Ahuja I, Bones AM, Ahmad JN. Gene Mining for Proline Based Signaling Proteins in Cell Wall of Arabidopsis thaliana. FRONTIERS IN PLANT SCIENCE 2017; 8:233. [PMID: 28289422 PMCID: PMC5326801 DOI: 10.3389/fpls.2017.00233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 02/07/2017] [Indexed: 05/29/2023]
Abstract
The cell wall (CW) as a first line of defense against biotic and abiotic stresses is of primary importance in plant biology. The proteins associated with cell walls play a significant role in determining a plant's sustainability to adverse environmental conditions. In this work, the genes encoding cell wall proteins (CWPs) in Arabidopsis were identified and functionally classified using geneMANIA and GENEVESTIGATOR with published microarrays data. This yielded 1605 genes, out of which 58 genes encoded proline-rich proteins (PRPs) and glycine-rich proteins (GRPs). Here, we have focused on the cellular compartmentalization, biological processes, and molecular functioning of proline-rich CWPs along with their expression at different plant developmental stages. The mined genes were categorized into five classes on the basis of the type of PRPs encoded in the cell wall of Arabidopsis thaliana. We review the domain structure and function of each class of protein, many with respect to the developmental stages of the plant. We have then used networks, hierarchical clustering and correlations to analyze co-expression, co-localization, genetic, and physical interactions and shared protein domains of these PRPs. This has given us further insight into these functionally important CWPs and identified a number of potentially new cell-wall related proteins in A. thaliana.
Collapse
Affiliation(s)
- Muhammad Z. Ihsan
- Cholistan Institute of Desert Studies, The Islamia University BahawalpurBahawalpur, Pakistan
| | - Samina J. N. Ahmad
- Plant Stress Physiology and Molecular Biology Lab, Department of Botany, University of Agriculture FaisalabadFaisalabad, Pakistan
- Integrated Genomics Cellular Developmental and Biotechnology Lab, Department of Entomology, University of Agriculture FaisalabadFaisalabad, Pakistan
| | - Zahid Hussain Shah
- Department of Arid Land Agriculture, Faculty of Meteorology, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Hafiz M. Rehman
- Department of Electronic and Biomedical Engineering, Chonnam National UniversityGwangju, South Korea
| | - Zubair Aslam
- Department of Agronomy, University of Agriculture FaisalabadFaisalabad, Pakistan
| | - Ishita Ahuja
- Department of Biology, Norwegian University of Science and TechnologyTrondheim, Norway
| | - Atle M. Bones
- Department of Biology, Norwegian University of Science and TechnologyTrondheim, Norway
| | - Jam N. Ahmad
- Plant Stress Physiology and Molecular Biology Lab, Department of Botany, University of Agriculture FaisalabadFaisalabad, Pakistan
- Integrated Genomics Cellular Developmental and Biotechnology Lab, Department of Entomology, University of Agriculture FaisalabadFaisalabad, Pakistan
| |
Collapse
|
21
|
Korla K, Chandra N. A Systems Perspective of Signalling Networks in Host–Pathogen Interactions. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
Cava C, Colaprico A, Bertoli G, Graudenzi A, Silva TC, Olsen C, Noushmehr H, Bontempi G, Mauri G, Castiglioni I. SpidermiR: An R/Bioconductor Package for Integrative Analysis with miRNA Data. Int J Mol Sci 2017; 18:ijms18020274. [PMID: 28134831 PMCID: PMC5343810 DOI: 10.3390/ijms18020274] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/24/2017] [Indexed: 02/08/2023] Open
Abstract
Gene Regulatory Networks (GRNs) control many biological systems, but how such network coordination is shaped is still unknown. GRNs can be subdivided into basic connections that describe how the network members interact e.g., co-expression, physical interaction, co-localization, genetic influence, pathways, and shared protein domains. The important regulatory mechanisms of these networks involve miRNAs. We developed an R/Bioconductor package, namely SpidermiR, which offers an easy access to both GRNs and miRNAs to the end user, and integrates this information with differentially expressed genes obtained from The Cancer Genome Atlas. Specifically, SpidermiR allows the users to: (i) query and download GRNs and miRNAs from validated and predicted repositories; (ii) integrate miRNAs with GRNs in order to obtain miRNA-gene-gene and miRNA-protein-protein interactions, and to analyze miRNA GRNs in order to identify miRNA-gene communities; and (iii) graphically visualize the results of the analyses. These analyses can be performed through a single interface and without the need for any downloads. The full data sets are then rapidly integrated and processed locally.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology National Research Council (IBFM-CNR), Segrate (Mi) 20090, Italy.
| | - Antonio Colaprico
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels 1050, Belgium.
- Machine Learning Group (MLG), Department d'Informatique, Universite libre de Bruxelles (ULB), Brussels 1050, Belgium.
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology National Research Council (IBFM-CNR), Segrate (Mi) 20090, Italy.
| | - Alex Graudenzi
- Institute of Molecular Bioimaging and Physiology National Research Council (IBFM-CNR), Segrate (Mi) 20090, Italy.
| | - Tiago C Silva
- Department of Genetics Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo 14049-900, Brazil.
| | - Catharina Olsen
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels 1050, Belgium.
- Machine Learning Group (MLG), Department d'Informatique, Universite libre de Bruxelles (ULB), Brussels 1050, Belgium.
| | - Houtan Noushmehr
- Department of Genetics Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo 14049-900, Brazil.
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Gianluca Bontempi
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels 1050, Belgium.
- Machine Learning Group (MLG), Department d'Informatique, Universite libre de Bruxelles (ULB), Brussels 1050, Belgium.
| | - Giancarlo Mauri
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan 20125, Italy.
- SYSBIO Centre of Systems Biology (SYSBIO), Milan 20126, Italy.
