1
|
del Giudice G, Serra A, Pavel A, Torres Maia M, Saarimäki LA, Fratello M, Federico A, Alenius H, Fadeel B, Greco D. A Network Toxicology Approach for Mechanistic Modelling of Nanomaterial Hazard and Adverse Outcomes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400389. [PMID: 38923832 PMCID: PMC11348149 DOI: 10.1002/advs.202400389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/10/2024] [Indexed: 06/28/2024]
Abstract
Hazard assessment is the first step in evaluating the potential adverse effects of chemicals. Traditionally, toxicological assessment has focused on the exposure, overlooking the impact of the exposed system on the observed toxicity. However, systems toxicology emphasizes how system properties significantly contribute to the observed response. Hence, systems theory states that interactions store more information than individual elements, leading to the adoption of network based models to represent complex systems in many fields of life sciences. Here, they develop a network-based approach to characterize toxicological responses in the context of a biological system, inferring biological system specific networks. They directly link molecular alterations to the adverse outcome pathway (AOP) framework, establishing direct connections between omics data and toxicologically relevant phenotypic events. They apply this framework to a dataset including 31 engineered nanomaterials with different physicochemical properties in two different in vitro and one in vivo models and demonstrate how the biological system is the driving force of the observed response. This work highlights the potential of network-based methods to significantly improve their understanding of toxicological mechanisms from a systems biology perspective and provides relevant considerations and future data-driven approaches for the hazard assessment of nanomaterials and other advanced materials.
Collapse
Affiliation(s)
- Giusy del Giudice
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinki00790Finland
| | - Angela Serra
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinki00790Finland
- Tampere Institute for Advanced StudyTampere UniversityTampere33100Finland
| | - Alisa Pavel
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
| | - Marcella Torres Maia
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
| | - Laura Aliisa Saarimäki
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinki00790Finland
| | - Michele Fratello
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
| | - Antonio Federico
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinki00790Finland
- Tampere Institute for Advanced StudyTampere UniversityTampere33100Finland
| | - Harri Alenius
- Human Microbiome Research Program (HUMI)University of HelsinkiHelsinki00014Finland
- Institute of Environmental MedicineKarolinska InstitutetStockholm171 77Sweden
| | - Bengt Fadeel
- Institute of Environmental MedicineKarolinska InstitutetStockholm171 77Sweden
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinki00790Finland
- Tampere Institute for Advanced StudyTampere UniversityTampere33100Finland
- Institute of BiotechnologyUniversity of HelsinkiHelsinki00790Finland
| |
Collapse
|
2
|
Wei L, Xin Y, Pu M, Zhang Y. Patient-specific analysis of co-expression to measure biological network rewiring in individuals. Life Sci Alliance 2024; 7:e202302253. [PMID: 37977656 PMCID: PMC10656351 DOI: 10.26508/lsa.202302253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
To effectively understand the underlying mechanisms of disease and inform the development of personalized therapies, it is critical to harness the power of differential co-expression (DCE) network analysis. Despite the promise of DCE network analysis in precision medicine, current approaches have a major limitation: they measure an average differential network across multiple samples, which means the specific etiology of individual patients is often overlooked. To address this, we present Cosinet, a DCE-based single-sample network rewiring degree quantification tool. By analyzing two breast cancer datasets, we demonstrate that Cosinet can identify important differences in gene co-expression patterns between individual patients and generate scores for each individual that are significantly associated with overall survival, recurrence-free interval, and other clinical outcomes, even after adjusting for risk factors such as age, tumor size, HER2 status, and PAM50 subtypes. Cosinet represents a remarkable development toward unlocking the potential of DCE analysis in the context of precision medicine.
Collapse
Affiliation(s)
- Lanying Wei
- Beijing StoneWise Technology Co Ltd, Danling SOHO, Beijing, China
| | - Yucui Xin
- Beijing StoneWise Technology Co Ltd, Danling SOHO, Beijing, China
| | - Mengchen Pu
- Beijing StoneWise Technology Co Ltd, Danling SOHO, Beijing, China
| | - Yingsheng Zhang
- Beijing StoneWise Technology Co Ltd, Danling SOHO, Beijing, China
| |
Collapse
|
3
|
Chen Y, Zhang H, Sun X. Improving the performance of single-cell RNA-seq data mining based on relative expression orderings. Brief Bioinform 2022; 24:6931720. [PMID: 36528803 PMCID: PMC9851298 DOI: 10.1093/bib/bbac556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
The advent of single-cell RNA-sequencing (scRNA-seq) provides an unprecedented opportunity to explore gene expression profiles at the single-cell level. However, gene expression values vary over time and under different conditions even within the same cell. There is an urgent need for more stable and reliable feature variables at the single-cell level to depict cell heterogeneity. Thus, we construct a new feature matrix called the delta rank matrix (DRM) from scRNA-seq data by integrating an a priori gene interaction network, which transforms the unreliable gene expression value into a stable gene interaction/edge value on a single-cell basis. This is the first time that a gene-level feature has been transformed into an interaction/edge-level for scRNA-seq data analysis based on relative expression orderings. Experiments on various scRNA-seq datasets have demonstrated that DRM performs better than the original gene expression matrix in cell clustering, cell identification and pseudo-trajectory reconstruction. More importantly, the DRM really achieves the fusion of gene expressions and gene interactions and provides a method of measuring gene interactions at the single-cell level. Thus, the DRM can be used to find changes in gene interactions among different cell types, which may open up a new way to analyze scRNA-seq data from an interaction perspective. In addition, DRM provides a new method to construct a cell-specific network for each single cell instead of a group of cells as in traditional network construction methods. DRM's exceptional performance is due to its extraction of rich gene-association information on biological systems and stable characterization of cells.
