1
|
Kumar R, Romano JD, Ritchie MD. Network-based analyses of multiomics data in biomedicine. BioData Min 2025; 18:37. [PMID: 40426270 PMCID: PMC12117783 DOI: 10.1186/s13040-025-00452-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Network representations of data are designed to encode relationships between concepts as sets of edges between nodes. Human biology is inherently complex and is represented by data that often exists in a hierarchical nature. One canonical example is the relationship that exists within and between various -omics datasets, including genomics, transcriptomics, and proteomics, among others. Encoding such data in a network-based or graph-based representation allows the explicit incorporation of such relationships into various biomedical big data tasks, including (but not limited to) disease subtyping, interaction prediction, biomarker identification, and patient classification. This review will present various existing approaches in using network representations and analysis of data in multiomics in the framework of deep learning and machine learning approaches, subdivided into supervised and unsupervised approaches, to identify benefits and drawbacks of various approaches as well as the possible next steps for the field.
Collapse
Affiliation(s)
- Rachit Kumar
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph D Romano
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Deng Z, Wu J, Chen X, Li G, Liu J, Hu Z, Li R, Deng W. MNMO: discover driver genes from a multi-omics data based-multi-layer network. Bioinformatics 2025; 41:btaf134. [PMID: 40152235 PMCID: PMC12033032 DOI: 10.1093/bioinformatics/btaf134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 03/29/2025] Open
Abstract
MOTIVATION Cancer as a public health problem is driven by genomic variations in "cancer driver" genes. The identification of driver genes is critical for the discovery of key biomarkers and the development of personalized therapy. RESULTS We propose a prediction method MNMO: a multi-layer network model based on multi-omics data. MNMO firstly constructs a dynamically adjusted four-layer network composed of miRNAs and three kinds of genes with different features. Then three kinds of scores, i.e. control capacity, mutation score, and network score, are devised and calculated by harmonic mean to produce the integrated gene score. Experiments were performed on three kinds of real cancer data to compare the identification performance of method MNMO with that of six state-of-the-art ones. The results indicate that method MNMO presents the best identification performance under most circumstances. The genes prioritized by method MNMO not only have a better match to the benchmark ones than those identified by the other methods, but also are all associated with the development and progression of cancers. In addition, some extended versions of method MNMO can further achieve better performance on most evaluation metrics for some specific datasets. They may be more conducive to identifying tissue-specific genes, which has been verified through a number of experiments. AVAILABILITY AND IMPLEMENTATION The source code and the R package "MNMO" are available at https://github.com/Zheng-D/MNMO. The dataset and code are archived at https://doi.org/10.5281/zenodo.14969986.
Collapse
Affiliation(s)
- Zheng Deng
- Guangxi Key Lab of Multi-source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- College of Computer Science and Information Engineering, Guangxi Normal University, Guilin 541004, China
| | - Jingli Wu
- Guangxi Key Lab of Multi-source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- College of Computer Science and Information Engineering, Guangxi Normal University, Guilin 541004, China
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
| | - Xiaorong Chen
- College of Computer, National University of Defense Technology, Changsha 410073, China
| | - Gaoshi Li
- Guangxi Key Lab of Multi-source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- College of Computer Science and Information Engineering, Guangxi Normal University, Guilin 541004, China
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
| | - Jiafei Liu
- Guangxi Key Lab of Multi-source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- College of Computer Science and Information Engineering, Guangxi Normal University, Guilin 541004, China
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
| | - Zhipeng Hu
- Guangxi Key Lab of Multi-source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- College of Computer Science and Information Engineering, Guangxi Normal University, Guilin 541004, China
| | - Rongyuan Li
- Guangxi Key Lab of Multi-source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- College of Computer Science and Information Engineering, Guangxi Normal University, Guilin 541004, China
| | - Wansu Deng
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
3
|
Zi R, Shen K, Zheng P, Su X, Yang Y, Chen Y, Dai H, Mao C, Lu Y, Wang L, Ma H, Wang W, Li Q, Lu W, Li C, Zheng S, Shi H, Liu X, Chen Z, Liang H, Ou J. NPC1L1 on pancreatic adenocarcinoma cell functions as a two-pronged checkpoint against antitumor activity. Innovation (N Y) 2025; 6:100783. [PMID: 40098667 PMCID: PMC11910884 DOI: 10.1016/j.xinn.2024.100783] [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: 04/21/2024] [Accepted: 12/30/2024] [Indexed: 03/19/2025] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a highly lethal malignancy with an immunosuppressive microenvironment and a limited immunotherapy response. Cholesterol is necessary for rapid growth of cancer cells, and cholesterol metabolism reprogramming is a hallmark of PAAD. How PAAD cells initiate cholesterol reprogramming to sustain their growth demand and suppressive immunomicroenvironment remains elusive. In this study, we for the first time revealed that PAAD cells overcome cholesterol shortage and immune surveillance via ectopically overexpressing NPC1L1, a cholesterol transporter, but function as a two-pronged checkpoint, which not only directly suppresses TCR activation of CD8+T cells but also hijacks the intracellular cholesterol from CD8+T cells. In vivo, we showed that ezetimibe, an NPC1L1 inhibitor usually for hypercholesterolemia, efficiently prevented PAAD cells from depriving cholesterol of CD8+T cells, and improved the anti-tumor immunity of PAAD to synergize with PD-1 blockade, suggesting NPC1L1 as a promising target to rescue the anti-tumor activity in PAAD.
Collapse
Affiliation(s)
- Ruiyang Zi
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kaicheng Shen
- Department of Oncology, Fuling Hospital of Chongqing University, Chongqing 408000, China
| | - Pengfei Zheng
- College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xingxing Su
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yishi Yang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yanrong Chen
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Haisu Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chengyi Mao
- Department of Pathology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Yongling Lu
- Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Hongbo Ma
- Department of Oncology, Fuling Hospital of Chongqing University, Chongqing 408000, China
| | - Wei Wang
- Department of Oncology, Fuling Hospital of Chongqing University, Chongqing 408000, China
| | - Qingyun Li
- Genecast Biotechnology Co., Wuxi 214104, China
| | - Wei Lu
- Galixir Technologies, Beijing 100086, China
| | | | | | - Hui Shi
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaohong Liu
- National University of Singapore (Chongqing) Research Institute, Chongqing 401123, China
| | - Zhiyu Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Juanjuan Ou
- Yu-Yue Pathology Scientific Research Center, Chongqing 401329, China
- Center for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu 610042, China
| |
Collapse
|
4
|
Li G, Hu Z, Luo X, Liu J, Wu J, Peng W, Zhu X. Identification of cancer driver genes based on hierarchical weak consensus model. Health Inf Sci Syst 2024; 12:21. [PMID: 38464463 PMCID: PMC10917728 DOI: 10.1007/s13755-024-00279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 01/31/2024] [Indexed: 03/12/2024] Open
Abstract
Cancer is a complex gene mutation disease that derives from the accumulation of mutations during somatic cell evolution. With the advent of high-throughput technology, a large amount of omics data has been generated, and how to find cancer-related driver genes from a large number of omics data is a challenge. In the early stage, the researchers developed many frequency-based driver genes identification methods, but they could not identify driver genes with low mutation rates well. Afterwards, researchers developed network-based methods by fusing multi-omics data, but they rarely considered the connection among features. In this paper, after analyzing a large number of methods for integrating multi-omics data, a hierarchical weak consensus model for fusing multiple features is proposed according to the connection among features. By analyzing the connection between PPI network and co-mutation hypergraph network, this paper firstly proposes a new topological feature, called co-mutation clustering coefficient (CMCC). Then, a hierarchical weak consensus model is used to integrate CMCC, mRNA and miRNA differential expression scores, and a new driver genes identification method HWC is proposed. In this paper, the HWC method and current 7 state-of-the-art methods are compared on three types of cancers. The comparison results show that HWC has the best identification performance in statistical evaluation index, functional consistency and the partial area under ROC curve. Supplementary Information The online version contains supplementary material available at 10.1007/s13755-024-00279-6.
Collapse
Affiliation(s)
- Gaoshi Li
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, 541004 China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin, 541004 Guangxi China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, 541004 Guangxi China
| | - Zhipeng Hu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, 541004 China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin, 541004 Guangxi China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, 541004 Guangxi China
| | - Xinlong Luo
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, 541004 China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin, 541004 Guangxi China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, 541004 Guangxi China
| | - Jiafei Liu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, 541004 China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin, 541004 Guangxi China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, 541004 Guangxi China
| | - Jingli Wu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, 541004 China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin, 541004 Guangxi China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, 541004 Guangxi China
| | - Wei Peng
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology, Kunming, 650500 Yunnan China
| | - Xiaoshu Zhu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, 541004 China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin, 541004 Guangxi China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, 541004 Guangxi China
- School of Computer and Information Security & School of Software Engineering, Guilin University of Electronic Science and Technology, Guilin, China
| |
Collapse
|
5
|
Kim JH, Koo B, Kim S. PONYTA: prioritization of phenotype-related genes from mouse KO events using PU learning on a biological network. Bioinformatics 2024; 40:btae634. [PMID: 39432684 PMCID: PMC11561041 DOI: 10.1093/bioinformatics/btae634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
MOTIVATION Transcriptome data from gene knock-out (KO) experiments in mice provide crucial insights into the intricate interactions between genotype and phenotype. Differentially expressed gene (DEG) analysis and network propagation (NP) are well-established methods for analysing transcriptome data. To determine genes related to phenotype changes from a KO experiment, we need to choose a cutoff value for the corresponding criterion based on the specific method. Using a rigorous cutoff value for DEG analysis and NP is likely to select mostly positive genes related to the phenotype, but many will be rejected as false negatives. On the other hand, using a loose cutoff value for either method is prone to include a number of genes that are not phenotype-related, which are false positives. Thus, the research problem at hand is how to deal with the trade-off between false negatives and false positives. RESULTS We propose a novel framework called PONYTA for gene prioritization via positive-unlabeled (PU) learning on biological networks. Beginning with the selection of true phenotype-related genes using a rigorous cutoff value for DEG analysis and NP, we address the issue of handling false negatives by rescuing them through PU learning. Evaluations on transcriptome data from multiple studies show that our approach has superior gene prioritization ability compared to benchmark models. Therefore, PONYTA effectively prioritizes genes related to phenotypes derived from gene KO events and guides in vitro and in vivo gene KO experiments for increased efficiency. AVAILABILITY AND IMPLEMENTATION The source code of PONYTA is available at https://github.com/Jun-Hyeong-Kim/PONYTA.