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology National Research Council (IBFM-CNR), Segrate (Mi) 20090, Italy.
| |
Collapse
|
23
|
Jaitin DA, Weiner A, Yofe I, Lara-Astiaso D, Keren-Shaul H, David E, Salame TM, Tanay A, van Oudenaarden A, Amit I. Dissecting Immune Circuits by Linking CRISPR-Pooled Screens with Single-Cell RNA-Seq. Cell 2016; 167:1883-1896.e15. [DOI: 10.1016/j.cell.2016.11.039] [Citation(s) in RCA: 448] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 11/14/2016] [Accepted: 11/19/2016] [Indexed: 12/15/2022]
|
24
|
Ung MH, Liu CC, Cheng C. Integrative analysis of cancer genes in a functional interactome. Sci Rep 2016; 6:29228. [PMID: 27356765 PMCID: PMC4928112 DOI: 10.1038/srep29228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/16/2016] [Indexed: 11/09/2022] Open
Abstract
The post-genomic era has resulted in the accumulation of high-throughput cancer data from a vast array of genomic technologies including next-generation sequencing and microarray. As such, the large amounts of germline variant and somatic mutation data that have been generated from GWAS and sequencing projects, respectively, show great promise in providing a systems-level view of these genetic aberrations. In this study, we analyze publicly available GWAS, somatic mutation, and drug target data derived from large databanks using a network-based approach that incorporates directed edge information under a randomized network hypothesis testing procedure. We show that these three classes of disease-associated nodes exhibit non-random topological characteristics in the context of a functional interactome. Specifically, we show that drug targets tend to lie upstream of somatic mutations and disease susceptibility germline variants. In addition, we introduce a new approach to measuring hierarchy between drug targets, somatic mutants, and disease susceptibility genes by utilizing directionality and path length information. Overall, our results provide new insight into the intrinsic relationships between these node classes that broaden our understanding of cancer. In addition, our results align with current knowledge on the therapeutic actionability of GWAS and somatic mutant nodes, while demonstrating relationships between node classes from a global network perspective.
Collapse
Affiliation(s)
- Matthew H Ung
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755 USA.,Program in Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03755 USA
| | - Chun-Chi Liu
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taiwan
| | - Chao Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755 USA.,Program in Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03755 USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03766 USA
| |
Collapse
|
25
|
Campbell J, Ryan CJ, Brough R, Bajrami I, Pemberton HN, Chong IY, Costa-Cabral S, Frankum J, Gulati A, Holme H, Miller R, Postel-Vinay S, Rafiq R, Wei W, Williamson CT, Quigley DA, Tym J, Al-Lazikani B, Fenton T, Natrajan R, Strauss SJ, Ashworth A, Lord CJ. Large-Scale Profiling of Kinase Dependencies in Cancer Cell Lines. Cell Rep 2016; 14:2490-501. [PMID: 26947069 PMCID: PMC4802229 DOI: 10.1016/j.celrep.2016.02.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/07/2015] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
Abstract
One approach to identifying cancer-specific vulnerabilities and therapeutic targets is to profile genetic dependencies in cancer cell lines. Here, we describe data from a series of siRNA screens that identify the kinase genetic dependencies in 117 cancer cell lines from ten cancer types. By integrating the siRNA screen data with molecular profiling data, including exome sequencing data, we show how vulnerabilities/genetic dependencies that are associated with mutations in specific cancer driver genes can be identified. By integrating additional data sets into this analysis, including protein-protein interaction data, we also demonstrate that the genetic dependencies associated with many cancer driver genes form dense connections on functional interaction networks. We demonstrate the utility of this resource by using it to predict the drug sensitivity of genetically or histologically defined subsets of tumor cell lines, including an increased sensitivity of osteosarcoma cell lines to FGFR inhibitors and SMAD4 mutant tumor cells to mitotic inhibitors.
Collapse
MESH Headings
- Cell Line, Tumor
- Gene Expression Profiling
- Humans
- Mutation
- Neoplasms/enzymology
- Neoplasms/genetics
- Neoplasms/pathology
- Protein Kinases/chemistry
- Protein Kinases/genetics
- Protein Kinases/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Smad4 Protein/antagonists & inhibitors
- Smad4 Protein/genetics
- Smad4 Protein/metabolism
Collapse
Affiliation(s)
- James Campbell
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Colm J Ryan
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Rachel Brough
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Ilirjana Bajrami
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Helen N Pemberton
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Irene Y Chong
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; Royal Marsden Hospital, London SW3 6JJ, UK
| | - Sara Costa-Cabral
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Jessica Frankum
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Aditi Gulati
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Holme
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Rowan Miller
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Sophie Postel-Vinay
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Rumana Rafiq
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Wenbin Wei
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Chris T Williamson
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - David A Quigley
- UCSF Helen Diller Family Comprehensive Cancer Centre, San Francisco, CA 94158, USA
| | - Joe Tym
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Bissan Al-Lazikani
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Timothy Fenton
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Rachael Natrajan
- Functional Genomics Laboratory, The Breast Cancer Now Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Sandra J Strauss
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alan Ashworth
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Christopher J Lord
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
26
|
Nguyen TTT, Chua JKK, Seah KS, Koo SH, Yee JY, Yang EG, Lim KK, Pang SYW, Yuen A, Zhang L, Ang WH, Dymock B, Lee EJD, Chen ES. Predicting chemotherapeutic drug combinations through gene network profiling. Sci Rep 2016; 6:18658. [PMID: 26791325 PMCID: PMC4726371 DOI: 10.1038/srep18658] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 11/23/2015] [Indexed: 12/29/2022] Open
Abstract
Contemporary chemotherapeutic treatments incorporate the use of several agents in combination. However, selecting the most appropriate drugs for such therapy is not necessarily an easy or straightforward task. Here, we describe a targeted approach that can facilitate the reliable selection of chemotherapeutic drug combinations through the interrogation of drug-resistance gene networks. Our method employed single-cell eukaryote fission yeast (Schizosaccharomyces pombe) as a model of proliferating cells to delineate a drug resistance gene network using a synthetic lethality workflow. Using the results of a previous unbiased screen, we assessed the genetic overlap of doxorubicin with six other drugs harboring varied mechanisms of action. Using this fission yeast model, drug-specific ontological sub-classifications were identified through the computation of relative hypersensitivities. We found that human gastric adenocarcinoma cells can be sensitized to doxorubicin by concomitant treatment with cisplatin, an intra-DNA strand crosslinking agent, and suberoylanilide hydroxamic acid, a histone deacetylase inhibitor. Our findings point to the utility of fission yeast as a model and the differential targeting of a conserved gene interaction network when screening for successful chemotherapeutic drug combinations for human cells.