Collapse
Affiliation(s)
- Yuanyuan Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China,College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Hao Zhang
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiao Sun
- Corresponding author: Xiao Sun, State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China. Tel: +8613951989906; E-mail:
| |
Collapse
|
4
|
Wang L, Xie W, Li K, Wang Z, Li X, Feng W, Li J. DysPIA: A Novel Dysregulated Pathway Identification Analysis Method. Front Genet 2021; 12:647653. [PMID: 34290733 PMCID: PMC8287415 DOI: 10.3389/fgene.2021.647653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
Differential co-expression-based pathway analysis is still limited and not widely used. In most current methods, the pathways were considered as gene sets, but the gene regulation relationships were not considered, and the computational speed was slow. In this article, we proposed a novel Dysregulated Pathway Identification Analysis (DysPIA) method to overcome these shortcomings. We adopted the idea of Correlation by Individual Level Product into analysis and performed a fast enrichment analysis. We constructed a combined gene-pair background which was much more sufficient than the background used in Edge Set Enrichment Analysis. In simulation study, DysPIA was able to identify the causal pathways with high AUC (0.9584 to 0.9896). In p53 mutation data, DysPIA obtained better performance than other methods. It obtained more potential dysregulated pathways that could be literature verified, and it ran much faster (∼1,700-8,000 times faster than other methods when 10,000 permutations). DysPIA was also applied to breast cancer relapse dataset and breast cancer subtype dataset. The results show that DysPIA is effective and has a great biological significance. R packages "DysPIA" and "DysPIAData" are constructed and freely available on R CRAN (https://cran.r-project.org/web/packages/DysPIA/index.html and https://cran.r-project.org/web/packages/DysPIAData/index.html), and on GitHub (https://github.com/lemonwang2020).
Collapse
Affiliation(s)
- Limei Wang
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China.,Key Laboratory of Tropical Translational Medicine, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Weixin Xie
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Kongning Li
- Key Laboratory of Tropical Translational Medicine, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Zhenzhen Wang
- Key Laboratory of Tropical Translational Medicine, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Weixing Feng
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Jin Li
- Key Laboratory of Tropical Translational Medicine, Ministry of Education, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Chen J, Zhang J, Gao Y, Li Y, Feng C, Song C, Ning Z, Zhou X, Zhao J, Feng M, Zhang Y, Wei L, Pan Q, Jiang Y, Qian F, Han J, Yang Y, Wang Q, Li C. LncSEA: a platform for long non-coding RNA related sets and enrichment analysis. Nucleic Acids Res 2021; 49:D969-D980. [PMID: 33045741 PMCID: PMC7778898 DOI: 10.1093/nar/gkaa806] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 02/01/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been proven to play important roles in transcriptional processes and various biological functions. Establishing a comprehensive collection of human lncRNA sets is urgent work at present. Using reference lncRNA sets, enrichment analyses will be useful for analyzing lncRNA lists of interest submitted by users. Therefore, we developed a human lncRNA sets database, called LncSEA, which aimed to document a large number of available resources for human lncRNA sets and provide annotation and enrichment analyses for lncRNAs. LncSEA supports >40 000 lncRNA reference sets across 18 categories and 66 sub-categories, and covers over 50 000 lncRNAs. We not only collected lncRNA sets based on downstream regulatory data sources, but also identified a large number of lncRNA sets regulated by upstream transcription factors (TFs) and DNA regulatory elements by integrating TF ChIP-seq, DNase-seq, ATAC-seq and H3K27ac ChIP-seq data. Importantly, LncSEA provides annotation and enrichment analyses of lncRNA sets associated with upstream regulators and downstream targets. In summary, LncSEA is a powerful platform that provides a variety of types of lncRNA sets for users, and supports lncRNA annotations and enrichment analyses. The LncSEA database is freely accessible at http://bio.liclab.net/LncSEA/index.php.
Collapse
Affiliation(s)
- Jiaxin Chen
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Jian Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Yu Gao
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Yanyu Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Chenchen Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Chao Song
- Department of Pharmacology, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Ziyu Ning
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Xinyuan Zhou
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Jianmei Zhao
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Minghong Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Yuexin Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Ling Wei
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Qi Pan
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Yong Jiang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Fengcui Qian
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yongsan Yang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Qiuyu Wang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, China
| |
Collapse
|
6
|
Savino A, Provero P, Poli V. Differential Co-Expression Analyses Allow the Identification of Critical Signalling Pathways Altered during Tumour Transformation and Progression. Int J Mol Sci 2020; 21:E9461. [PMID: 33322692 PMCID: PMC7764314 DOI: 10.3390/ijms21249461] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 02/02/2023] Open
Abstract
Biological systems respond to perturbations through the rewiring of molecular interactions, organised in gene regulatory networks (GRNs). Among these, the increasingly high availability of transcriptomic data makes gene co-expression networks the most exploited ones. Differential co-expression networks are useful tools to identify changes in response to an external perturbation, such as mutations predisposing to cancer development, and leading to changes in the activity of gene expression regulators or signalling. They can help explain the robustness of cancer cells to perturbations and identify promising candidates for targeted therapy, moreover providing higher specificity with respect to standard co-expression methods. Here, we comprehensively review the literature about the methods developed to assess differential co-expression and their applications to cancer biology. Via the comparison of normal and diseased conditions and of different tumour stages, studies based on these methods led to the definition of pathways involved in gene network reorganisation upon oncogenes' mutations and tumour progression, often converging on immune system signalling. A relevant implementation still lagging behind is the integration of different data types, which would greatly improve network interpretability. Most importantly, performance and predictivity evaluation of the large variety of mathematical models proposed would urgently require experimental validations and systematic comparisons. We believe that future work on differential gene co-expression networks, complemented with additional omics data and experimentally tested, will considerably improve our insights into the biology of tumours.
Collapse
Affiliation(s)
- Aurora Savino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Corso Massimo D’Ázeglio 52, 10126 Turin, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Valeria Poli
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
7
|
Han X, Wang D, Zhao P, Liu C, Hao Y, Chang L, Zhao J, Zhao W, Mu L, Wang J, Li H, Kong Q, Han J. Inference of Subpathway Activity Profiles Reveals Metabolism Abnormal Subpathway Regions in Glioblastoma Multiforme. Front Oncol 2020; 10:1549. [PMID: 33072547 PMCID: PMC7533644 DOI: 10.3389/fonc.2020.01549] [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: 12/23/2019] [Accepted: 07/20/2020] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma, also known as glioblastoma multiforme (GBM), is the most malignant form of glioma and represents 81% of malignant brain and central nervous system (CNS) tumors. Like most cancers, GBM causes metabolic recombination to promote cell survival, proliferation, and invasion of cancer cells. In this study, we propose a method for constructing the metabolic subpathway activity score matrix to accurately identify abnormal targets of GBM metabolism. By integrating gene expression data from different sequencing methods, our method identified 25 metabolic subpathways that were significantly abnormal in the GBM patient population, and most of these subpathways have been reported to have an effect on GBM. Through the analysis of 25 GBM-related metabolic subpathways, we found that (S)-2,3-Epoxysqualene, which was at the central region of the sterol biosynthesis subpathway, may have a greater impact on the entire pathway, suggesting a potential high association with GBM. Analysis of CCK8 cell activity indicated that (S)-2,3-Epoxysqualene can indeed inhibit the activity of U87-MG cells. By flow cytometry, we demonstrated that (S)-2,3-Epoxysqualene not only arrested the U87-MG cell cycle in the G0/G1 phase but also induced cell apoptosis. These results confirm the reliability of our proposed metabolic subpathway identification method and suggest that (S)-2,3-Epoxysqualene has potential therapeutic value for GBM. In order to make the method more broadly applicable, we have developed an R system package crmSubpathway to perform disease-related metabolic subpathway identification and it is freely available on the GitHub (https://github.com/hanjunwei-lab/crmSubpathway).