Collapse
Affiliation(s)
- Jun Hyeong Kim
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul 08826, Republic of Korea
| | - Bonil Koo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul 08758, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul 08758, Republic of Korea
- Department of Computer Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
6
|
Sanches PHG, de Melo NC, Porcari AM, de Carvalho LM. Integrating Molecular Perspectives: Strategies for Comprehensive Multi-Omics Integrative Data Analysis and Machine Learning Applications in Transcriptomics, Proteomics, and Metabolomics. BIOLOGY 2024; 13:848. [PMID: 39596803 PMCID: PMC11592251 DOI: 10.3390/biology13110848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 11/29/2024]
Abstract
With the advent of high-throughput technologies, the field of omics has made significant strides in characterizing biological systems at various levels of complexity. Transcriptomics, proteomics, and metabolomics are the three most widely used omics technologies, each providing unique insights into different layers of a biological system. However, analyzing each omics data set separately may not provide a comprehensive understanding of the subject under study. Therefore, integrating multi-omics data has become increasingly important in bioinformatics research. In this article, we review strategies for integrating transcriptomics, proteomics, and metabolomics data, including co-expression analysis, metabolite-gene networks, constraint-based models, pathway enrichment analysis, and interactome analysis. We discuss combined omics integration approaches, correlation-based strategies, and machine learning techniques that utilize one or more types of omics data. By presenting these methods, we aim to provide researchers with a better understanding of how to integrate omics data to gain a more comprehensive view of a biological system, facilitating the identification of complex patterns and interactions that might be missed by single-omics analyses.
Collapse
Affiliation(s)
- Pedro H. Godoy Sanches
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Nicolly Clemente de Melo
- Graduate Program in Biomedicine, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Andreia M. Porcari
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Lucas Miguel de Carvalho
- Post Graduate Program in Health Sciences, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| |
Collapse
|
7
|
Hernández-Lemus E, Ochoa S. Methods for multi-omic data integration in cancer research. Front Genet 2024; 15:1425456. [PMID: 39364009 PMCID: PMC11446849 DOI: 10.3389/fgene.2024.1425456] [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: 04/29/2024] [Accepted: 08/28/2024] [Indexed: 10/05/2024] Open
Abstract
Multi-omics data integration is a term that refers to the process of combining and analyzing data from different omic experimental sources, such as genomics, transcriptomics, methylation assays, and microRNA sequencing, among others. Such data integration approaches have the potential to provide a more comprehensive functional understanding of biological systems and has numerous applications in areas such as disease diagnosis, prognosis and therapy. However, quantitative integration of multi-omic data is a complex task that requires the use of highly specialized methods and approaches. Here, we discuss a number of data integration methods that have been developed with multi-omics data in view, including statistical methods, machine learning approaches, and network-based approaches. We also discuss the challenges and limitations of such methods and provide examples of their applications in the literature. Overall, this review aims to provide an overview of the current state of the field and highlight potential directions for future research.
Collapse
Affiliation(s)
- Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Soledad Ochoa
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
8
|
Nam Y, Kim J, Jung SH, Woerner J, Suh EH, Lee DG, Shivakumar M, Lee ME, Kim D. Harnessing Artificial Intelligence in Multimodal Omics Data Integration: Paving the Path for the Next Frontier in Precision Medicine. Annu Rev Biomed Data Sci 2024; 7:225-250. [PMID: 38768397 PMCID: PMC11972123 DOI: 10.1146/annurev-biodatasci-102523-103801] [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: 05/22/2024]
Abstract
The integration of multiomics data with detailed phenotypic insights from electronic health records marks a paradigm shift in biomedical research, offering unparalleled holistic views into health and disease pathways. This review delineates the current landscape of multimodal omics data integration, emphasizing its transformative potential in generating a comprehensive understanding of complex biological systems. We explore robust methodologies for data integration, ranging from concatenation-based to transformation-based and network-based strategies, designed to harness the intricate nuances of diverse data types. Our discussion extends from incorporating large-scale population biobanks to dissecting high-dimensional omics layers at the single-cell level. The review underscores the emerging role of large language models in artificial intelligence, anticipating their influence as a near-future pivot in data integration approaches. Highlighting both achievements and hurdles, we advocate for a concerted effort toward sophisticated integration models, fortifying the foundation for groundbreaking discoveries in precision medicine.
Collapse
Affiliation(s)
- Yonghyun Nam
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Jaesik Kim
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Erica H Suh
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Dong-Gi Lee
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Matthew E Lee
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Dokyoon Kim
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
9
|
Cesnik A, Schaffer LV, Gaur I, Jain M, Ideker T, Lundberg E. Mapping the Multiscale Proteomic Organization of Cellular and Disease Phenotypes. Annu Rev Biomed Data Sci 2024; 7:369-389. [PMID: 38748859 PMCID: PMC11343683 DOI: 10.1146/annurev-biodatasci-102423-113534] [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: 06/23/2024]
Abstract
While the primary sequences of human proteins have been cataloged for over a decade, determining how these are organized into a dynamic collection of multiprotein assemblies, with structures and functions spanning biological scales, is an ongoing venture. Systematic and data-driven analyses of these higher-order structures are emerging, facilitating the discovery and understanding of cellular phenotypes. At present, knowledge of protein localization and function has been primarily derived from manual annotation and curation in resources such as the Gene Ontology, which are biased toward richly annotated genes in the literature. Here, we envision a future powered by data-driven mapping of protein assemblies. These maps can capture and decode cellular functions through the integration of protein expression, localization, and interaction data across length scales and timescales. In this review, we focus on progress toward constructing integrated cell maps that accelerate the life sciences and translational research.
Collapse
Affiliation(s)
- Anthony Cesnik
- Department of Bioengineering, Stanford University, Stanford, California, USA;
| | - Leah V Schaffer
- Department of Medicine, University of California San Diego, La Jolla, California, USA;
| | - Ishan Gaur
- Department of Bioengineering, Stanford University, Stanford, California, USA;
| | - Mayank Jain
- Department of Medicine, University of California San Diego, La Jolla, California, USA;
| | - Trey Ideker
- Departments of Computer Science and Engineering and Bioengineering, University of California San Diego, La Jolla, California, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA;
| | - Emma Lundberg
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Pathology, Stanford University, Palo Alto, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA;
| |
Collapse
|
10
|
Niu R, Guo Y, Shang X. GLIMS: A two-stage gradual-learning method for cancer genes prediction using multi-omics data and co-splicing network. iScience 2024; 27:109387. [PMID: 38510118 PMCID: PMC10951990 DOI: 10.1016/j.isci.2024.109387] [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: 09/26/2023] [Revised: 11/30/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
Identifying cancer genes is vital for cancer diagnosis and treatment. However, because of the complexity of cancer occurrence and limited cancer genes knowledge, it is hard to identify cancer genes accurately using only a few omics data, and the overall performance of existing methods is being called for further improvement. Here, we introduce a two-stage gradual-learning strategy GLIMS to predict cancer genes using integrative features from multi-omics data. Firstly, it uses a semi-supervised hierarchical graph neural network to predict the initial candidate cancer genes by integrating multi-omics data and protein-protein interaction (PPI) network. Then, it uses an unsupervised approach to further optimize the initial prediction by integrating the co-splicing network in post-transcriptional regulation, which plays an important role in cancer development. Systematic experiments on multi-omics cancer data demonstrated that GLIMS outperforms the state-of-the-art methods for the identification of cancer genes and it could be a useful tool to help advance cancer analysis.
Collapse
Affiliation(s)
- Rui Niu
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710129, China
| | - Yang Guo
- School of Information Science and Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710129, China
| |
Collapse
|
11
|
Song J, Song Z, Zhang J, Gong Y. Privacy-Preserving Identification of Cancer Subtype-Specific Driver Genes Based on Multigenomics Data with Privatedriver. J Comput Biol 2024; 31:99-116. [PMID: 38271572 DOI: 10.1089/cmb.2023.0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
Identifying cancer subtype-specific driver genes from a large number of irrelevant passengers is crucial for targeted therapy in cancer treatment. Recently, the rapid accumulation of large-scale cancer genomics data from multiple institutions has presented remarkable opportunities for identification of cancer subtype-specific driver genes. However, the insufficient subtype samples, privacy issues, and heterogenous of aberration events pose great challenges in precisely identifying cancer subtype-specific driver genes. To address this, we introduce privatedriver, the first model for identifying subtype-specific driver genes that integrates genomics data from multiple institutions in a data privacy-preserving collaboration manner. The process of identifying subtype-specific cancer driver genes using privatedriver involves the following two steps: genomics data integration and collaborative training. In the integration process, the aberration events from multiple genomics data sources are combined for each institution using the forward and backward propagation method of NetICS. In the collaborative training process, each institution utilizes the federated learning framework to upload encrypted model parameters instead of raw data of all institutions to train a global model by using the non-negative matrix factorization algorithm. We applied privatedriver on head and neck squamous cell and colon cancer from The Cancer Genome Atlas website and evaluated it with two benchmarks using macro-Fscore. The comparison analysis demonstrates that privatedriver achieves comparable results to centralized learning models and outperforms most other nonprivacy preserving models, all while ensuring the confidentiality of patient information. We also demonstrate that, for varying predicted driver gene distributions in subtype, our model fully considers the heterogeneity of subtype and identifies subtype-specific driver genes corresponding to the given prognosis and therapeutic effect. The success of privatedriver reveals the feasibility and effectiveness of identifying cancer subtype-specific driver genes in a data protection manner, providing new insights for future privacy-preserving driver gene identification studies.