Collapse
Affiliation(s)
- Thi Thuy Trang Nguyen
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore
| | - Jacqueline Kia Kee Chua
- Department of Biochemistry, National University of Singapore, Singapore.,Department of Chemistry, Faculty of Science, National University of Singapore, Singapore
| | - Kwi Shan Seah
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore
| | - Seok Hwee Koo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Changi General Hospital, Ministry of Health, Singapore
| | - Jie Yin Yee
- National University Health System (NUHS), Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Eugene Guorong Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Kim Kiat Lim
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore
| | | | - Audrey Yuen
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore
| | - Louxin Zhang
- Department of Mathematics, Faculty of Science, National University of Singapore, Singapore
| | - Wee Han Ang
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.,Department of Chemistry, Faculty of Science, National University of Singapore, Singapore
| | - Brian Dymock
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Edmund Jon Deoon Lee
- National University Health System (NUHS), Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ee Sin Chen
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
27
|
Linking Genes to Cardiovascular Diseases: Gene Action and Gene-Environment Interactions. J Cardiovasc Transl Res 2015; 8:506-27. [PMID: 26545598 DOI: 10.1007/s12265-015-9658-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/08/2015] [Indexed: 01/22/2023]
Abstract
A unique myocardial characteristic is its ability to grow/remodel in order to adapt; this is determined partly by genes and partly by the environment and the milieu intérieur. In the "post-genomic" era, a need is emerging to elucidate the physiologic functions of myocardial genes, as well as potential adaptive and maladaptive modulations induced by environmental/epigenetic factors. Genome sequencing and analysis advances have become exponential lately, with escalation of our knowledge concerning sometimes controversial genetic underpinnings of cardiovascular diseases. Current technologies can identify candidate genes variously involved in diverse normal/abnormal morphomechanical phenotypes, and offer insights into multiple genetic factors implicated in complex cardiovascular syndromes. The expression profiles of thousands of genes are regularly ascertained under diverse conditions. Global analyses of gene expression levels are useful for cataloging genes and correlated phenotypes, and for elucidating the role of genes in maladies. Comparative expression of gene networks coupled to complex disorders can contribute insights as to how "modifier genes" influence the expressed phenotypes. Increasingly, a more comprehensive and detailed systematic understanding of genetic abnormalities underlying, for example, various genetic cardiomyopathies is emerging. Implementing genomic findings in cardiology practice may well lead directly to better diagnosing and therapeutics. There is currently evolving a strong appreciation for the value of studying gene anomalies, and doing so in a non-disjointed, cohesive manner. However, it is challenging for many-practitioners and investigators-to comprehend, interpret, and utilize the clinically increasingly accessible and affordable cardiovascular genomics studies. This survey addresses the need for fundamental understanding in this vital area.
Collapse
|
28
|
Bourras S, McNally KE, Ben-David R, Parlange F, Roffler S, Praz CR, Oberhaensli S, Menardo F, Stirnweis D, Frenkel Z, Schaefer LK, Flückiger S, Treier G, Herren G, Korol AB, Wicker T, Keller B. Multiple Avirulence Loci and Allele-Specific Effector Recognition Control the Pm3 Race-Specific Resistance of Wheat to Powdery Mildew. THE PLANT CELL 2015; 27:2991-3012. [PMID: 26452600 PMCID: PMC4682313 DOI: 10.1105/tpc.15.00171] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 09/01/2015] [Accepted: 09/11/2015] [Indexed: 05/20/2023]
Abstract
In cereals, several mildew resistance genes occur as large allelic series; for example, in wheat (Triticum aestivum and Triticum turgidum), 17 functional Pm3 alleles confer agronomically important race-specific resistance to powdery mildew (Blumeria graminis). The molecular basis of race specificity has been characterized in wheat, but little is known about the corresponding avirulence genes in powdery mildew. Here, we dissected the genetics of avirulence for six Pm3 alleles and found that three major Avr loci affect avirulence, with a common locus_1 involved in all AvrPm3-Pm3 interactions. We cloned the effector gene AvrPm3(a2/f2) from locus_2, which is recognized by the Pm3a and Pm3f alleles. Induction of a Pm3 allele-dependent hypersensitive response in transient assays in Nicotiana benthamiana and in wheat demonstrated specificity. Gene expression analysis of Bcg1 (encoded by locus_1) and AvrPm3 (a2/f2) revealed significant differences between isolates, indicating that in addition to protein polymorphisms, expression levels play a role in avirulence. We propose a model for race specificity involving three components: an allele-specific avirulence effector, a resistance gene allele, and a pathogen-encoded suppressor of avirulence. Thus, whereas a genetically simple allelic series controls specificity in the plant host, recognition on the pathogen side is more complex, allowing flexible evolutionary responses and adaptation to resistance genes.
Collapse
Affiliation(s)
- Salim Bourras
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | | | - Roi Ben-David
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Francis Parlange
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Stefan Roffler
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | | | - Simone Oberhaensli
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Fabrizio Menardo
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Daniel Stirnweis
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Zeev Frenkel
- Institute of Evolution, University of Haifa, Mount Carmel, 31905 Haifa, Israel
| | | | - Simon Flückiger
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Georges Treier
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Gerhard Herren
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Abraham B Korol
- Institute of Evolution, University of Haifa, Mount Carmel, 31905 Haifa, Israel
| | - Thomas Wicker
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| | - Beat Keller
- Institute of Plant Biology, University of Zurich, CH-8008 Zürich, Switzerland
| |
Collapse
|
29
|
Boloc D, Castillo-Lara S, Marfany G, Gonzàlez-Duarte R, Abril JF. Distilling a Visual Network of Retinitis Pigmentosa Gene-Protein Interactions to Uncover New Disease Candidates. PLoS One 2015; 10:e0135307. [PMID: 26267445 PMCID: PMC4534355 DOI: 10.1371/journal.pone.0135307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 07/20/2015] [Indexed: 01/18/2023] Open
Abstract
Background Retinitis pigmentosa (RP) is a highly heterogeneous genetic visual disorder with more than 70 known causative genes, some of them shared with other non-syndromic retinal dystrophies (e.g. Leber congenital amaurosis, LCA). The identification of RP genes has increased steadily during the last decade, and the 30% of the cases that still remain unassigned will soon decrease after the advent of exome/genome sequencing. A considerable amount of genetic and functional data on single RD genes and mutations has been gathered, but a comprehensive view of the RP genes and their interacting partners is still very fragmentary. This is the main gap that needs to be filled in order to understand how mutations relate to progressive blinding disorders and devise effective therapies. Methodology We have built an RP-specific network (RPGeNet) by merging data from different sources: high-throughput data from BioGRID and STRING databases, manually curated data for interactions retrieved from iHOP, as well as interactions filtered out by syntactical parsing from up-to-date abstracts and full-text papers related to the RP research field. The paths emerging when known RP genes were used as baits over the whole interactome have been analysed, and the minimal number of connections among the RP genes and their close neighbors were distilled in order to simplify the search space. Conclusions In contrast to the analysis of single isolated genes, finding the networks linking disease genes renders powerful etiopathological insights. We here provide an interactive interface, RPGeNet, for the molecular biologist to explore the network centered on the non-syndromic and syndromic RP and LCA causative genes. By integrating tissue-specific expression levels and phenotypic data on top of that network, a more comprehensive biological view will highlight key molecular players of retinal degeneration and unveil new RP disease candidates.