Collapse
Affiliation(s)
- Xudong Han
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Donghua Wang
- Department of General Surgery, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin, China
| | - Ping Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Chonghui Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Yue Hao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Lulu Chang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Jiarui Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Wei Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, China.,Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Liang D, Liu Q, Zhou K, Jia W, Xie G, Chen T. IP4M: an integrated platform for mass spectrometry-based metabolomics data mining. BMC Bioinformatics 2020; 21:444. [PMID: 33028191 PMCID: PMC7542974 DOI: 10.1186/s12859-020-03786-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Metabolomics data analyses rely on the use of bioinformatics tools. Many integrated multi-functional tools have been developed for untargeted metabolomics data processing and have been widely used. More alternative platforms are expected for both basic and advanced users. Results Integrated mass spectrometry-based untargeted metabolomics data mining (IP4M) software was designed and developed. The IP4M, has 62 functions categorized into 8 modules, covering all the steps of metabolomics data mining, including raw data preprocessing (alignment, peak de-convolution, peak picking, and isotope filtering), peak annotation, peak table preprocessing, basic statistical description, classification and biomarker detection, correlation analysis, cluster and sub-cluster analysis, regression analysis, ROC analysis, pathway and enrichment analysis, and sample size and power analysis. Additionally, a KEGG-derived metabolic reaction database was embedded and a series of ratio variables (product/substrate) can be generated with enlarged information on enzyme activity. A new method, GRaMM, for correlation analysis between metabolome and microbiome data was also provided. IP4M provides both a number of parameters for customized and refined analysis (for expert users), as well as 4 simplified workflows with few key parameters (for beginners who are unfamiliar with computational metabolomics). The performance of IP4M was evaluated and compared with existing computational platforms using 2 data sets derived from standards mixture and 2 data sets derived from serum samples, from GC–MS and LC–MS respectively. Conclusion IP4M is powerful, modularized, customizable and easy-to-use. It is a good choice for metabolomics data processing and analysis. Free versions for Windows, MAC OS, and Linux systems are provided.
Collapse
Affiliation(s)
- Dandan Liang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Quan Liu
- Human Metabolomics Institute, Inc., Shenzhen, 518109, Guangdong, China
| | - Kejun Zhou
- Human Metabolomics Institute, Inc., Shenzhen, 518109, Guangdong, China
| | - Wei Jia
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Guoxiang Xie
- Human Metabolomics Institute, Inc., Shenzhen, 518109, Guangdong, China.
| | - Tianlu Chen
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
9
|
Li M, Zhao J, Li X, Chen Y, Feng C, Qian F, Liu Y, Zhang J, He J, Ai B, Ning Z, Liu W, Bai X, Han X, Wu Z, Xu X, Tang Z, Pan Q, Xu L, Li C, Wang Q, Li E. HiFreSP: A novel high-frequency sub-pathway mining approach to identify robust prognostic gene signatures. Brief Bioinform 2020; 21:1411-1424. [PMID: 31350847 DOI: 10.1093/bib/bbz078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/19/2019] [Accepted: 06/04/2019] [Indexed: 02/05/2023] Open
Abstract
With the increasing awareness of heterogeneity in cancers, better prediction of cancer prognosis is much needed for more personalized treatment. Recently, extensive efforts have been made to explore the variations in gene expression for better prognosis. However, the prognostic gene signatures predicted by most existing methods have little robustness among different datasets of the same cancer. To improve the robustness of the gene signatures, we propose a novel high-frequency sub-pathways mining approach (HiFreSP), integrating a randomization strategy with gene interaction pathways. We identified a six-gene signature (CCND1, CSF3R, E2F2, JUP, RARA and TCF7) in esophageal squamous cell carcinoma (ESCC) by HiFreSP. This signature displayed a strong ability to predict the clinical outcome of ESCC patients in two independent datasets (log-rank test, P = 0.0045 and 0.0087). To further show the predictive performance of HiFreSP, we applied it to two other cancers: pancreatic adenocarcinoma and breast cancer. The identified signatures show high predictive power in all testing datasets of the two cancers. Furthermore, compared with the two popular prognosis signature predicting methods, the least absolute shrinkage and selection operator penalized Cox proportional hazards model and the random survival forest, HiFreSP showed better predictive accuracy and generalization across all testing datasets of the above three cancers. Lastly, we applied HiFreSP to 8137 patients involving 20 cancer types in the TCGA database and found high-frequency prognosis-associated pathways in many cancers. Taken together, HiFreSP shows higher prognostic capability and greater robustness, and the identified signatures provide clinical guidance for cancer prognosis. HiFreSP is freely available via GitHub: https://github.com/chunquanlipathway/HiFreSP.