Collapse
Affiliation(s)
- Junrong Song
- School of Information; Kunming, P.R. China
- Yunnan Key Laboratory of Service Computing; Yunnan University of Finance and Economics, Kunming, P.R. China
| | - Zhiming Song
- School of Information; Kunming, P.R. China
- Yunnan Key Laboratory of Service Computing; Yunnan University of Finance and Economics, Kunming, P.R. China
| | - Jinpeng Zhang
- School of Information; Kunming, P.R. China
- Yunnan Key Laboratory of Service Computing; Yunnan University of Finance and Economics, Kunming, P.R. China
- The School of Computer Science and Engineering, Yunnan University, Kunming, P.R. China
| | | |
Collapse
|
12
|
Sharma V, Singh A, Chauhan S, Sharma PK, Chaudhary S, Sharma A, Porwal O, Fuloria NK. Role of Artificial Intelligence in Drug Discovery and Target Identification in Cancer. Curr Drug Deliv 2024; 21:870-886. [PMID: 37670704 DOI: 10.2174/1567201821666230905090621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/08/2023] [Accepted: 03/24/2023] [Indexed: 09/07/2023]
Abstract
Drug discovery and development (DDD) is a highly complex process that necessitates precise monitoring and extensive data analysis at each stage. Furthermore, the DDD process is both timeconsuming and costly. To tackle these concerns, artificial intelligence (AI) technology can be used, which facilitates rapid and precise analysis of extensive datasets within a limited timeframe. The pathophysiology of cancer disease is complicated and requires extensive research for novel drug discovery and development. The first stage in the process of drug discovery and development involves identifying targets. Cell structure and molecular functioning are complex due to the vast number of molecules that function constantly, performing various roles. Furthermore, scientists are continually discovering novel cellular mechanisms and molecules, expanding the range of potential targets. Accurately identifying the correct target is a crucial step in the preparation of a treatment strategy. Various forms of AI, such as machine learning, neural-based learning, deep learning, and network-based learning, are currently being utilised in applications, online services, and databases. These technologies facilitate the identification and validation of targets, ultimately contributing to the success of projects. This review focuses on the different types and subcategories of AI databases utilised in the field of drug discovery and target identification for cancer.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Amit Singh
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Sanjana Chauhan
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Shubham Chaudhary
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Astha Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Omji Porwal
- Department of Pharmacognosy, Faculty of Pharmacy, Tishk International University, Erbil 44001, Iraq
| | | |
Collapse
|
13
|
Das S, Srivastava DK. ioSearch: An approach for identifying interacting multiomics biomarkers using a novel algorithm with application on breast cancer data sets. Genet Epidemiol 2023; 47:600-616. [PMID: 37795815 DOI: 10.1002/gepi.22536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 08/04/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
Identification of biomarkers by integrating multiple omics together is important because complex diseases occur due to an intricate interplay of various genetic materials. Traditional single-omics association tests neither explore this crucial interomics dependence nor identify moderately weak signals due to the multiple-testing burden. Conversely, multiomics data integration imparts complementary information but suffers from an increased multiple-testing burden, data diversity inherent with different omics features, high-dimensionality, and so forth. Most of the available methods address subtype classification using dimension-reduction techniques to circumvent the sample size issue but interacting multiomics biomarker identification methods are unavailable. We propose a two-step model that first investigates phenotype-omics association using logistic regression. Then, selects disease-associated omics using sparse principal components which explores the interrelationship of multiple variables from two omics in a multivariate multiple regression framework. On the basis of this model, we developed a multiomics biomarker identification algorithm, interacting omics search (ioSearch), that jointly tests the effect of multiple omics with disease and between-omics associations by using pathway information that subsequently reduces the multiple-testing burden. Further, inference in terms of p values potentially makes it an easily interpretable biomarker identification tool. Extensive simulation demonstrates ioSearch as statistically powerful with a controlled Type-I error rate. Its application to publicly available breast cancer data sets identified relevant omics features in important pathways.
Collapse
Affiliation(s)
- Sarmistha Das
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Deo Kumar Srivastava
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
14
|
Hernández Sánchez LF, Burger B, Castro Campos RA, Johansson S, Njølstad PR, Barsnes H, Vaudel M. Extending protein interaction networks using proteoforms and small molecules. Bioinformatics 2023; 39:btad598. [PMID: 37756698 PMCID: PMC10564616 DOI: 10.1093/bioinformatics/btad598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 09/29/2023] Open
Abstract
MOTIVATION Biological network analysis for high-throughput biomedical data interpretation relies heavily on topological characteristics. Networks are commonly composed of nodes representing genes or proteins that are connected by edges when interacting. In this study, we use the rich information available in the Reactome pathway database to build biological networks accounting for small molecules and proteoforms modeled using protein isoforms and post-translational modifications to study the topological changes induced by this refinement of the network representation. RESULTS We find that improving the interactome modeling increases the number of nodes and interactions, but that isoform and post-translational modification annotation is still limited compared to what can be expected biologically. We also note that small molecule information can distort the topology of the network due to the high connectedness of these molecules, which does not necessarily represent the reality of biology. However, by restricting the connections of small molecules to the context of biochemical reactions, we find that these improve the overall connectedness of the network and reduce the prevalence of isolated components and nodes. Overall, changing the representation of the network alters the prevalence of articulation points and bridges globally but also within and across pathways. Hence, some molecules can gain or lose in biological importance depending on the level of detail of the representation of the biological system, which might in turn impact network-based studies of diseases or druggability. AVAILABILITY AND IMPLEMENTATION Networks are constructed based on data publicly available in the Reactome Pathway knowledgebase: reactome.org.
Collapse
Affiliation(s)
- Luis Francisco Hernández Sánchez
- Department of Clinical Science, Mohn Center for Diabetes Precision Medicine, University of Bergen, Bergen 5020, Norway
- Department of Medical Genetics, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5020, Norway
| | - Bram Burger
- Department of Clinical Science, Mohn Center for Diabetes Precision Medicine, University of Bergen, Bergen 5020, Norway
- Department of Medical Genetics, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5020, Norway
- Department of Biomedicine, Proteomics Unit, University of Bergen, Bergen 5020, Norway
- Department of Informatics, Computational Biology Unit, University of Bergen, Bergen 5020, Norway
| | | | - Stefan Johansson
- Department of Clinical Science, Mohn Center for Diabetes Precision Medicine, University of Bergen, Bergen 5020, Norway
- Department of Medical Genetics, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5020, Norway
| | - Pål Rasmus Njølstad
- Department of Clinical Science, Mohn Center for Diabetes Precision Medicine, University of Bergen, Bergen 5020, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen 5020, Norway
| | - Harald Barsnes
- Department of Biomedicine, Proteomics Unit, University of Bergen, Bergen 5020, Norway
- Department of Informatics, Computational Biology Unit, University of Bergen, Bergen 5020, Norway
| | - Marc Vaudel
- Department of Clinical Science, Mohn Center for Diabetes Precision Medicine, University of Bergen, Bergen 5020, Norway
- Department of Informatics, Computational Biology Unit, University of Bergen, Bergen 5020, Norway
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo 0213, Norway
| |
Collapse
|
15
|
Jeong D, Koo B, Oh M, Kim TB, Kim S. GOAT: Gene-level biomarker discovery from multi-Omics data using graph ATtention neural network for eosinophilic asthma subtype. Bioinformatics 2023; 39:btad582. [PMID: 37740295 PMCID: PMC10547929 DOI: 10.1093/bioinformatics/btad582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 09/24/2023] Open
Abstract
MOTIVATION Asthma is a heterogeneous disease where various subtypes are established and molecular biomarkers of the subtypes are yet to be discovered. Recent availability of multi-omics data paved a way to discover molecular biomarkers for the subtypes. However, multi-omics biomarker discovery is challenging because of the complex interplay between different omics layers. RESULTS We propose a deep attention model named Gene-level biomarker discovery from multi-Omics data using graph ATtention neural network (GOAT) for identifying molecular biomarkers for eosinophilic asthma subtypes with multi-omics data. GOAT identifies genes that discriminate subtypes using a graph neural network by modeling complex interactions among genes as the attention mechanism in the deep learning model. In experiments with multi-omics profiles of the COREA (Cohort for Reality and Evolution of Adult Asthma in Korea) asthma cohort of 300 patients, GOAT outperforms existing models and suggests interpretable biological mechanisms underlying asthma subtypes. Importantly, GOAT identified genes that are distinct only in terms of relationship with other genes through attention. To better understand the role of biomarkers, we further investigated two transcription factors, CTNNB1 and JUN, captured by GOAT. We were successful in showing the role of the transcription factors in eosinophilic asthma pathophysiology in a network propagation and transcriptional network analysis, which were not distinct in terms of gene expression level differences. AVAILABILITY AND IMPLEMENTATION Source code is available https://github.com/DabinJeong/Multi-omics_biomarker. The preprocessed data underlying this article is accessible in data folder of the github repository. Raw data are available in Multi-Omics Platform at http://203.252.206.90:5566/, and it can be accessible when requested.