Collapse
Affiliation(s)
- Daniel Boloc
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Sergio Castillo-Lara
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Marfany
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBERER, Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - Roser Gonzàlez-Duarte
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBERER, Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
- * E-mail: (JFA); (RGD)
| | - Josep F. Abril
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
- * E-mail: (JFA); (RGD)
| |
Collapse
|
30
|
Krogan NJ, Lippman S, Agard DA, Ashworth A, Ideker T. The cancer cell map initiative: defining the hallmark networks of cancer. Mol Cell 2015; 58:690-8. [PMID: 26000852 PMCID: PMC5359018 DOI: 10.1016/j.molcel.2015.05.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Progress in DNA sequencing has revealed the startling complexity of cancer genomes, which typically carry thousands of somatic mutations. However, it remains unclear which are the key driver mutations or dependencies in a given cancer and how these influence pathogenesis and response to therapy. Although tumors of similar types and clinical outcomes can have patterns of mutations that are strikingly different, it is becoming apparent that these mutations recurrently hijack the same hallmark molecular pathways and networks. For this reason, it is likely that successful interpretation of cancer genomes will require comprehensive knowledge of the molecular networks under selective pressure in oncogenesis. Here we announce the creation of a new effort, The Cancer Cell Map Initiative (CCMI), aimed at systematically detailing these complex interactions among cancer genes and how they differ between diseased and healthy states. We discuss recent progress that enables creation of these cancer cell maps across a range of tumor types and how they can be used to target networks disrupted in individual patients, significantly accelerating the development of precision medicine.
Collapse
Affiliation(s)
- Nevan J Krogan
- California Institute for Quantitative Biosciences (QB3), University of California, San Francisco, San Francisco, CA 94143, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA; J. David Gladstone Institutes, San Francisco, CA 94143, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Scott Lippman
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA; Moores Cancer Center, University of California, San Diego, San Diego, CA 92093, USA
| | - David A Agard
- California Institute for Quantitative Biosciences (QB3), University of California, San Francisco, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 92093, USA
| | - Alan Ashworth
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 92093, USA
| | - Trey Ideker
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA; Moores Cancer Center, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
31
|
Parikshak NN, Gandal MJ, Geschwind DH. Systems biology and gene networks in neurodevelopmental and neurodegenerative disorders. Nat Rev Genet 2015; 16:441-58. [PMID: 26149713 PMCID: PMC4699316 DOI: 10.1038/nrg3934] [Citation(s) in RCA: 287] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic and genomic approaches have implicated hundreds of genetic loci in neurodevelopmental disorders and neurodegeneration, but mechanistic understanding continues to lag behind the pace of gene discovery. Understanding the role of specific genetic variants in the brain involves dissecting a functional hierarchy that encompasses molecular pathways, diverse cell types, neural circuits and, ultimately, cognition and behaviour. With a focus on transcriptomics, this Review discusses how high-throughput molecular, integrative and network approaches inform disease biology by placing human genetics in a molecular systems and neurobiological context. We provide a framework for interpreting network biology studies and leveraging big genomics data sets in neurobiology.
Collapse
Affiliation(s)
- Neelroop N Parikshak
- 1] Program in Neurobehavioral Genetics, Semel Institute, and Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [2] Interdepartmental Program in Neuroscience, University of California, Los Angeles, California 90095, USA
| | - Michael J Gandal
- 1] Program in Neurobehavioral Genetics, Semel Institute, and Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [2] Center for Autism Treatment and Research, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | - Daniel H Geschwind
- 1] Program in Neurobehavioral Genetics, Semel Institute, and Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [2] Interdepartmental Program in Neuroscience, University of California, Los Angeles, California 90095, USA. [3] Center for Autism Treatment and Research, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [4] Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
32
|
Bennett L, Kittas A, Muirhead G, Papageorgiou LG, Tsoka S. Detection of composite communities in multiplex biological networks. Sci Rep 2015; 5:10345. [PMID: 26012716 PMCID: PMC4446847 DOI: 10.1038/srep10345] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/26/2015] [Indexed: 12/23/2022] Open
Abstract
The detection of community structure is a widely accepted means of investigating the
principles governing biological systems. Recent efforts are exploring ways in which
multiple data sources can be integrated to generate a more comprehensive model of
cellular interactions, leading to the detection of more biologically relevant
communities. In this work, we propose a mathematical programming model to cluster
multiplex biological networks, i.e. multiple network slices, each with a different
interaction type, to determine a single representative partition of composite
communities. Our method, known as SimMod, is evaluated through its application to
yeast networks of physical, genetic and co-expression interactions. A comparative
analysis involving partitions of the individual networks, partitions of aggregated
networks and partitions generated by similar methods from the literature highlights
the ability of SimMod to identify functionally enriched modules. It is further shown
that SimMod offers enhanced results when compared to existing approaches without the
need to train on known cellular interactions.