Collapse
Affiliation(s)
- Meng Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jianmei Zhao
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xuecang Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yang Chen
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Chenchen Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Fengcui Qian
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yuejuan Liu
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jian Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jianzhong He
- Institute of Oncologic Pathology, Shantou University Medical College
| | - Bo Ai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Ziyu Ning
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Wei Liu
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Xuefeng Bai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xiaole Han
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Zhiyong Wu
- Departments of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University
| | - Xiue Xu
- Institute of Oncologic Pathology, Shantou University Medical College
| | - Zhidong Tang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Qi Pan
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Liyan Xu
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Qiuyu Wang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Enmin Li
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| |
Collapse
|
10
|
Piran M, Karbalaei R, Piran M, Aldahdooh J, Mirzaie M, Ansari-Pour N, Tang J, Jafari M. Can We Assume the Gene Expression Profile as a Proxy for Signaling Network Activity? Biomolecules 2020; 10:biom10060850. [PMID: 32503292 PMCID: PMC7355924 DOI: 10.3390/biom10060850] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Studying relationships among gene products by expression profile analysis is a common approach in systems biology. Many studies have generalized the outcomes to the different levels of central dogma information flow and assumed a correlation of transcript and protein expression levels. However, the relation between the various types of interaction (i.e., activation and inhibition) of gene products to their expression profiles has not been widely studied. In fact, looking for any perturbation according to differentially expressed genes is the common approach, while analyzing the effects of altered expression on the activity of signaling pathways is often ignored. In this study, we examine whether significant changes in gene expression necessarily lead to dysregulated signaling pathways. Using four commonly used and comprehensive databases, we extracted all relevant gene expression data and all relationships among directly linked gene pairs. We aimed to evaluate the ratio of coherency or sign consistency between the expression level as well as the causal relationships among the gene pairs. Through a comparison with random unconnected gene pairs, we illustrate that the signaling network is incoherent, and inconsistent with the recorded expression profile. Finally, we demonstrate that, to infer perturbed signaling pathways, we need to consider the type of relationships in addition to gene-product expression data, especially at the transcript level. We assert that identifying enriched biological processes via differentially expressed genes is limited when attempting to infer dysregulated pathways.
Collapse
Affiliation(s)
- Mehran Piran
- Bioinformatics and Computational Biology Research Center, Shiraz University of Medical Sciences, Shiraz P.O. Box 71336-54361, Iran;
| | - Reza Karbalaei
- Department of Biology, Temple University, Philadelphia, PA 19122, USA;
| | - Mehrdad Piran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran P.O. Box 14177-55469, Iran;
| | - Jehad Aldahdooh
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00270 Helsinki, Finland;
| | - Mehdi Mirzaie
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-134, Iran;
| | - Naser Ansari-Pour
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LF, UK;
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00270 Helsinki, Finland;
- Correspondence: (J.T.); (M.J.)
| | - Mohieddin Jafari
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00270 Helsinki, Finland;
- Correspondence: (J.T.); (M.J.)
| |
Collapse
|
11
|
Zhao Y, Piekos S, Hoang TH, Shin DG. A framework using topological pathways for deeper analysis of transcriptome data. BMC Genomics 2020; 21:834. [PMID: 32138666 PMCID: PMC7057456 DOI: 10.1186/s12864-019-6155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/30/2019] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Pathway analysis is one of the later stage data analysis steps essential in interpreting high-throughput gene expression data. We propose a set of algorithms which given gene expression data can recognize which portion of sub-pathways are actively utilized in the biological system being studied. The degree of activation is measured by conditional probability of the input expression data based on the Bayesian Network model constructed from the topological pathway. RESULTS We demonstrate the effectiveness of our pathway analysis method by conducting two case studies. The first one applies our method to a well-studied temporal microarray data set for the cell cycle using the KEGG Cell Cycle pathway. Our method closely reproduces the biological claims associated with the data sets, but unlike the original work ours can produce how pathway routes interact with each other above and beyond merely identifying which pathway routes are involved in the process. The second study applies the method to the p53 mutation microarray data to perform a comparative study. CONCLUSIONS We show that our method achieves comparable performance against all other pathway analysis systems included in this study in identifying p53 altered pathways. Our method could pave a new way of carrying out next generation pathway analysis.
Collapse
Affiliation(s)
- Yue Zhao
- Computer Science and Engineering Department, University of Connecticut, 371 Fairfield Way, Unit 4155, Storrs, 06269 USA
| | - Stephanie Piekos
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, USA
| | - Tham H. Hoang
- Computer Science and Engineering Department, University of Connecticut, 371 Fairfield Way, Unit 4155, Storrs, 06269 USA
| | - Dong-Guk Shin
- Computer Science and Engineering Department, University of Connecticut, 371 Fairfield Way, Unit 4155, Storrs, 06269 USA
| |
Collapse
|
12
|
Gasparini S, Del Vecchio G, Gioiosa S, Flati T, Castrignano T, Legnini I, Licursi V, Ricceri L, Scattoni ML, Rinaldi A, Presutti C, Mannironi C. Differential Expression of Hippocampal Circular RNAs in the BTBR Mouse Model for Autism Spectrum Disorder. Mol Neurobiol 2020; 57:2301-2313. [PMID: 32020500 DOI: 10.1007/s12035-020-01878-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/13/2020] [Indexed: 01/02/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition with unknown etiology. Recent experimental evidences suggest the contribution of non-coding RNAs (ncRNAs) in the pathophysiology of ASD. In this work, we aimed to investigate the expression profile of the ncRNA class of circular RNAs (circRNAs) in the hippocampus of the BTBR T + tf/J (BTBR) mouse model and age-matched C57BL/6J (B6) mice. Alongside, we analyzed BTBR hippocampal gene expression profile to evaluate possible correlations between the differential abundance of circular and linear gene products. From RNA sequencing data, we identified circRNAs highly modulated in BTBR mice. Thirteen circRNAs and their corresponding linear isoforms were validated by RT-qPCR analysis. The BTBR-regulated circCdh9 was better characterized in terms of molecular structure and expression, highlighting altered levels not only in the hippocampus, but also in the cerebellum, prefrontal cortex, and amygdala. Finally, gene expression analysis of the BTBR hippocampus pinpointed altered biological and molecular pathways relevant for the ASD phenotype. By comparison of circRNA and gene expression profiles, we identified 6 genes significantly regulated at either circRNA or mRNA gene products, suggesting low overall correlation between circRNA and host gene expression. In conclusion, our results indicate a consistent deregulation of circRNA expression in the hippocampus of BTBR mice. ASD-related circRNAs should be considered in functional studies to identify their contribution to the etiology of the disorder. In addition, as abundant and highly stable molecules, circRNAs represent interesting potential biomarkers for autism.