Collapse
Affiliation(s)
- Dabin Jeong
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
| | - Bonil Koo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd, Seoul 08826, Republic of Korea
| | - Minsik Oh
- School of Software Convergence, Myongji University, Seoul 03674, Republic of Korea
| | - Tae-Bum Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd, Seoul 08826, Republic of Korea
- Department of Computer Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Artificial Intelligence,, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
16
|
Peng W, Yu P, Dai W, Fu X, Liu L, Pan Y. A Graph Convolution Network-Based Model for Prioritizing Personalized Cancer Driver Genes of Individual Patients. IEEE Trans Nanobioscience 2023; 22:744-754. [PMID: 37195839 DOI: 10.1109/tnb.2023.3277316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Cancer driver genes are mutated genes that play a key role in the growth of cancer cells. Accurately identifying the cancer driver genes helps us understand cancer's pathogenesis and develop effective treatment strategies. However, cancers are highly heterogeneous diseases; patients with the same cancer type may have different genomic characteristics and clinical symptoms. Hence, it is urgent to devise effective methods to identify personalized cancer driver genes of individual patients to help determine whether a patient can be treated with a certain targeted drug. This work presents a method for predicting personalized cancer Driver genes of individual patients based on Graph Convolution Networks and Neighbor Interactions called NIGCNDriver. NIGCNDriver first constructs a gene-sample association matrix using the associations between a sample and its known driver genes. Then, it employs graph convolution models on the gene-sample network to aggregate neighbor node features, and themself features, and then combines with the element-wise level interactions between neighbors to learn new feature representations for the samples and gene nodes. Finally, a linear correlation coefficient decoder is used to reconstruct the association between the sample and the mutant gene, enabling the prediction of a personalized driver gene for the individual sample. We applied the NIGCNDriver method to predict cancer driver genes for individual samples in the TCGA and cancer cell line datasets. The results show that our method outperforms the baseline methods in cancer driver gene prediction for individual samples.
Collapse
|
17
|
Xiao S, Lin H, Wang C, Wang S, Rajapakse JC. Graph Neural Networks With Multiple Prior Knowledge for Multi-Omics Data Analysis. IEEE J Biomed Health Inform 2023; 27:4591-4600. [PMID: 37307177 DOI: 10.1109/jbhi.2023.3284794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
With the development of biotechnology, a large amount of multi-omics data have been collected for precision medicine. There exists multiple graph-based prior biological knowledge about omics data, such as gene-gene interaction networks. Recently, there has been an increasing interest in introducing graph neural networks (GNNs) into multi-omics learning. However, existing methods have not fully exploited these graphical priors since none have been able to integrate knowledge from multiple sources simultaneously. To solve this problem, we propose a multi-omics data analysis framework by incorporating multiple prior knowledge into graph neural network (MPK-GNN). To the best of our knowledge, this is the first attempt to introduce multiple prior graphs into multi-omics data analysis. Specifically, the proposed method contains four parts: (1) a feature-level learning module to aggregate information from prior graphs; (2) a projection module to maximize the agreement among prior networks by optimizing a contrastive loss; (3) a sample-level module to learn a global representation from input multi-omics features; (4) a task-specific module to flexibly extend MPK-GNN for various downstream multi-omics analysis tasks. Finally, we verify the effectiveness of the proposed multi-omics learning algorithm on the cancer molecular subtype classification task. Experimental results show that MPK-GNN outperforms other state-of-the-art algorithms, including multi-view learning methods and multi-omics integrative approaches.
Collapse
|
18
|
Demir Karaman E, Işık Z. Multi-Omics Data Analysis Identifies Prognostic Biomarkers across Cancers. Med Sci (Basel) 2023; 11:44. [PMID: 37489460 PMCID: PMC10366886 DOI: 10.3390/medsci11030044] [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: 05/19/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Combining omics data from different layers using integrative methods provides a better understanding of the biology of a complex disease such as cancer. The discovery of biomarkers related to cancer development or prognosis helps to find more effective treatment options. This study integrates multi-omics data of different cancer types with a network-based approach to explore common gene modules among different tumors by running community detection methods on the integrated network. The common modules were evaluated by several biological metrics adapted to cancer. Then, a new prognostic scoring method was developed by weighting mRNA expression, methylation, and mutation status of genes. The survival analysis pointed out statistically significant results for GNG11, CBX2, CDKN3, ARHGEF10, CLN8, SEC61G and PTDSS1 genes. The literature search reveals that the identified biomarkers are associated with the same or different types of cancers. Our method does not only identify known cancer-specific biomarker genes, but also proposes new potential biomarkers. Thus, this study provides a rationale for identifying new gene targets and expanding treatment options across cancer types.
Collapse
Affiliation(s)
- Ezgi Demir Karaman
- Department of Computer Engineering, Institute of Natural and Applied Sciences, Dokuz Eylul University, Izmir 35390, Turkey
| | - Zerrin Işık
- Department of Computer Engineering, Faculty of Engineering, Dokuz Eylul University, Izmir 35390, Turkey
| |
Collapse
|
19
|
Xu Z, Marchionni L, Wang S. MultiNEP: a multi-omics network enhancement framework for prioritizing disease genes and metabolites simultaneously. Bioinformatics 2023; 39:btad333. [PMID: 37216914 PMCID: PMC10250081 DOI: 10.1093/bioinformatics/btad333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023] Open
Abstract
MOTIVATION Many studies have successfully used network information to prioritize candidate omics profiles associated with diseases. The metabolome, as the link between genotypes and phenotypes, has accumulated growing attention. Using a "multi-omics" network constructed with a gene-gene network, a metabolite-metabolite network, and a gene-metabolite network to simultaneously prioritize candidate disease-associated metabolites and gene expressions could further utilize gene-metabolite interactions that are not used when prioritizing them separately. However, the number of metabolites is usually 100 times fewer than that of genes. Without accounting for this imbalance issue, we cannot effectively use gene-metabolite interactions when simultaneously prioritizing disease-associated metabolites and genes. RESULTS Here, we developed a Multi-omics Network Enhancement Prioritization (MultiNEP) framework with a weighting scheme to reweight contributions of different sub-networks in a multi-omics network to effectively prioritize candidate disease-associated metabolites and genes simultaneously. In simulation studies, MultiNEP outperforms competing methods that do not address network imbalances and identifies more true signal genes and metabolites simultaneously when we down-weight relative contributions of the gene-gene network and up-weight that of the metabolite-metabolite network to the gene-metabolite network. Applications to two human cancer cohorts show that MultiNEP prioritizes more cancer-related genes by effectively using both within- and between-omics interactions after handling network imbalance. AVAILABILITY AND IMPLEMENTATION The developed MultiNEP framework is implemented in an R package and available at: https://github.com/Karenxzr/MultiNep.
Collapse
Affiliation(s)
- Zhuoran Xu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Shuang Wang
- Department of Biostatistics, Columbia University, New York, NY 10032, United States
| |
Collapse
|
20
|
Unlu Yazici M, Marron JS, Bakir-Gungor B, Zou F, Yousef M. Invention of 3Mint for feature grouping and scoring in multi-omics. Front Genet 2023; 14:1093326. [PMID: 37007972 PMCID: PMC10050723 DOI: 10.3389/fgene.2023.1093326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
Advanced genomic and molecular profiling technologies accelerated the enlightenment of the regulatory mechanisms behind cancer development and progression, and the targeted therapies in patients. Along this line, intense studies with immense amounts of biological information have boosted the discovery of molecular biomarkers. Cancer is one of the leading causes of death around the world in recent years. Elucidation of genomic and epigenetic factors in Breast Cancer (BRCA) can provide a roadmap to uncover the disease mechanisms. Accordingly, unraveling the possible systematic connections between-omics data types and their contribution to BRCA tumor progression is crucial. In this study, we have developed a novel machine learning (ML) based integrative approach for multi-omics data analysis. This integrative approach combines information from gene expression (mRNA), microRNA (miRNA) and methylation data. Due to the complexity of cancer, this integrated data is expected to improve the prediction, diagnosis and treatment of disease through patterns only available from the 3-way interactions between these 3-omics datasets. In addition, the proposed method bridges the interpretation gap between the disease mechanisms that drive onset and progression. Our fundamental contribution is the 3 Multi-omics integrative tool (3Mint). This tool aims to perform grouping and scoring of groups using biological knowledge. Another major goal is improved gene selection via detection of novel groups of cross-omics biomarkers. Performance of 3Mint is assessed using different metrics. Our computational performance evaluations showed that the 3Mint classifies the BRCA molecular subtypes with lower number of genes when compared to the miRcorrNet tool which uses miRNA and mRNA gene expression profiles in terms of similar performance metrics (95% Accuracy). The incorporation of methylation data in 3Mint yields a much more focused analysis. The 3Mint tool and all other supplementary files are available at https://github.com/malikyousef/3Mint/.