Collapse
Affiliation(s)
- Laura Bennett
- Centre for Process Systems Engineering, Department of Chemical Engineering,University College London, Torrington Place, London WC1E 7JE, United Kingdom
| | - Aristotelis Kittas
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Strand, London WC2R 2LS, UnitedKingdom
| | - Gareth Muirhead
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Strand, London WC2R 2LS, UnitedKingdom
| | - Lazaros G Papageorgiou
- Centre for Process Systems Engineering, Department of Chemical Engineering,University College London, Torrington Place, London WC1E 7JE, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Strand, London WC2R 2LS, UnitedKingdom
| |
Collapse
|
33
|
Wang H, Cai H, Ao L, Yan H, Zhao W, Qi L, Gu Y, Guo Z. Individualized identification of disease-associated pathways with disrupted coordination of gene expression. Brief Bioinform 2015; 17:78-87. [PMID: 26023086 DOI: 10.1093/bib/bbv030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 01/08/2023] Open
Abstract
Current pathway analysis approaches are primarily dedicated to capturing deregulated pathways at the population level and cannot provide patient-specific pathway deregulation information. In this article, the authors present a simple approach, called individPath, to detect pathways with significantly disrupted intra-pathway relative expression orderings for each disease sample compared with the stable, normal intra-pathway relative expression orderings pre-determined in previously accumulated normal samples. Through the analysis of multiple microarray data sets for lung and breast cancer, the authors demonstrate individPath's effectiveness for detecting cancer-associated pathways with disrupted relative expression orderings at the individual level and dissecting the heterogeneity of pathway deregulation among different patients. The portable use of this simple approach in clinical contexts is exemplified by the identification of prognostic intra-pathway gene pair signatures to predict overall survival of resected early-stage lung adenocarcinoma patients and signatures to predict relapse-free survival of estrogen receptor-positive breast cancer patients after tamoxifen treatment.
Collapse
|
34
|
Rangarajan N, Kulkarni P, Hannenhalli S. Evolutionarily conserved network properties of intrinsically disordered proteins. PLoS One 2015; 10:e0126729. [PMID: 25974317 PMCID: PMC4431869 DOI: 10.1371/journal.pone.0126729] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 04/07/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Intrinsically disordered proteins (IDPs) lack a stable tertiary structure in isolation. Remarkably, however, a substantial portion of IDPs undergo disorder-to-order transitions upon binding to their cognate partners. Structural flexibility and binding plasticity enable IDPs to interact with a broad range of partners. However, the broader network properties that could provide additional insights into the functional role of IDPs are not known. RESULTS Here, we report the first comprehensive survey of network properties of IDP-induced sub-networks in multiple species from yeast to human. Our results show that IDPs exhibit greater-than-expected modularity and are connected to the rest of the protein interaction network (PIN) via proteins that exhibit the highest betweenness centrality and connect to fewer-than-expected IDP communities, suggesting that they form critical communication links from IDP modules to the rest of the PIN. Moreover, we found that IDPs are enriched at the top level of regulatory hierarchy. CONCLUSION Overall, our analyses reveal coherent and remarkably conserved IDP-centric network properties, namely, modularity in IDP-induced network and a layer of critical nodes connecting IDPs with the rest of the PIN.
Collapse
Affiliation(s)
| | - Prakash Kulkarni
- Institute for Bioscience & Biotechnology Research, University of Maryland, Rockville, Maryland, United States of America
| | - Sridhar Hannenhalli
- Center for Bioinformatics and Computational Biology, Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
35
|
Chen JJ, Wang Y. Microsatellite Development and Potential Application in Authentication, Conservation, and Genetic Improvement of Chinese Medicinal Plants. CHINESE HERBAL MEDICINES 2015. [DOI: 10.1016/s1674-6384(15)60029-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
36
|
Goldfarb D, Hast BE, Wang W, Major MB. Spotlite: web application and augmented algorithms for predicting co-complexed proteins from affinity purification--mass spectrometry data. J Proteome Res 2014; 13:5944-55. [PMID: 25300367 DOI: 10.1021/pr5008416] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protein-protein interactions defined by affinity purification and mass spectrometry (APMS) suffer from high false discovery rates. Consequently, lists of potential interactions must be pruned of contaminants before network construction and interpretation, historically an expensive, time-intensive, and error-prone task. In recent years, numerous computational methods were developed to identify genuine interactions from the hundreds of candidates. Here, comparative analysis of three popular algorithms, HGSCore, CompPASS, and SAINT, revealed complementarity in their classification accuracies, which is supported by their divergent scoring strategies. We improved each algorithm by an average area under a receiver operating characteristics curve increase of 16% by integrating a variety of indirect data known to correlate with established protein-protein interactions, including mRNA coexpression, gene ontologies, domain-domain binding affinities, and homologous protein interactions. Each APMS scoring approach was incorporated into a separate logistic regression model along with the indirect features; the resulting three classifiers demonstrate improved performance on five diverse APMS data sets. To facilitate APMS data scoring within the scientific community, we created Spotlite, a user-friendly and fast web application. Within Spotlite, data can be scored with the augmented classifiers, annotated, and visualized ( http://cancer.unc.edu/majorlab/software.php ). The utility of the Spotlite platform to reveal physical, functional, and disease-relevant characteristics within APMS data is established through a focused analysis of the KEAP1 E3 ubiquitin ligase.
Collapse
Affiliation(s)
- Dennis Goldfarb
- Department of Computer Science, University of North Carolina at Chapel Hill , Box #3175, Chapel Hill, North Carolina 27599, United States
| | | | | | | |
Collapse
|
37
|
Sedaghat N, Saegusa T, Randolph T, Shojaie A. Comparative study of computational methods for reconstructing genetic networks of cancer-related pathways. Cancer Inform 2014; 13:55-66. [PMID: 25288880 PMCID: PMC4179645 DOI: 10.4137/cin.s13781] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/08/2014] [Accepted: 05/10/2014] [Indexed: 12/16/2022] Open
Abstract
Network reconstruction is an important yet challenging task in systems biology. While many methods have been recently proposed for reconstructing biological networks from diverse data types, properties of estimated networks and differences between reconstruction methods are not well understood. In this paper, we conduct a comprehensive empirical evaluation of seven existing network reconstruction methods, by comparing the estimated networks with different sparsity levels for both normal and tumor samples. The results suggest substantial heterogeneity in networks reconstructed using different reconstruction methods. Our findings also provide evidence for significant differences between networks of normal and tumor samples, even after accounting for the considerable variability in structures of networks estimated using different reconstruction methods. These differences can offer new insight into changes in mechanisms of genetic interaction associated with cancer initiation and progression.