Collapse
Affiliation(s)
- Silvia Gasparini
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Giorgia Del Vecchio
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Silvia Gioiosa
- SCAI-Super Computing Applications and Innovation Department, CINECA, Rome, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, National Research Council, Bari, Italy
| | - Tiziano Flati
- SCAI-Super Computing Applications and Innovation Department, CINECA, Rome, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, National Research Council, Bari, Italy
| | - Tiziana Castrignano
- SCAI-Super Computing Applications and Innovation Department, CINECA, Rome, Italy
- Department of Ecological and Biological, Sciences University of Tuscia, Viterbo, Italy
| | - Ivano Legnini
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | | | | | - Arianna Rinaldi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Carlo Presutti
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy.
| | - Cecilia Mannironi
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy.
| |
Collapse
|
13
|
Li Y, Wu Y, Zhang X, Bai Y, Akthar LM, Lu X, Shi M, Zhao J, Jiang Q, Li Y. SCIA: A Novel Gene Set Analysis Applicable to Data With Different Characteristics. Front Genet 2019; 10:598. [PMID: 31293623 PMCID: PMC6603225 DOI: 10.3389/fgene.2019.00598] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/05/2019] [Indexed: 01/06/2023] Open
Abstract
Gene set analysis is commonly used in functional enrichment and molecular pathway analyses. Most of the present methods are based on the competitive testing methods which assume each gene is independent of the others. However, the false discovery rates of competitive methods are amplified when they are applied to datasets with high inter-gene correlations. The self-contained testing methods could solve this problem, but there are other restrictions on data characteristics. Therefore, a statistically rigorous testing method applicable to different datasets with various complex characteristics is needed to obtain unbiased and comparable results. We propose a self-contained and competitive incorporated analysis (SCIA) to alleviate the bias caused by the limited application scope of existing gene set analysis methods. This is accomplished through a novel permutation strategy using a priori biological networks to selectively permute gene labels with different probabilities. In simulation studies, SCIA was compared with four representative analysis methods (GSEA, CAMERA, ROAST, and NES), and produced the best performance in both false discovery rate and sensitivity under most conditions with different parameter settings. Further, the KEGG pathway analysis on two real datasets of lung cancer showed that the results found by SCIA in both of the two datasets are much more than that of GSEA and most of them could be supported by literature. Overall, SCIA promisingly offers researchers more reliable and comparable results with different datasets.
Collapse
Affiliation(s)
- Yiqun Li
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ying Wu
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaohan Zhang
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yunfan Bai
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Luqman Muhammad Akthar
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xin Lu
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ming Shi
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jianxiang Zhao
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Qinghua Jiang
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yu Li
- Department of Laboratory of Cancer Biology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
14
|
Andrejeva D, Kugler JM, Nguyen HT, Malmendal A, Holm ML, Toft BG, Loya AC, Cohen SM. Metabolic control of PPAR activity by aldehyde dehydrogenase regulates invasive cell behavior and predicts survival in hepatocellular and renal clear cell carcinoma. BMC Cancer 2018; 18:1180. [PMID: 30486822 PMCID: PMC6264057 DOI: 10.1186/s12885-018-5061-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/07/2018] [Indexed: 01/16/2023] Open
Abstract
Background Changes in cellular metabolism are now recognized as potential drivers of cancer development, rather than as secondary consequences of disease. Here, we explore the mechanism by which metabolic changes dependent on aldehyde dehydrogenase impact cancer development. Methods ALDH7A1 was identified as a potential cancer gene using a Drosophila in vivo metastasis model. The role of the human ortholog was examined using RNA interference in cell-based assays of cell migration and invasion. 1H-NMR metabolite profiling was used to identify metabolic changes in ALDH7A1-depleted cells. Publically available cancer gene expression data was interrogated to identify a gene-expression signature associated with depletion of ALDH7A1. Computational pathway and gene set enrichment analysis was used to identify signaling pathways and cellular processes that were correlated with reduced ALDH7A1 expression in cancer. A variety of statistical tests used to evaluate these analyses are described in detail in the methods section. Immunohistochemistry was used to assess ALDH7A1 expression in tissue samples from cancer patients. Results Depletion of ALDH7A1 increased cellular migration and invasiveness in vitro. Depletion of ALDH7A1 led to reduced levels of metabolites identified as ligands for Peroxisome proliferator-activated receptor (PPARα). Analysis of publically available cancer gene expression data revealed that ALDH7A1 mRNA levels were reduced in many human cancers, and that this correlated with poor survival in kidney and liver cancer patients. Using pathway and gene set enrichment analysis, we establish a correlation between low ALDH7A1 levels, reduced PPAR signaling and reduced patient survival. Metabolic profiling showed that endogenous PPARα ligands were reduced in ALDH7A1-depleted cells. ALDH7A1-depletion led to reduced PPAR transcriptional activity. Treatment with a PPARα agonist restored normal cellular behavior. Low ALDH7A1 protein levels correlated with poor clinical outcome in hepatocellular and renal clear cell carcinoma patients. Conclusions We provide evidence that low ALDH7A1 expression is a useful prognostic marker of poor clinical outcome for hepatocellular and renal clear cell carcinomas and hypothesize that patients with low ALDH7A1 might benefit from therapeutic approaches addressing PPARα activity. Electronic supplementary material The online version of this article (10.1186/s12885-018-5061-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diana Andrejeva
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Jan-Michael Kugler
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark.
| | - Hung Thanh Nguyen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Anders Malmendal
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Mette Lind Holm
- Department of Urology, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen Ø, Denmark
| | | | - Anand C Loya
- Department of Pathology, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen Ø, Denmark
| | - Stephen M Cohen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
15
|
DynSig: Modelling Dynamic Signaling Alterations along Gene Pathways for Identifying Differential Pathways. Genes (Basel) 2018; 9:genes9070323. [PMID: 29954150 PMCID: PMC6071020 DOI: 10.3390/genes9070323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 11/16/2022] Open
Abstract
Although a number of methods have been proposed for identifying differentially expressed pathways (DEPs), few efforts consider the dynamic components of pathway networks, i.e., gene links. We here propose a signaling dynamics detection method for identification of DEPs, DynSig, which detects the molecular signaling changes in cancerous cells along pathway topology. Specifically, DynSig relies on gene links, instead of gene nodes, in pathways, and models the dynamic behavior of pathways based on Markov chain model (MCM). By incorporating the dynamics of molecular signaling, DynSig allows for an in-depth characterization of pathway activity. To identify DEPs, a novel statistic of activity alteration of pathways was formulated as an overall signaling perturbation score between sample classes. Experimental results on both simulation and real-world datasets demonstrate the effectiveness and efficiency of the proposed method in identifying differential pathways.