Collapse
Affiliation(s)
- Miray Unlu Yazici
- Department of Bioengineering, Abdullah Gül University, Kayseri, Türkiye
| | - J. S. Marron
- Department of Statistics and Operations Research, University of North Carolina, Chapel Hill, NC, United States
| | - Burcu Bakir-Gungor
- Department of Bioengineering, Abdullah Gül University, Kayseri, Türkiye
- Department of Computer Engineering, Abdullah Gul University, Kayseri, Türkiye
| | - Fei Zou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Malik Yousef
- Department of Information Systems, Zefat Academic College, Zefat, Israel
- Galilee Digital Health Research Center, Zefat Academic College, Zefat, Israel
- *Correspondence: Malik Yousef,
| |
Collapse
|
21
|
Chen X, Han M, Li Y, Li X, Zhang J, Zhu Y. Identification of functional gene modules by integrating multi-omics data and known molecular interactions. Front Genet 2023; 14:1082032. [PMID: 36760999 PMCID: PMC9902936 DOI: 10.3389/fgene.2023.1082032] [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: 10/27/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023] Open
Abstract
Multi-omics data integration has emerged as a promising approach to identify patient subgroups. However, in terms of grouping genes (or gene products) into co-expression modules, data integration methods suffer from two main drawbacks. First, most existing methods only consider genes or samples measured in all different datasets. Second, known molecular interactions (e.g., transcriptional regulatory interactions, protein-protein interactions and biological pathways) cannot be utilized to assist in module detection. Herein, we present a novel data integration framework, Correlation-based Local Approximation of Membership (CLAM), which provides two methodological innovations to address these limitations: 1) constructing a trans-omics neighborhood matrix by integrating multi-omics datasets and known molecular interactions, and 2) using a local approximation procedure to define gene modules from the matrix. Applying Correlation-based Local Approximation of Membership to human colorectal cancer (CRC) and mouse B-cell differentiation multi-omics data obtained from The Cancer Genome Atlas (TCGA), Clinical Proteomics Tumor Analysis Consortium (CPTAC), Gene Expression Omnibus (GEO) and ProteomeXchange database, we demonstrated its superior ability to recover biologically relevant modules and gene ontology (GO) terms. Further investigation of the colorectal cancer modules revealed numerous transcription factors and KEGG pathways that played crucial roles in colorectal cancer progression. Module-based survival analysis constructed four survival-related networks in which pairwise gene correlations were significantly correlated with colorectal cancer patient survival. Overall, the series of evaluations demonstrated the great potential of Correlation-based Local Approximation of Membership for identifying modular biomarkers for complex diseases. We implemented Correlation-based Local Approximation of Membership as a user-friendly application available at https://github.com/free1234hm/CLAM.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Basic Medical School, Anhui Medical University, Hefei, China,National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Mingfei Han
- National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yingxing Li
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Li
- National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Jiaqi Zhang
- National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yunping Zhu
- Basic Medical School, Anhui Medical University, Hefei, China,National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China,*Correspondence: Yunping Zhu,
| |
Collapse
|
22
|
Liu XY, Mei XY. Prediction of drug sensitivity based on multi-omics data using deep learning and similarity network fusion approaches. Front Bioeng Biotechnol 2023; 11:1156372. [PMID: 37139048 PMCID: PMC10150883 DOI: 10.3389/fbioe.2023.1156372] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
With the rapid development of multi-omics technologies and accumulation of large-scale bio-datasets, many studies have conducted a more comprehensive understanding of human diseases and drug sensitivity from multiple biomolecules, such as DNA, RNA, proteins and metabolites. Using single omics data is difficult to systematically and comprehensively analyze the complex disease pathology and drug pharmacology. The molecularly targeted therapy-based approaches face some challenges, such as insufficient target gene labeling ability, and no clear targets for non-specific chemotherapeutic drugs. Consequently, the integrated analysis of multi-omics data has become a new direction for scientists to explore the mechanism of disease and drug. However, the available drug sensitivity prediction models based on multi-omics data still have problems such as overfitting, lack of interpretability, difficulties in integrating heterogeneous data, and the prediction accuracy needs to be improved. In this paper, we proposed a novel drug sensitivity prediction (NDSP) model based on deep learning and similarity network fusion approaches, which extracts drug targets using an improved sparse principal component analysis (SPCA) method for each omics data, and construct sample similarity networks based on the sparse feature matrices. Furthermore, the fused similarity networks are put into a deep neural network for training, which greatly reduces the data dimensionality and weakens the risk of overfitting problem. We use three omics of data, RNA sequence, copy number aberration and methylation, and select 35 drugs from Genomics of Drug Sensitivity in Cancer (GDSC) for experiments, including Food and Drug Administration (FDA)-approved targeted drugs, FDA-unapproved targeted drugs and non-specific therapies. Compared with some current deep learning methods, our proposed method can extract highly interpretable biological features to achieve highly accurate sensitivity prediction of targeted and non-specific cancer drugs, which is beneficial for the development of precision oncology beyond targeted therapy.
Collapse
Affiliation(s)
- Xiao-Ying Liu
- Guangdong Polytechnic of Science and Technology, Zhuhai, China
- *Correspondence: Xiao-Ying Liu,
| | - Xin-Yue Mei
- Institute of Systems Engineering, Macau University of Science and Technology, Taipa, China
| |
Collapse
|
23
|
Vu T, Litkowski EM, Liu W, Pratte KA, Lange L, Bowler RP, Banaei-Kashani F, Kechris KJ. NetSHy: network summarization via a hybrid approach leveraging topological properties. Bioinformatics 2023; 39:6957083. [PMID: 36548341 PMCID: PMC9831052 DOI: 10.1093/bioinformatics/btac818] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/30/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Biological networks can provide a system-level understanding of underlying processes. In many contexts, networks have a high degree of modularity, i.e. they consist of subsets of nodes, often known as subnetworks or modules, which are highly interconnected and may perform separate functions. In order to perform subsequent analyses to investigate the association between the identified module and a variable of interest, a module summarization, that best explains the module's information and reduces dimensionality is often needed. Conventional approaches for obtaining network representation typically rely only on the profiles of the nodes within the network while disregarding the inherent network topological information. RESULTS In this article, we propose NetSHy, a hybrid approach which is capable of reducing the dimension of a network while incorporating topological properties to aid the interpretation of the downstream analyses. In particular, NetSHy applies principal component analysis (PCA) on a combination of the node profiles and the well-known Laplacian matrix derived directly from the network similarity matrix to extract a summarization at a subject level. Simulation scenarios based on random and empirical networks at varying network sizes and sparsity levels show that NetSHy outperforms the conventional PCA approach applied directly on node profiles, in terms of recovering the true correlation with a phenotype of interest and maintaining a higher amount of explained variation in the data when networks are relatively sparse. The robustness of NetSHy is also demonstrated by a more consistent correlation with the observed phenotype as the sample size decreases. Lastly, a genome-wide association study is performed as an application of a downstream analysis, where NetSHy summarization scores on the biological networks identify more significant single nucleotide polymorphisms than the conventional network representation. AVAILABILITY AND IMPLEMENTATION R code implementation of NetSHy is available at https://github.com/thaovu1/NetSHy. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Thao Vu
- To whom correspondence should be addressed. or
| | - Elizabeth M Litkowski
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Biomedical Informatics & Personalized Medicine, School of Medicine, Colorado University Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Weixuan Liu
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katherine A Pratte
- Department of Biostatistics, National Jewish Health, Denver, CO 80206, USA
| | - Leslie Lange
- Division of Biomedical Informatics & Personalized Medicine, School of Medicine, Colorado University Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Russell P Bowler
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Farnoush Banaei-Kashani
- Department of Computer Science and Engineering, College of Engineering, Design and Computing, University of Colorado Denver, Denver, CO 80204, USA
| | | |
Collapse
|
24
|
Tanvir Ahmed K, Cheng S, Li Q, Yong J, Zhang W. Incomplete time-series gene expression in integrative study for islet autoimmunity prediction. Brief Bioinform 2022; 24:6895461. [PMID: 36513375 PMCID: PMC9851333 DOI: 10.1093/bib/bbac537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) outcome prediction plays a vital role in identifying novel risk factors, ensuring early patient care and designing cohort studies. TEDDY is a longitudinal cohort study that collects a vast amount of multi-omics and clinical data from its participants to explore the progression and markers of T1D. However, missing data in the omics profiles make the outcome prediction a difficult task. TEDDY collected time series gene expression for less than 6% of enrolled participants. Additionally, for the participants whose gene expressions are collected, 79% time steps are missing. This study introduces an advanced bioinformatics framework for gene expression imputation and islet autoimmunity (IA) prediction. The imputation model generates synthetic data for participants with partially or entirely missing gene expression. The prediction model integrates the synthetic gene expression with other risk factors to achieve better predictive performance. Comprehensive experiments on TEDDY datasets show that: (1) Our pipeline can effectively integrate synthetic gene expression with family history, HLA genotype and SNPs to better predict IA status at 2 years (sensitivity 0.622, AUC 0.715) compared with the individual datasets and state-of-the-art results in the literature (AUC 0.682). (2) The synthetic gene expression contains predictive signals as strong as the true gene expression, reducing reliance on expensive and long-term longitudinal data collection. (3) Time series gene expression is crucial to the proposed improvement and shows significantly better predictive ability than cross-sectional gene expression. (4) Our pipeline is robust to limited data availability. Availability: Code is available at https://github.com/compbiolabucf/TEDDY.