Collapse
Affiliation(s)
- Nafiseh Sedaghat
- Computer Engineering Department, Iran University of Science and Technology, Tehran, Iran
| | - Takumi Saegusa
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Timothy Randolph
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| |
Collapse
|
38
|
Abstract
The challenging task of studying and modeling complex dynamics of biological systems in order to describe various human diseases has gathered great interest in recent years. Major biological processes are mediated through protein interactions, hence there is a need to understand the chaotic network that forms these processes in pursuance of understanding human diseases. The applications of protein interaction networks to disease datasets allow the identification of genes and proteins associated with diseases, the study of network properties, identification of subnetworks, and network-based disease gene classification. Although various protein interaction network analysis strategies have been employed, grand challenges are still existing. Global understanding of protein interaction networks via integration of high-throughput functional genomics data from different levels will allow researchers to examine the disease pathways and identify strategies to control them. As a result, it seems likely that more personalized, more accurate and more rapid disease gene diagnostic techniques will be devised in the future, as well as novel strategies that are more personalized. This mini-review summarizes the current practice of protein interaction networks in medical research as well as challenges to be overcome.
Collapse
Affiliation(s)
- Tuba Sevimoglu
- Department of Bioengineering, Marmara University, Goztepe, 34722 Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, Goztepe, 34722 Istanbul, Turkey
| |
Collapse
|
39
|
Kim Y, Jang JH, Choi S, Hwang D. TEMPI: probabilistic modeling time-evolving differential PPI networks with multiPle information. Bioinformatics 2014; 30:i453-60. [PMID: 25161233 PMCID: PMC4147907 DOI: 10.1093/bioinformatics/btu454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Motivation: Time-evolving differential protein–protein interaction (PPI) networks are essential to understand serial activation of differentially regulated (up- or downregulated) cellular processes (DRPs) and their interplays over time. Despite developments in the network inference, current methods are still limited in identifying temporal transition of structures of PPI networks, DRPs associated with the structural transition and the interplays among the DRPs over time. Results: Here, we present a probabilistic model for estimating Time-Evolving differential PPI networks with MultiPle Information (TEMPI). This model describes probabilistic relationships among network structures, time-course gene expression data and Gene Ontology biological processes (GOBPs). By maximizing the likelihood of the probabilistic model, TEMPI estimates jointly the time-evolving differential PPI networks (TDNs) describing temporal transition of PPI network structures together with serial activation of DRPs associated with transiting networks. This joint estimation enables us to interpret the TDNs in terms of temporal transition of the DRPs. To demonstrate the utility of TEMPI, we applied it to two time-course datasets. TEMPI identified the TDNs that correctly delineated temporal transition of DRPs and time-dependent associations between the DRPs. These TDNs provide hypotheses for mechanisms underlying serial activation of key DRPs and their temporal associations. Availability and implementation: Source code and sample data files are available at http://sbm.postech.ac.kr/tempi/sources.zip. Contact:seungjin@postech.ac.kr or dhwang@dgist.ac.kr Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yongsoo Kim
- School of Interdisciplinary Bioscience and Bioengineering and Department of Computer Science and Engineering, Pohang University of Science and Technology, Pohang 790-784, Korea and Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu 711-873, Korea
| | - Jin-Hyeok Jang
- School of Interdisciplinary Bioscience and Bioengineering and Department of Computer Science and Engineering, Pohang University of Science and Technology, Pohang 790-784, Korea and Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu 711-873, Korea
| | - Seungjin Choi
- School of Interdisciplinary Bioscience and Bioengineering and Department of Computer Science and Engineering, Pohang University of Science and Technology, Pohang 790-784, Korea and Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu 711-873, Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering and Department of Computer Science and Engineering, Pohang University of Science and Technology, Pohang 790-784, Korea and Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu 711-873, Korea School of Interdisciplinary Bioscience and Bioengineering and Department of Computer Science and Engineering, Pohang University of Science and Technology, Pohang 790-784, Korea and Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu 711-873, Korea
| |
Collapse
|
40
|
Prediction of dynamical drug sensitivity and resistance by module network rewiring-analysis based on transcriptional profiling. Drug Resist Updat 2014; 17:64-76. [PMID: 25156319 DOI: 10.1016/j.drup.2014.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Revealing functional reorganization or module rewiring between modules at network levels during drug treatment is important to systematically understand therapies and drug responses. The present article proposed a novel model of module network rewiring to characterize functional reorganization of a complex biological system, and described a new framework named as module network rewiring-analysis (MNR) for systematically studying dynamical drug sensitivity and resistance during drug treatment. MNR was used to investigate functional reorganization or rewiring on the module network, rather than molecular network or individual molecules. Our experiments on expression data of patients with Hepatitis C virus infection receiving Interferon therapy demonstrated that consistent module genes derived by MNR could be directly used to reveal new genotypes relevant to drug sensitivity, unlike the other differential analyses of gene expressions. Our results showed that functional connections and reconnections among consistent modules bridged by biological paths were necessary for achieving effective responses of a drug. The hierarchical structures of the temporal module network can be considered as spatio-temporal biomarkers to monitor the efficacy, efficiency, toxicity, and resistance of the therapy. Our study indicates that MNR is a useful tool to identify module biomarkers and further predict dynamical drug sensitivity and resistance, characterize complex dynamic processes for therapy response, and provide biologically systematic clues for pharmacogenomic applications.
Collapse
|
41
|
Deciphering early development of complex diseases by progressive module network. Methods 2014; 67:334-43. [DOI: 10.1016/j.ymeth.2014.01.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/20/2014] [Accepted: 01/23/2014] [Indexed: 11/23/2022] Open
|
42
|
Selection of higher order regression models in the analysis of multi-factorial transcription data. PLoS One 2014; 9:e91840. [PMID: 24658540 PMCID: PMC3962375 DOI: 10.1371/journal.pone.0091840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 02/16/2014] [Indexed: 11/19/2022] Open
Abstract
Introduction Many studies examine gene expression data that has been obtained under the influence of multiple factors, such as genetic background, environmental conditions, or exposure to diseases. The interplay of multiple factors may lead to effect modification and confounding. Higher order linear regression models can account for these effects. We present a new methodology for linear model selection and apply it to microarray data of bone marrow-derived macrophages. This experiment investigates the influence of three variable factors: the genetic background of the mice from which the macrophages were obtained, Yersinia enterocolitica infection (two strains, and a mock control), and treatment/non-treatment with interferon-γ. Results We set up four different linear regression models in a hierarchical order. We introduce the eruption plot as a new practical tool for model selection complementary to global testing. It visually compares the size and significance of effect estimates between two nested models. Using this methodology we were able to select the most appropriate model by keeping only relevant factors showing additional explanatory power. Application to experimental data allowed us to qualify the interaction of factors as either neutral (no interaction), alleviating (co-occurring effects are weaker than expected from the single effects), or aggravating (stronger than expected). We find a biologically meaningful gene cluster of putative C2TA target genes that appear to be co-regulated with MHC class II genes. Conclusions We introduced the eruption plot as a tool for visual model comparison to identify relevant higher order interactions in the analysis of expression data obtained under the influence of multiple factors. We conclude that model selection in higher order linear regression models should generally be performed for the analysis of multi-factorial microarray data.