Collapse
|
16
|
Park HJ, Kim S, Li W. Model-based analysis of competing-endogenous pathways (MACPath) in human cancers. PLoS Comput Biol 2018; 14:e1006074. [PMID: 29565967 PMCID: PMC5882149 DOI: 10.1371/journal.pcbi.1006074] [Citation(s) in RCA: 8] [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: 09/05/2017] [Revised: 04/03/2018] [Accepted: 03/06/2018] [Indexed: 01/24/2023] Open
Abstract
Competing endogenous RNA (ceRNA) has emerged as an important post-transcriptional mechanism that simultaneously alters expressions of thousands genes in cancers. However, only a few ceRNA genes have been studied for their functions to date. To understand the major biological functions of thousands ceRNA genes as a whole, we designed Model-based Analysis of Competing-endogenous Pathways (MACPath) to infer pathways co-regulated through ceRNA mechanism (cePathways). Our analysis on breast tumors suggested that NGF (nerve growth factor)-induced tumor cell proliferation might be associated with tumor-related growth factor pathways through ceRNA. MACPath also identified indirect cePathways, whose ceRNA relationship is mediated by mediating ceRNAs. Finally, MACPath identified mediating ceRNAs that connect the indirect cePathways based on efficient integer linear programming technique. Mediating ceRNAs are unexpectedly enriched in tumor suppressor genes, whose down-regulation is suspected to disrupt indirect cePathways, such as between DNA replication and WNT signaling pathways. Altogether, MACPath is the first computational method to comprehensively understand functions of thousands ceRNA genes, both direct and indirect, at the pathway level.
Collapse
Affiliation(s)
- Hyun Jung Park
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (HJP); (WL)
| | - Soyeon Kim
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Wei Li
- Division of Biostatistics, Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (HJP); (WL)
| |
Collapse
|
17
|
Xie XP, Gan B, Yang W, Wang HQ. ctPath: Demixing pathway crosstalk effect from transcriptomics data for differential pathway identification. J Biomed Inform 2017; 73:104-114. [PMID: 28756161 DOI: 10.1016/j.jbi.2017.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 12/17/2022]
Abstract
Identifying differentially expressed pathways (DEPs) plays important roles in understanding tumor etiology and promoting clinical treatment of cancer or other diseases. By assuming gene expression to be a sparse non-negative linear combination of hidden pathway signals, we propose a pathway crosstalk-based transcriptomics data analysis method (ctPath) for identifying differentially expressed pathways. Biologically, pathways of different functions work in concert at the systematic level. The proposed method interrogates the crosstalks between pathways and discovers hidden pathway signals by mapping high-dimensional transcriptomics data into a low-dimensional pathway space. The resulted pathway signals reflect the activity level of pathways after removing pathway crosstalk effect and allow a robust identification of DEPs from inherently complex and noisy transcriptomics data. CtPath can also correct incomplete and inaccurate pathway annotations which frequently occur in public repositories. Experimental results on both simulation data and real-world cancer data demonstrate the superior performance of ctPath over other popular approaches. R code for ctPath is available for non-commercial use at the URL http://micblab.iim.ac.cn/Download/.
Collapse
Affiliation(s)
- Xin-Ping Xie
- School of Mathematics and Physics, Anhui Jianzhu University, Hefei, Anhui, China
| | - Bin Gan
- Biological Molecular Information System Lab., Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, Hefei, Anhui, China
| | - Wulin Yang
- Center for Medical Physics and Technology, Hefei Institutes of Physical Science, CAS, Hefei, Anhui, China; Cancer Hospital, CAS, Hefei, Anhui, China
| | - Hong-Qiang Wang
- Biological Molecular Information System Lab., Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, Hefei, Anhui, China; Center for Medical Physics and Technology, Hefei Institutes of Physical Science, CAS, Hefei, Anhui, China; Cancer Hospital, CAS, Hefei, Anhui, China.
| |
Collapse
|
18
|
Mohammadi S, Grama A. A convex optimization approach for identification of human tissue-specific interactomes. Bioinformatics 2017; 32:i243-i252. [PMID: 27307623 PMCID: PMC4908329 DOI: 10.1093/bioinformatics/btw245] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Motivation: Analysis of organism-specific interactomes has yielded novel insights into cellular function and coordination, understanding of pathology, and identification of markers and drug targets. Genes, however, can exhibit varying levels of cell type specificity in their expression, and their coordinated expression manifests in tissue-specific function and pathology. Tissue-specific/tissue-selective interaction mechanisms have significant applications in drug discovery, as they are more likely to reveal drug targets. Furthermore, tissue-specific transcription factors (tsTFs) are significantly implicated in human disease, including cancers. Finally, disease genes and protein complexes have the tendency to be differentially expressed in tissues in which defects cause pathology. These observations motivate the construction of refined tissue-specific interactomes from organism-specific interactomes. Results: We present a novel technique for constructing human tissue-specific interactomes. Using a variety of validation tests (Edge Set Enrichment Analysis, Gene Ontology Enrichment, Disease-Gene Subnetwork Compactness), we show that our proposed approach significantly outperforms state-of-the-art techniques. Finally, using case studies of Alzheimer’s and Parkinson’s diseases, we show that tissue-specific interactomes derived from our study can be used to construct pathways implicated in pathology and demonstrate the use of these pathways in identifying novel targets. Availability and implementation:http://www.cs.purdue.edu/homes/mohammas/projects/ActPro.html Contact:mohammadi@purdue.edu
Collapse
Affiliation(s)
- Shahin Mohammadi
- Department of Computer Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Ananth Grama
- Department of Computer Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
19
|
Will T, Helms V. Rewiring of the inferred protein interactome during blood development studied with the tool PPICompare. BMC SYSTEMS BIOLOGY 2017; 11:44. [PMID: 28376810 PMCID: PMC5379774 DOI: 10.1186/s12918-017-0400-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/26/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Differential analysis of cellular conditions is a key approach towards understanding the consequences and driving causes behind biological processes such as developmental transitions or diseases. The progress of whole-genome expression profiling enabled to conveniently capture the state of a cell's transcriptome and to detect the characteristic features that distinguish cells in specific conditions. In contrast, mapping the physical protein interactome for many samples is experimentally infeasible at the moment. For the understanding of the whole system, however, it is equally important how the interactions of proteins are rewired between cellular states. To overcome this deficiency, we recently showed how condition-specific protein interaction networks that even consider alternative splicing can be inferred from transcript expression data. Here, we present the differential network analysis tool PPICompare that was specifically designed for isoform-sensitive protein interaction networks. RESULTS Besides detecting significant rewiring events between the interactomes of grouped samples, PPICompare infers which alterations to the transcriptome caused each rewiring event and what is the minimal set of alterations necessary to explain all between-group changes. When applied to the development of blood cells, we verified that a reasonable amount of rewiring events were reported by the tool and found that differential gene expression was the major determinant of cellular adjustments to the interactome. Alternative splicing events were consistently necessary in each developmental step to explain all significant alterations and were especially important for rewiring in the context of transcriptional control. CONCLUSIONS Applying PPICompare enabled us to investigate the dynamics of the human protein interactome during developmental transitions. A platform-independent implementation of the tool PPICompare is available at https://sourceforge.net/projects/ppicompare/ .