Collapse
Affiliation(s)
| | - Sze Cheng
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Qian Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jeongsik Yong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Wei Zhang
- Corresponding author. Wei Zhang, Computer Science Department, University of Central Florida. Tel.: 407-823-2763;
| |
Collapse
|
25
|
Alfatemi A, Peng H, Rong W, Zhang B, Cai H. Patient subgrouping with distinct survival rates via integration of multiomics data on a Grassmann manifold. BMC Med Inform Decis Mak 2022; 22:190. [PMID: 35870923 PMCID: PMC9308936 DOI: 10.1186/s12911-022-01938-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
Background Patient subgroups are important for easily understanding a disease and for providing precise yet personalized treatment through multiple omics dataset integration. Multiomics datasets are produced daily. Thus, the fusion of heterogeneous big data into intrinsic structures is an urgent problem. Novel mathematical methods are needed to process these data in a straightforward way. Results We developed a novel method for subgrouping patients with distinct survival rates via the integration of multiple omics datasets and by using principal component analysis to reduce the high data dimensionality. Then, we constructed similarity graphs for patients, merged the graphs in a subspace, and analyzed them on a Grassmann manifold. The proposed method could identify patient subgroups that had not been reported previously by selecting the most critical information during the merging at each level of the omics dataset. Our method was tested on empirical multiomics datasets from The Cancer Genome Atlas. Conclusion Through the integration of microRNA, gene expression, and DNA methylation data, our method accurately identified patient subgroups and achieved superior performance compared with popular methods. Supplementary Information The online version contains supplementary material available at 10.1186/s12911-022-01938-y.
Collapse
|
26
|
Zhang L, Fan S, Vera J, Lai X. A network medicine approach for identifying diagnostic and prognostic biomarkers and exploring drug repurposing in human cancer. Comput Struct Biotechnol J 2022; 21:34-45. [PMID: 36514340 PMCID: PMC9732137 DOI: 10.1016/j.csbj.2022.11.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer is a heterogeneous disease mainly driven by abnormal gene perturbations in regulatory networks. Therefore, it is appealing to identify the common and specific perturbed genes from multiple cancer networks. We developed an integrative network medicine approach to identify novel biomarkers and investigate drug repurposing across cancer types. We used a network-based method to prioritize genes in cancer-specific networks reconstructed using human transcriptome and interactome data. The prioritized genes show extensive perturbation and strong regulatory interaction with other highly perturbed genes, suggesting their vital contribution to tumorigenesis and tumor progression, and are therefore regarded as cancer genes. The cancer genes detected show remarkable performances in discriminating tumors from normal tissues and predicting survival times of cancer patients. Finally, we developed a network proximity approach to systematically screen drugs and identified dozens of candidates with repurposable potential in several cancer types. Taken together, we demonstrated the power of the network medicine approach to identify novel biomarkers and repurposable drugs in multiple cancer types. We have also made the data and code freely accessible to ensure reproducibility and reusability of the developed computational workflow.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer Science, Sichuan University, Chengdu, China
| | - Shiwei Fan
- College of Computer Science, Sichuan University, Chengdu, China
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany,Deutsches Zentrum Immuntherapie, Erlangen, Germany,Comprehensive Cancer Center Erlangen, Erlangen, Germany
| | - Xin Lai
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany,Deutsches Zentrum Immuntherapie, Erlangen, Germany,Comprehensive Cancer Center Erlangen, Erlangen, Germany,BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland,Corresponding author at: Universitätsklinikum Erlangen, Erlangen, Germany; Tampere University, Tampere, Finland.
| |
Collapse
|
27
|
Agamah FE, Bayjanov JR, Niehues A, Njoku KF, Skelton M, Mazandu GK, Ederveen THA, Mulder N, Chimusa ER, 't Hoen PAC. Computational approaches for network-based integrative multi-omics analysis. Front Mol Biosci 2022; 9:967205. [PMID: 36452456 PMCID: PMC9703081 DOI: 10.3389/fmolb.2022.967205] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/20/2022] [Indexed: 08/27/2023] Open
Abstract
Advances in omics technologies allow for holistic studies into biological systems. These studies rely on integrative data analysis techniques to obtain a comprehensive view of the dynamics of cellular processes, and molecular mechanisms. Network-based integrative approaches have revolutionized multi-omics analysis by providing the framework to represent interactions between multiple different omics-layers in a graph, which may faithfully reflect the molecular wiring in a cell. Here we review network-based multi-omics/multi-modal integrative analytical approaches. We classify these approaches according to the type of omics data supported, the methods and/or algorithms implemented, their node and/or edge weighting components, and their ability to identify key nodes and subnetworks. We show how these approaches can be used to identify biomarkers, disease subtypes, crosstalk, causality, and molecular drivers of physiological and pathological mechanisms. We provide insight into the most appropriate methods and tools for research questions as showcased around the aetiology and treatment of COVID-19 that can be informed by multi-omics data integration. We conclude with an overview of challenges associated with multi-omics network-based analysis, such as reproducibility, heterogeneity, (biological) interpretability of the results, and we highlight some future directions for network-based integration.
Collapse
Affiliation(s)
- Francis E. Agamah
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jumamurat R. Bayjanov
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anna Niehues
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kelechi F. Njoku
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gaston K. Mazandu
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- African Institute for Mathematical Sciences, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. 't Hoen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
28
|
Azadifar S, Ahmadi A. A novel candidate disease gene prioritization method using deep graph convolutional networks and semi-supervised learning. BMC Bioinformatics 2022; 23:422. [PMID: 36241966 PMCID: PMC9563530 DOI: 10.1186/s12859-022-04954-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Selecting and prioritizing candidate disease genes is necessary before conducting laboratory studies as identifying disease genes from a large number of candidate genes using laboratory methods, is a very costly and time-consuming task. There are many machine learning-based gene prioritization methods. These methods differ in various aspects including the feature vectors of genes, the used datasets with different structures, and the learning model. Creating a suitable feature vector for genes and an appropriate learning model on a variety of data with different and non-Euclidean structures, including graphs, as well as the lack of negative data are very important challenges of these methods. The use of graph neural networks has recently emerged in machine learning and other related fields, and they have demonstrated superior performance for a broad range of problems. METHODS In this study, a new semi-supervised learning method based on graph convolutional networks is presented using the novel constructing feature vector for each gene. In the proposed method, first, we construct three feature vectors for each gene using terms from the Gene Ontology (GO) database. Then, we train a graph convolution network on these vectors using protein-protein interaction (PPI) network data to identify disease candidate genes. Our model discovers hidden layer representations encoding in both local graph structure as well as features of nodes. This method is characterized by the simultaneous consideration of topological information of the biological network (e.g., PPI) and other sources of evidence. Finally, a validation has been done to demonstrate the efficiency of our method. RESULTS Several experiments are performed on 16 diseases to evaluate the proposed method's performance. The experiments demonstrate that our proposed method achieves the best results, in terms of precision, the area under the ROC curve (AUCs), and F1-score values, when compared with eight state-of-the-art network and machine learning-based disease gene prioritization methods. CONCLUSION This study shows that the proposed semi-supervised learning method appropriately classifies and ranks candidate disease genes using a graph convolutional network and an innovative method to create three feature vectors for genes based on the molecular function, cellular component, and biological process terms from GO data.
Collapse
Affiliation(s)
- Saeid Azadifar
- Faculty of Computer Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Ali Ahmadi
- Faculty of Computer Engineering, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
29
|
Hiort P, Hugo J, Zeinert J, Müller N, Kashyap S, Rajapakse JC, Azuaje F, Renard BY, Baum K. DrDimont: explainable drug response prediction from differential analysis of multi-omics networks. Bioinformatics 2022; 38:ii113-ii119. [PMID: 36124784 PMCID: PMC9486584 DOI: 10.1093/bioinformatics/btac477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MOTIVATION While it has been well established that drugs affect and help patients differently, personalized drug response predictions remain challenging. Solutions based on single omics measurements have been proposed, and networks provide means to incorporate molecular interactions into reasoning. However, how to integrate the wealth of information contained in multiple omics layers still poses a complex problem. RESULTS We present DrDimont, Drug response prediction from Differential analysis of multi-omics networks. It allows for comparative conclusions between two conditions and translates them into differential drug response predictions. DrDimont focuses on molecular interactions. It establishes condition-specific networks from correlation within an omics layer that are then reduced and combined into heterogeneous, multi-omics molecular networks. A novel semi-local, path-based integration step ensures integrative conclusions. Differential predictions are derived from comparing the condition-specific integrated networks. DrDimont's predictions are explainable, i.e. molecular differences that are the source of high differential drug scores can be retrieved. We predict differential drug response in breast cancer using transcriptomics, proteomics, phosphosite and metabolomics measurements and contrast estrogen receptor positive and receptor negative patients. DrDimont performs better than drug prediction based on differential protein expression or PageRank when evaluating it on ground truth data from cancer cell lines. We find proteomic and phosphosite layers to carry most information for distinguishing drug response. AVAILABILITY AND IMPLEMENTATION DrDimont is available on CRAN: https://cran.r-project.org/package=DrDimont. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Pauline Hiort
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam 14482, Germany
| | - Julian Hugo
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam 14482, Germany
| | - Justus Zeinert
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam 14482, Germany
| | - Nataniel Müller
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam 14482, Germany
| | - Spoorthi Kashyap
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam 14482, Germany
| | - Jagath C Rajapakse
- School of Computer Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | | | - Bernhard Y Renard
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam 14482, Germany
| | | |
Collapse
|
30
|
Mercatelli D, Cabrelle C, Veltri P, Giorgi FM, Guzzi PH. Detection of pan-cancer surface protein biomarkers via a network-based approach on transcriptomics data. Brief Bioinform 2022; 23:6695270. [DOI: 10.1093/bib/bbac400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Cell surface proteins have been used as diagnostic and prognostic markers in cancer research and as targets for the development of anticancer agents. Many of these proteins lie at the top of signaling cascades regulating cell responses and gene expression, therefore acting as ‘signaling hubs’. It has been previously demonstrated that the integrated network analysis on transcriptomic data is able to infer cell surface protein activity in breast cancer. Such an approach has been implemented in a publicly available method called ‘SURFACER’. SURFACER implements a network-based analysis of transcriptomic data focusing on the overall activity of curated surface proteins, with the final aim to identify those proteins driving major phenotypic changes at a network level, named surface signaling hubs. Here, we show the ability of SURFACER to discover relevant knowledge within and across cancer datasets. We also show how different cancers can be stratified in surface-activity-specific groups. Our strategy may identify cancer-wide markers to design targeted therapies and biomarker-based diagnostic approaches.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna , 40138 Bologna , Italy
| | - Chiara Cabrelle
- Department of Pharmacy and Biotechnology, University of Bologna , 40138 Bologna , Italy
| | - Pierangelo Veltri
- Department of Surgical and Medical Sciences, Magna Graecia University , 88100 Catanzaro , Italy
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna , 40138 Bologna , Italy
| | - Pietro H Guzzi
- Department of Surgical and Medical Sciences, Magna Graecia University , 88100 Catanzaro , Italy
| |
Collapse
|
31
|
Suter P, Dazert E, Kuipers J, Ng CKY, Boldanova T, Hall MN, Heim MH, Beerenwinkel N. Multi-omics subtyping of hepatocellular carcinoma patients using a Bayesian network mixture model. PLoS Comput Biol 2022; 18:e1009767. [PMID: 36067230 PMCID: PMC9481159 DOI: 10.1371/journal.pcbi.1009767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/16/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Comprehensive molecular characterization of cancer subtypes is essential for predicting clinical outcomes and searching for personalized treatments. We present bnClustOmics, a statistical model and computational tool for multi-omics unsupervised clustering, which serves a dual purpose: Clustering patient samples based on a Bayesian network mixture model and learning the networks of omics variables representing these clusters. The discovered networks encode interactions among all omics variables and provide a molecular characterization of each patient subgroup. We conducted simulation studies that demonstrated the advantages of our approach compared to other clustering methods in the case where the generative model is a mixture of Bayesian networks. We applied bnClustOmics to a hepatocellular carcinoma (HCC) dataset comprising genome (mutation and copy number), transcriptome, proteome, and phosphoproteome data. We identified three main HCC subtypes together with molecular characteristics, some of which are associated with survival even when adjusting for the clinical stage. Cluster-specific networks shed light on the links between genotypes and molecular phenotypes of samples within their respective clusters and suggest targets for personalized treatments.