Collapse
|
43
|
Canuel V, Rance B, Avillach P, Degoulet P, Burgun A. Translational research platforms integrating clinical and omics data: a review of publicly available solutions. Brief Bioinform 2014; 16:280-90. [PMID: 24608524 PMCID: PMC4364065 DOI: 10.1093/bib/bbu006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The rise of personalized medicine and the availability of high-throughput molecular analyses in the context of clinical care have increased the need for adequate tools for translational researchers to manage and explore these data. We reviewed the biomedical literature for translational platforms allowing the management and exploration of clinical and omics data, and identified seven platforms: BRISK, caTRIP, cBio Cancer Portal, G-DOC, iCOD, iDASH and tranSMART. We analyzed these platforms along seven major axes. (1) The community axis regrouped information regarding initiators and funders of the project, as well as availability status and references. (2) We regrouped under the information content axis the nature of the clinical and omics data handled by each system. (3) The privacy management environment axis encompassed functionalities allowing control over data privacy. (4) In the analysis support axis, we detailed the analytical and statistical tools provided by the platforms. We also explored (5) interoperability support and (6) system requirements. The final axis (7) platform support listed the availability of documentation and installation procedures. A large heterogeneity was observed in regard to the capability to manage phenotype information in addition to omics data, their security and interoperability features. The analytical and visualization features strongly depend on the considered platform. Similarly, the availability of the systems is variable. This review aims at providing the reader with the background to choose the platform best suited to their needs. To conclude, we discuss the desiderata for optimal translational research platforms, in terms of privacy, interoperability and technical features.
Collapse
|
44
|
Mu Y, Cai P, Hu S, Ma S, Gao Y. Characterization of diverse internal binding specificities of PDZ domains by yeast two-hybrid screening of a special peptide library. PLoS One 2014; 9:e88286. [PMID: 24505465 PMCID: PMC3913781 DOI: 10.1371/journal.pone.0088286] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/09/2014] [Indexed: 01/07/2023] Open
Abstract
Protein-protein interactions (PPIs) are essential events to play important roles in a series of biological processes. There are probably more ways of PPIs than we currently realized. Structural and functional investigations of weak PPIs have lagged behind those of strong PPIs due to technical difficulties. Weak PPIs are often short-lived, which may result in more dynamic signals with important biological roles within and/or between cells. For example, the characteristics of PSD-95/Dlg/ZO-1 (PDZ) domain binding to internal sequences, which are primarily weak interactions, have not yet been systematically explored. In the present study, we constructed a nearly random octapeptide yeast two-hybrid library. A total of 24 PDZ domains were used as baits for screening the library. Fourteen of these domains were able to bind internal PDZ-domain binding motifs (PBMs), and PBMs screened for nine PDZ domains exhibited strong preferences. Among 11 PDZ domains that have not been reported their internal PBM binding ability, six were confirmed to bind internal PBMs. The first PDZ domain of LNX2, which has not been reported to bind C-terminal PBMs, was found to bind internal PBMs. These results suggest that the internal PBMs binding ability of PDZ domains may have been underestimated. The data provided diverse internal binding properties for several PDZ domains that may help identify their novel binding partners.
Collapse
Affiliation(s)
- Yi Mu
- National Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, P.R. China
| | - Pengfei Cai
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Siqi Hu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Sucan Ma
- National Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, P.R. China
| | - Youhe Gao
- National Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, P.R. China
- * E-mail:
| |
Collapse
|
45
|
Yang TH, Wu WS. Inferring functional transcription factor-gene binding pairs by integrating transcription factor binding data with transcription factor knockout data. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 6:S13. [PMID: 24565265 PMCID: PMC4029220 DOI: 10.1186/1752-0509-7-s6-s13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Chromatin immunoprecipitation (ChIP) experiments are now the most comprehensive experimental approaches for mapping the binding of transcription factors (TFs) to their target genes. However, ChIP data alone is insufficient for identifying functional binding target genes of TFs for two reasons. First, there is an inherent high false positive/negative rate in ChIP-chip or ChIP-seq experiments. Second, binding signals in the ChIP data do not necessarily imply functionality. Methods It is known that ChIP-chip data and TF knockout (TFKO) data reveal complementary information on gene regulation. While ChIP-chip data can provide TF-gene binding pairs, TFKO data can provide TF-gene regulation pairs. Therefore, we propose a novel network approach for identifying functional TF-gene binding pairs by integrating the ChIP-chip data with the TFKO data. In our method, a TF-gene binding pair from the ChIP-chip data is regarded to be functional if it also has high confident curated TFKO TF-gene regulatory relation or deduced hypostatic TF-gene regulatory relation. Results and conclusions We first validated our method on a gathered ground truth set. Then we applied our method to the ChIP-chip data to identify functional TF-gene binding pairs. The biological significance of our identified functional TF-gene binding pairs was shown by assessing their functional enrichment, the prevalence of protein-protein interaction, and expression coherence. Our results outperformed the results of three existing methods across all measures. And our identified functional targets of TFs also showed statistical significance over the randomly assigned TF-gene pairs. We also showed that our method is dataset independent and can apply to ChIP-seq data and the E. coli genome. Finally, we provided an example showing the biological applicability of our notion.
Collapse
|
46
|
Integrative approaches for finding modular structure in biological networks. Nat Rev Genet 2013; 14:719-32. [PMID: 24045689 DOI: 10.1038/nrg3552] [Citation(s) in RCA: 351] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A central goal of systems biology is to elucidate the structural and functional architecture of the cell. To this end, large and complex networks of molecular interactions are being rapidly generated for humans and model organisms. A recent focus of bioinformatics research has been to integrate these networks with each other and with diverse molecular profiles to identify sets of molecules and interactions that participate in a common biological function - that is, 'modules'. Here, we classify such integrative approaches into four broad categories, describe their bioinformatic principles and review their applications.