Collapse
Affiliation(s)
- Thorsten Will
- Center for Bioinformatics, Saarland University, Campus E2.1, Saarbrücken, 66123 Germany
- Graduate School of Computer Science, Saarland University, Campus E1.3, Saarbrücken, 66123 Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Campus E2.1, Saarbrücken, 66123 Germany
| |
Collapse
|
20
|
Azad AKM, Lawen A, Keith JM. Bayesian model of signal rewiring reveals mechanisms of gene dysregulation in acquired drug resistance in breast cancer. PLoS One 2017; 12:e0173331. [PMID: 28288164 PMCID: PMC5348014 DOI: 10.1371/journal.pone.0173331] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/20/2017] [Indexed: 11/24/2022] Open
Abstract
Small molecule inhibitors, such as lapatinib, are effective against breast cancer in clinical trials, but tumor cells ultimately acquire resistance to the drug. Maintaining sensitization to drug action is essential for durable growth inhibition. Recently, adaptive reprogramming of signaling circuitry has been identified as a major cause of acquired resistance. We developed a computational framework using a Bayesian statistical approach to model signal rewiring in acquired resistance. We used the p1-model to infer potential aberrant gene-pairs with differential posterior probabilities of appearing in resistant-vs-parental networks. Results were obtained using matched gene expression profiles under resistant and parental conditions. Using two lapatinib-treated ErbB2-positive breast cancer cell-lines: SKBR3 and BT474, our method identified similar dysregulated signaling pathways including EGFR-related pathways as well as other receptor-related pathways, many of which were reported previously as compensatory pathways of EGFR-inhibition via signaling cross-talk. A manual literature survey provided strong evidence that aberrant signaling activities in dysregulated pathways are closely related to acquired resistance in EGFR tyrosine kinase inhibitors. Our approach predicted literature-supported dysregulated pathways complementary to both node-centric (SPIA, DAVID, and GATHER) and edge-centric (ESEA and PAGI) methods. Moreover, by proposing a novel pattern of aberrant signaling called V-structures, we observed that genes were dysregulated in resistant-vs-sensitive conditions when they were involved in the switch of dependencies from targeted to bypass signaling events. A literature survey of some important V-structures suggested they play a role in breast cancer metastasis and/or acquired resistance to EGFR-TKIs, where the mRNA changes of TGFBR2, LEF1 and TP53 in resistant-vs-sensitive conditions were related to the dependency switch from targeted to bypass signaling links. Our results suggest many signaling pathway structures are compromised in acquired resistance, and V-structures of aberrant signaling within/among those pathways may provide further insights into the bypass mechanism of targeted inhibition.
Collapse
Affiliation(s)
- A. K. M. Azad
- School of Mathematical Sciences, Monash University, Clayton, VIC, Australia
- * E-mail:
| | - Alfons Lawen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, Australia
| | - Jonathan M. Keith
- School of Mathematical Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
Kaushik A, Ali S, Gupta D. Altered Pathway Analyzer: A gene expression dataset analysis tool for identification and prioritization of differentially regulated and network rewired pathways. Sci Rep 2017; 7:40450. [PMID: 28084397 PMCID: PMC5233954 DOI: 10.1038/srep40450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Gene connection rewiring is an essential feature of gene network dynamics. Apart from its normal functional role, it may also lead to dysregulated functional states by disturbing pathway homeostasis. Very few computational tools measure rewiring within gene co-expression and its corresponding regulatory networks in order to identify and prioritize altered pathways which may or may not be differentially regulated. We have developed Altered Pathway Analyzer (APA), a microarray dataset analysis tool for identification and prioritization of altered pathways, including those which are differentially regulated by TFs, by quantifying rewired sub-network topology. Moreover, APA also helps in re-prioritization of APA shortlisted altered pathways enriched with context-specific genes. We performed APA analysis of simulated datasets and p53 status NCI-60 cell line microarray data to demonstrate potential of APA for identification of several case-specific altered pathways. APA analysis reveals several altered pathways not detected by other tools evaluated by us. APA analysis of unrelated prostate cancer datasets identifies sample-specific as well as conserved altered biological processes, mainly associated with lipid metabolism, cellular differentiation and proliferation. APA is designed as a cross platform tool which may be transparently customized to perform pathway analysis in different gene expression datasets. APA is freely available at http://bioinfo.icgeb.res.in/APA.
Collapse
Affiliation(s)
- Abhinav Kaushik
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Shakir Ali
- Department of Biochemistry, Jamia Hamdard, Deemed University, New Delhi 110062, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| |
Collapse
|
22
|
Yuan Z, Ji J, Zhang T, Liu Y, Zhang X, Chen W, Xue F. A novel chi-square statistic for detecting group differences between pathways in systems epidemiology. Stat Med 2016; 35:5512-5524. [PMID: 27605026 DOI: 10.1002/sim.7094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/01/2016] [Accepted: 08/16/2016] [Indexed: 12/15/2022]
Abstract
Traditional epidemiology often pays more attention to the identification of a single factor rather than to the pathway that is related to a disease, and therefore, it is difficult to explore the disease mechanism. Systems epidemiology aims to integrate putative lifestyle exposures and biomarkers extracted from multiple omics platforms to offer new insights into the pathway mechanisms that underlie disease at the human population level. One key but inadequately addressed question is how to develop powerful statistics to identify whether one candidate pathway is associated with a disease. Bearing in mind that a pathway difference can result from not only changes in the nodes but also changes in the edges, we propose a novel statistic for detecting group differences between pathways, which in principle, captures the nodes changes and edge changes, as well as simultaneously accounting for the pathway structure simultaneously. The proposed test has been proven to follow the chi-square distribution, and various simulations have shown it has better performance than other existing methods. Integrating genome-wide DNA methylation data, we analyzed one real data set from the Bogalusa cohort study and significantly identified a potential pathway, Smoking → SOCS3 → PIK3R1, which was strongly associated with abdominal obesity. The proposed test was powerful and efficient at identifying pathway differences between two groups, and it can be extended to other disciplines that involve statistical comparisons between pathways. The source code in R is available on our website. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Jiadong Ji
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China.,Department of Epidemiology, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, U.S.A
| | - Yi Liu
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaoshuai Zhang
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| | - Wei Chen
- Department of Epidemiology, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, U.S.A
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, Shandong, China
| |
Collapse
|
23
|
Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, McDermott MG, Monteiro CD, Gundersen GW, Ma'ayan A. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 2016; 44. [PMID: 27141961 PMCID: PMC4987924 DOI: 10.1093/nar%2fgkw377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Enrichment analysis is a popular method for analyzing gene sets generated by genome-wide experiments. Here we present a significant update to one of the tools in this domain called Enrichr. Enrichr currently contains a large collection of diverse gene set libraries available for analysis and download. In total, Enrichr currently contains 180 184 annotated gene sets from 102 gene set libraries. New features have been added to Enrichr including the ability to submit fuzzy sets, upload BED files, improved application programming interface and visualization of the results as clustergrams. Overall, Enrichr is a comprehensive resource for curated gene sets and a search engine that accumulates biological knowledge for further biological discoveries. Enrichr is freely available at: http://amp.pharm.mssm.edu/Enrichr.