Collapse
Affiliation(s)
- Polina Suter
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, Basel, Switzerland
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Charlotte K. Y. Ng
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tuyana Boldanova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Markus H. Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
32
|
Gentili M, Martini L, Sponziello M, Becchetti L. Biological Random Walks: multi-omics integration for disease gene prioritization. Bioinformatics 2022; 38:4145-4152. [PMID: 35792834 DOI: 10.1093/bioinformatics/btac446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 06/22/2022] [Accepted: 07/05/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Over the past decade, network-based approaches have proven useful in identifying disease modules within the human interactome, often providing insights into key mechanisms and guiding the quest for therapeutic targets. This is all the more important, since experimental investigation of potential gene candidates is an expensive task, thus not always a feasible option. On the other hand, many sources of biological information exist beyond the interactome and an important research direction is the design of effective techniques for their integration. RESULTS In this work, we introduce the Biological Random Walks (BRW) approach for disease gene prioritization in the human interactome. The proposed framework leverages multiple biological sources within an integrated framework. We perform an extensive, comparative study of BRW's performance against well-established baselines. AVAILABILITY AND IMPLEMENTATION All codes are publicly available and can be downloaded at https://github.com/LeoM93/BiologicalRandomWalks. We used publicly available datasets, details on their retrieval and preprocessing are provided in the Supplementary Material. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Michele Gentili
- Department of Computer, Control, and Management Engineering Antonio Ruberti, Sapienza University of Rome, Rome, Italy
| | - Leonardo Martini
- Department of Computer, Control, and Management Engineering Antonio Ruberti, Sapienza University of Rome, Rome, Italy
| | - Marialuisa Sponziello
- Translational and Precision Medicine Department, Sapienza University of Rome, Rome, Italy
| | - Luca Becchetti
- Department of Computer, Control, and Management Engineering Antonio Ruberti, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Guo X, Han J, Song Y, Yin Z, Liu S, Shang X. Using expression quantitative trait loci data and graph-embedded neural networks to uncover genotype–phenotype interactions. Front Genet 2022; 13:921775. [PMID: 36046233 PMCID: PMC9421127 DOI: 10.3389/fgene.2022.921775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Motivation: A central goal of current biology is to establish a complete functional link between the genotype and phenotype, known as the so-called genotype–phenotype map. With the continuous development of high-throughput technology and the decline in sequencing costs, multi-omics analysis has become more widely employed. While this gives us new opportunities to uncover the correlation mechanisms between single-nucleotide polymorphism (SNP), genes, and phenotypes, multi-omics still faces certain challenges, specifically: 1) When the sample size is large enough, the number of omics types is often not large enough to meet the requirements of multi-omics analysis; 2) each omics’ internal correlations are often unclear, such as the correlation between genes in genomics; 3) when analyzing a large number of traits (p), the sample size (n) is often smaller than p, n << p, hindering the application of machine learning methods in the classification of disease outcomes.Results: To solve these issues with multi-omics and build a robust classification model, we propose a graph-embedded deep neural network (G-EDNN) based on expression quantitative trait loci (eQTL) data, which achieves sparse connectivity between network layers to prevent overfitting. The correlation within each omics is also considered such that the model more closely resembles biological reality. To verify the capabilities of this method, we conducted experimental analysis using the GSE28127 and GSE95496 data sets from the Gene Expression Omnibus (GEO) database, tested various neural network architectures, and used prior data for feature selection and graph embedding. Results show that the proposed method could achieve a high classification accuracy and easy-to-interpret feature selection. This method represents an extended application of genotype–phenotype association analysis in deep learning networks.
Collapse
Affiliation(s)
- Xinpeng Guo
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, China
| | - Jinyu Han
- School of Economics and Management, Chang ‘an University, Xi’an, China
| | - Yafei Song
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, China
| | - Zhilei Yin
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Shuaichen Liu
- School of Marine Science and Technology, Northwestern Polytechnical University, Xi’an, China
| | - Xuequn Shang
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Xuequn Shang,
| |
Collapse
|
34
|
Zhang S, Sun X, Mou M, Amahong K, Sun H, Zhang W, Shi S, Li Z, Gao J, Zhu F. REGLIV: Molecular regulation data of diverse living systems facilitating current multiomics research. Comput Biol Med 2022; 148:105825. [PMID: 35872412 DOI: 10.1016/j.compbiomed.2022.105825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022]
Abstract
Multiomics is a powerful technique in molecular biology that facilitates the identification of new associations among different molecules (genes, proteins & metabolites). It has attracted tremendous research interest from the scientists worldwide and has led to an explosive number of published studies. Most of these studies are based on the regulation data provided in available databases. Therefore, it is essential to have molecular regulation data that are strictly validated in the living systems of various cell lines and in vivo models. However, no database has been developed yet to provide comprehensive molecular regulation information validated by living systems. Herein, a new database, Molecular Regulation Data of Living System Facilitating Multiomics Study (REGLIV) is introduced to describe various types of molecular regulation tested by the living systems. (1) A total of 2996 regulations describe the changes in 1109 metabolites triggered by alterations in 284 genes or proteins, and (2) 1179 regulations describe the variations in 926 proteins induced by 125 endogenous metabolites. Overall, REGLIV is unique in (a) providing the molecular regulation of a clearly defined regulatory direction other than simple correlation, (b) focusing on molecular regulations that are validated in a living system not simply in an in vitro test, and (c) describing the disease/tissue/species specific property underlying each regulation. Therefore, REGLIV has important implications for the future practice of not only multiomics, but also other fields relevant to molecular regulation. REGLIV is freely accessible at: https://idrblab.org/regliv/.
Collapse
Affiliation(s)
- Song Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Kuerbannisha Amahong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shuiyang Shi
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhaorong Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China.
| |
Collapse
|
35
|
Bhardwaj A, Josse C, Van Daele D, Poulet C, Chavez M, Struman I, Van Steen K. Deeper insights into long-term survival heterogeneity of pancreatic ductal adenocarcinoma (PDAC) patients using integrative individual- and group-level transcriptome network analyses. Sci Rep 2022; 12:11027. [PMID: 35773268 PMCID: PMC9247075 DOI: 10.1038/s41598-022-14592-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 06/09/2022] [Indexed: 11/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is categorized as the leading cause of cancer mortality worldwide. However, its predictive markers for long-term survival are not well known. It is interesting to delineate individual-specific perturbed genes when comparing long-term (LT) and short-term (ST) PDAC survivors and integrate individual- and group-based transcriptome profiling. Using a discovery cohort of 19 PDAC patients from CHU-Liège (Belgium), we first performed differential gene expression analysis comparing LT to ST survivor. Second, we adopted systems biology approaches to obtain clinically relevant gene modules. Third, we created individual-specific perturbation profiles. Furthermore, we used Degree-Aware disease gene prioritizing (DADA) method to develop PDAC disease modules; Network-based Integration of Multi-omics Data (NetICS) to integrate group-based and individual-specific perturbed genes in relation to PDAC LT survival. We identified 173 differentially expressed genes (DEGs) in ST and LT survivors and five modules (including 38 DEGs) showing associations to clinical traits. Validation of DEGs in the molecular lab suggested a role of REG4 and TSPAN8 in PDAC survival. Via NetICS and DADA, we identified various known oncogenes such as CUL1 and TGFB1. Our proposed analytic workflow shows the advantages of combining clinical and omics data as well as individual- and group-level transcriptome profiling.