Collapse
|
47
|
Ostojić J, Glatigny A, Herbert CJ, Dujardin G, Bonnefoy N. Does the study of genetic interactions help predict the function of mitochondrial proteins in Saccharomyces cerevisiae? Biochimie 2013; 100:27-37. [PMID: 24262604 DOI: 10.1016/j.biochi.2013.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 11/06/2013] [Indexed: 10/26/2022]
Abstract
Mitochondria are complex organelles of eukaryotic cells that contain their own genome, encoding key subunits of the respiratory complexes. The successive steps of mitochondrial gene expression are intimately linked, and are under the control of a large number of nuclear genes encoding factors that are imported into mitochondria. Investigating the relationships between these genes and their interaction networks, and whether they reveal direct or indirect partners, can shed light on their role in mitochondrial biogenesis, as well as identify new actors in this process. These studies, mainly developed in yeasts, are significant because mammalian equivalents of such yeast genes are candidate genes in mitochondrial pathologies. In practice, studies of physical, chemical and genetic interactions can be undertaken. The search for genetic interactions, either aggravating or alleviating the phenotype of the starting mutants, has proved to be particularly powerful in yeast since even subtle changes in respiratory phenotypes can be screened in a very efficient way. In addition, several high throughput genetic approaches have recently been developed. In this review we analyze the genetic network of three genes involved in different steps of mitochondrial gene expression, from the transcription and translation of mitochondrial RNAs to the insertion of newly synthesized proteins into the inner mitochondrial membrane, and we examine their relevance to our understanding of mitochondrial biogenesis. We find that these genetic interactions are seldom redundant with physical interactions, and thus bring a considerable amount of original and significant information as well as open new areas of research.
Collapse
Affiliation(s)
- Jelena Ostojić
- Centre de Génétique Moléculaire, CNRS UPR3404 Associated to the University Paris XI-Sud, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Annie Glatigny
- Centre de Génétique Moléculaire, CNRS UPR3404 Associated to the University Paris XI-Sud, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Christopher J Herbert
- Centre de Génétique Moléculaire, CNRS UPR3404 Associated to the University Paris XI-Sud, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Geneviève Dujardin
- Centre de Génétique Moléculaire, CNRS UPR3404 Associated to the University Paris XI-Sud, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Nathalie Bonnefoy
- Centre de Génétique Moléculaire, CNRS UPR3404 Associated to the University Paris XI-Sud, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France.
| |
Collapse
|
48
|
Abstract
Proteins are not monolithic entities; rather, they can contain multiple domains that mediate distinct interactions, and their functionality can be regulated through post-translational modifications at multiple distinct sites. Traditionally, network biology has ignored such properties of proteins and has instead examined either the physical interactions of whole proteins or the consequences of removing entire genes. In this Review, we discuss experimental and computational methods to increase the resolution of protein-protein, genetic and drug-gene interaction studies to the domain and residue levels. Such work will be crucial for using interaction networks to connect sequence and structural information, and to understand the biological consequences of disease-associated mutations, which will hopefully lead to more effective therapeutic strategies.
Collapse
|
49
|
Identifying genes relevant to specific biological conditions in time course microarray experiments. PLoS One 2013; 8:e76561. [PMID: 24146889 PMCID: PMC3795718 DOI: 10.1371/journal.pone.0076561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/28/2013] [Indexed: 11/19/2022] Open
Abstract
Microarrays have been useful in understanding various biological processes by allowing the simultaneous study of the expression of thousands of genes. However, the analysis of microarray data is a challenging task. One of the key problems in microarray analysis is the classification of unknown expression profiles. Specifically, the often large number of non-informative genes on the microarray adversely affects the performance and efficiency of classification algorithms. Furthermore, the skewed ratio of sample to variable poses a risk of overfitting. Thus, in this context, feature selection methods become crucial to select relevant genes and, hence, improve classification accuracy. In this study, we investigated feature selection methods based on gene expression profiles and protein interactions. We found that in our setup, the addition of protein interaction information did not contribute to any significant improvement of the classification results. Furthermore, we developed a novel feature selection method that relies exclusively on observed gene expression changes in microarray experiments, which we call "relative Signal-to-Noise ratio" (rSNR). More precisely, the rSNR ranks genes based on their specificity to an experimental condition, by comparing intrinsic variation, i.e. variation in gene expression within an experimental condition, with extrinsic variation, i.e. variation in gene expression across experimental conditions. Genes with low variation within an experimental condition of interest and high variation across experimental conditions are ranked higher, and help in improving classification accuracy. We compared different feature selection methods on two time-series microarray datasets and one static microarray dataset. We found that the rSNR performed generally better than the other methods.
Collapse
|
50
|
Shimizu H, Kubo A, Uchibe K, Hashimoto M, Yokoyama S, Takada S, Mitsuoka K, Asahara H. The AERO system: a 3D-like approach for recording gene expression patterns in the whole mouse embryo. PLoS One 2013; 8:e75754. [PMID: 24146773 PMCID: PMC3797748 DOI: 10.1371/journal.pone.0075754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 08/20/2013] [Indexed: 12/30/2022] Open
Abstract
We have recently constructed a web-based database of gene expression in the mouse whole embryo, EMBRYS (http://embrys.jp/embrys/html/MainMenu.html). To allow examination of gene expression patterns to the fullest extent possible, this database provides both photo images and annotation data. However, since embryos develop via an intricate process of morphogenesis, it would be of great value to track embryonic gene expression from a three dimensional perspective. In fact, several methods have been developed to achieve this goal, but highly laborious procedures and specific operational skills are generally required. We utilized a novel microscopic technique that enables the easy capture of rotational, 3D-like images of the whole embryo. In this method, a rotary head equipped with two mirrors that are designed to obtain an image tilted at 45 degrees to the microscope stage captures serial images at 2-degree intervals. By a simple operation, 180 images are automatically collected. These 2D images obtained at multiple angles are then used to reconstruct 3D-like images, termed AERO images. By means of this system, over 800 AERO images of 191 gene expression patterns were captured. These images can be easily rotated on the computer screen using the EMBRYS database so that researchers can view an entire embryo by a virtual viewing on a computer screen in an unbiased or non-predetermined manner. The advantages afforded by this approach make it especially useful for generating data viewed in public databases.
Collapse
Affiliation(s)
- Hirohito Shimizu
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Atsushi Kubo
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenta Uchibe
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Megumi Hashimoto
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Shigetoshi Yokoyama
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Shuji Takada
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kazuhiko Mitsuoka
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hiroshi Asahara
- Department of Systems Biomedicine, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|