Collapse
Affiliation(s)
- Maxim V. Kuleshov
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Matthew R. Jones
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Andrew D. Rouillard
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Nicolas F. Fernandez
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Qiaonan Duan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Zichen Wang
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Simon Koplev
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Sherry L. Jenkins
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Kathleen M. Jagodnik
- Fluid Physics and Transport Processes Branch, NASA Glenn Research Center, 21000 Brookpark Rd., Cleveland, OH 44135, USA
| | - Alexander Lachmann
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Michael G. McDermott
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Caroline D. Monteiro
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Gregory W. Gundersen
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA,To whom correspondence should be addressed. Tel: +1 212 241 1153; Fax: +1 212 996 7214;
| |
Collapse
|
24
|
Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, McDermott MG, Monteiro CD, Gundersen GW, Ma'ayan A. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 2016. [PMID: 27141961 DOI: 10.1093/nar/gkw377)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Enrichment analysis is a popular method for analyzing gene sets generated by genome-wide experiments. Here we present a significant update to one of the tools in this domain called Enrichr. Enrichr currently contains a large collection of diverse gene set libraries available for analysis and download. In total, Enrichr currently contains 180 184 annotated gene sets from 102 gene set libraries. New features have been added to Enrichr including the ability to submit fuzzy sets, upload BED files, improved application programming interface and visualization of the results as clustergrams. Overall, Enrichr is a comprehensive resource for curated gene sets and a search engine that accumulates biological knowledge for further biological discoveries. Enrichr is freely available at: http://amp.pharm.mssm.edu/Enrichr.
Collapse
Affiliation(s)
- Maxim V Kuleshov
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Matthew R Jones
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Andrew D Rouillard
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Nicolas F Fernandez
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Qiaonan Duan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Zichen Wang
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Simon Koplev
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Sherry L Jenkins
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Kathleen M Jagodnik
- Fluid Physics and Transport Processes Branch, NASA Glenn Research Center, 21000 Brookpark Rd., Cleveland, OH 44135, USA
| | - Alexander Lachmann
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Michael G McDermott
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Caroline D Monteiro
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Gregory W Gundersen
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| |
Collapse
|
25
|
Identifying epigenetically dysregulated pathways from pathway-pathway interaction networks. Comput Biol Med 2016; 76:160-7. [PMID: 27454244 DOI: 10.1016/j.compbiomed.2016.06.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Identification of pathways that show significant difference in activity between disease and control samples have been an interesting topic of research for over a decade. Pathways so identified serve as potential indicators of aberrations in phenotype or a disease condition. Recently, epigenetic mechanisms such as DNA methylation are known to play an important role in altering the regulatory mechanism of biological pathways. It is reasonable to think that a set of genes that show significant difference in expression and methylation interact together to form a network of pathways. Existing pathway identification methods fail to capture the complex interplay between interacting pathways. RESULTS This paper proposes a novel framework to identify biological pathways that are dysregulated by epigenetic mechanisms. Experiments on four benchmark cancer datasets and comparison with state-of-the-art pathway identification methods reveal the effectiveness of the proposed approach. CONCLUSION The proposed framework incorporates both topology and biological relationships of pathways. Comparison with state-of-the-art techniques reveals promising results. Epigenetic signatures identified from pathway interaction networks can help to advance Molecular Pathological Epidemiology (MPE) research efforts by predicting tumor molecular changes.
Collapse
|
26
|
Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, McDermott MG, Monteiro CD, Gundersen GW, Ma'ayan A. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 2016; 44:W90-7. [PMID: 27141961 PMCID: PMC4987924 DOI: 10.1093/nar/gkw377] [Citation(s) in RCA: 5941] [Impact Index Per Article: 742.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/25/2016] [Indexed: 12/11/2022] Open
Abstract
Enrichment analysis is a popular method for analyzing gene sets generated by genome-wide experiments. Here we present a significant update to one of the tools in this domain called Enrichr. Enrichr currently contains a large collection of diverse gene set libraries available for analysis and download. In total, Enrichr currently contains 180 184 annotated gene sets from 102 gene set libraries. New features have been added to Enrichr including the ability to submit fuzzy sets, upload BED files, improved application programming interface and visualization of the results as clustergrams. Overall, Enrichr is a comprehensive resource for curated gene sets and a search engine that accumulates biological knowledge for further biological discoveries. Enrichr is freely available at: http://amp.pharm.mssm.edu/Enrichr.
Collapse
Affiliation(s)
- Maxim V Kuleshov
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Matthew R Jones
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Andrew D Rouillard
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Nicolas F Fernandez
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Qiaonan Duan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Zichen Wang
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Simon Koplev
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Sherry L Jenkins
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Kathleen M Jagodnik
- Fluid Physics and Transport Processes Branch, NASA Glenn Research Center, 21000 Brookpark Rd., Cleveland, OH 44135, USA
| | - Alexander Lachmann
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Michael G McDermott
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Caroline D Monteiro
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Gregory W Gundersen
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1215, New York, NY 10029, USA
| |
Collapse
|