Collapse
Affiliation(s)
- Archana Bhardwaj
- GIGA-R Centre, BIO3 - Medical Genomics, University of Liège, Avenue de L'Hôpital, 11, 4000, Liège, Belgium.
| | - Claire Josse
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU), Liège, Belgium
- Medical Oncology Department, CHU Liège, Liège, Belgium
| | - Daniel Van Daele
- Department of Gastro-Enterology, University Hospital (CHU), Liège, Belgium
| | - Christophe Poulet
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU), Liège, Belgium
- Laboratory of Rheumatology, GIGA-R, University Hospital (CHULiege), Liège, Belgium
| | - Marcela Chavez
- Department of Medicine, Division of Hematology, University Hospital (CHU), Liège, Belgium
| | - Ingrid Struman
- GIGA-R Centre, Laboratory of Molecular Angiogenesis, University of Liège, Liège, Belgium
| | - Kristel Van Steen
- GIGA-R Centre, BIO3 - Medical Genomics, University of Liège, Avenue de L'Hôpital, 11, 4000, Liège, Belgium
| |
Collapse
|
36
|
Van Daele D, Weytjens B, De Raedt L, Marchal K. OMEN: Network-based Driver Gene Identification using Mutual Exclusivity. Bioinformatics 2022; 38:3245-3251. [PMID: 35552634 DOI: 10.1093/bioinformatics/btac312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Network-based driver identification methods that can exploit mutual exclusivity typically fail to detect rare drivers because of their statistical rigor. Propagation-based methods in contrast allow recovering rare driver genes, but the interplay between network topology and high-scoring nodes often results in spurious predictions. The specificity of driver gene detection can be improved by taking into account both gene-specific and gene-set properties. Combining these requires a formalism that can adjust gene-set properties depending on the exact network context within which a gene is analyzed. RESULTS We developed OMEN: a logic programming framework based on random walk semantics. OMEN presents a number of novel concepts. In particular, its design is unique in that it presents an effective approach to combine both gene-specific driver properties and gene-set properties, and includes a novel method to avoid restrictive, a priori filtering of genes by exploiting the gene-set property of mutual exclusivity, expressed in terms of the functional impact scores of mutations, rather than in terms of simple binary mutation calls. Applying OMEN to a benchmark data set derived from TCGA illustrates how OMEN is able to robustly identify driver genes and modules of driver genes as proxies of driver pathways. AVAILABILITY The source code is freely available for download at www.github.com/DriesVanDaele/OMEN The data set is archived at https://doi.org/10.5281/zenodo.6419097 and the code at https://doi.org/10.5281/zenodo.6419764. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Bram Weytjens
- Department of Plant Biotechnology and Bioinformatics, Department of Information Technology, IDLab, Gent, 9000, Belgium IMEC
| | - Luc De Raedt
- Department of Computer Science, KU Leuven, 3001, Belgium
| | - Kathleen Marchal
- Department of Plant Biotechnology and Bioinformatics, Department of Information Technology, IDLab, Gent, 9000, Belgium IMEC
| |
Collapse
|
37
|
Zhang X, Zhou Z, Xu H, Liu CT. Integrative clustering methods for multi-omics data. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL STATISTICS 2022; 14. [PMID: 35573155 PMCID: PMC9097984 DOI: 10.1002/wics.1553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrative analysis of multi-omics data has drawn much attention from the scientific community due to the technological advancements which have generated various omics data. Leveraging these multi-omics data potentially provides a more comprehensive view of the disease mechanism or biological processes. Integrative multi-omics clustering is an unsupervised integrative method specifically used to find coherent groups of samples or features by utilizing information across multi-omics data. It aims to better stratify diseases and to suggest biological mechanisms and potential targeted therapies for the diseases. However, applying integrative multi-omics clustering is both statistically and computationally challenging due to various reasons such as high dimensionality and heterogeneity. In this review, we summarized integrative multi-omics clustering methods into three general categories: concatenated clustering, clustering of clusters, and interactive clustering based on when and how the multi-omics data are processed for clustering. We further classified the methods into different approaches under each category based on the main statistical strategy used during clustering. In addition, we have provided recommended practices tailored to four real-life scenarios to help researchers to strategize their selection in integrative multi-omics clustering methods for their future studies.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Zhenwei Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Vahabi N, Michailidis G. Unsupervised Multi-Omics Data Integration Methods: A Comprehensive Review. Front Genet 2022; 13:854752. [PMID: 35391796 PMCID: PMC8981526 DOI: 10.3389/fgene.2022.854752] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
Through the developments of Omics technologies and dissemination of large-scale datasets, such as those from The Cancer Genome Atlas, Alzheimer’s Disease Neuroimaging Initiative, and Genotype-Tissue Expression, it is becoming increasingly possible to study complex biological processes and disease mechanisms more holistically. However, to obtain a comprehensive view of these complex systems, it is crucial to integrate data across various Omics modalities, and also leverage external knowledge available in biological databases. This review aims to provide an overview of multi-Omics data integration methods with different statistical approaches, focusing on unsupervised learning tasks, including disease onset prediction, biomarker discovery, disease subtyping, module discovery, and network/pathway analysis. We also briefly review feature selection methods, multi-Omics data sets, and resources/tools that constitute critical components for carrying out the integration.
Collapse
Affiliation(s)
- Nasim Vahabi
- Informatics Institute, University of Florida, Gainesville, FL, United States
| | - George Michailidis
- Informatics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
39
|
Zhang B, Yao K, Cheng C. A network-based integration for understanding racial disparity in prostate cancer. Transl Oncol 2022; 17:101327. [PMID: 34998235 PMCID: PMC8738961 DOI: 10.1016/j.tranon.2021.101327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/24/2022] Open
Abstract
Compared to Caucasians (CAs), African Americans (AAs) have a higher rate of incidence and mortality in prostate cancer and are prone to be diagnosed at later stages. To understand this racial disparity, molecular features of different types, including gene expression, DNA methylation and other genomic alterations, have been compared between tumor samples from the two races, but led to different disparity associated genes (DAGs). In this study, we applied a network-based algorithm to integrate a comprehensive set of genomic datasets and identified 130 core DAGs. Out of these genes, 78 were not identified by any individual dataset but prioritized and selected through network propagation. We found DAGs were highly enriched in several critical prostate cancer-related signaling transduction and cell cycle pathways and were more likely to be associated with patient prognosis in prostate cancer. Furthermore, DAGs were over-represented in prostate cancer risk genes identified from previous genome wide association studies. We also found DAGs were enriched in kinase and transcription factor encoding genes. Interestingly, for many of these prioritized kinases their association with racial disparity did not manifest from the original genomic/transcriptomic data but was reflected by their differential phosphorylation levels between AA and CA prostate tumor samples. Similarly, the disparity relevance of some transcription factors was not reflected at the mRNA or protein expression level, but at the activity level as demonstrated by their differential ability in regulating target gene expression. Our integrative analysis provided new candidate targets for improving prostate cancer treatment and addressing the racial disparity problem.
Collapse
Affiliation(s)
- Baoyi Zhang
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX 77030, United States
| | - Kevin Yao
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, United States; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, United States; Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
40
|
Zhao L, Han L, Wei X, Zhou Y, Zhang Y, Si N, Wang H, Yang J, Bian B, Zhao H. Toxicokinetics of Arenobufagin and its Cardiotoxicity Mechanism Exploration Based on Lipidomics and Proteomics Approaches in Rats. Front Pharmacol 2022; 12:780016. [PMID: 35002716 PMCID: PMC8727535 DOI: 10.3389/fphar.2021.780016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
Arenobufagin (ArBu), one of the main active bufadienolides of toad venom with cardiotonic effect, analgesic effect, and outstanding anti-tumor potentiality, is also a potential cardiotoxic component. In the present study, the cardiac effect of ArBu and its underlying mechanism were explored by integrating data such as heart rates, toxicokinetics, myocardial enzyme and brain natriuretic peptide (BNP) activity, pathological sections, lipidomics and proteomics. Under different doses, the cardiac effects turned out to be different. The oral dose of 60 mg/kg of ArBu sped up the heart rate. However, 120 mg/kg ArBu mainly reduced the heart rate. Over time, they all returned to normal, consisting of the trend of ArBu concentration-time curve. High concentrations of myocardial enzymes and BNP indicated that ArBu inhibited or impaired the cardiac function of rats. Pathological sections of hearts also showed that ArBu caused myocardial fiber disorder and rupture, in which the high-dose group was more serious. At the same time, serum and heart tissue lipidomics were used to explore the changes in body lipid metabolism under different doses. The data indicated a larger difference in the high-dose ArBu group. There were likewise many significant differences in the proteomics of the heart. Furthermore, a multi-layered network was used to integrate the above information to explore the potential mechanism. Finally, 4 proteins that were shown to be significantly and differentially expressed were validated by targeted proteomics using parallel reaction monitoring (PRM) analysis. Our findings indicated that ArBu behaved as a bidirectional regulation of the heart. The potential mechanism of cardiac action was revealed with the increased dose, which provided a useful reference for the safety of clinical application of ArBu.
Collapse
Affiliation(s)
- Lijuan Zhao
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Shaanxi Chinese Medicine Institute (Shaanxi Pharmaceutical Information Center), Xianyang, China
| | - Lingyu Han
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xiaolu Wei
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanyan Zhou
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanqiong Zhang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Si
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjie Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian Yang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baolin Bian
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Abstract
DNA microarrays are widely used to investigate gene expression. Even though the classical analysis of microarray data is based on the study of differentially expressed genes, it is well known that genes do not act individually. Network analysis can be applied to study association patterns of the genes in a biological system. Moreover, it finds wide application in differential coexpression analysis between different systems. Network based coexpression studies have for example been used in (complex) disease gene prioritization, disease subtyping, and patient stratification.In this chapter we provide an overview of the methods and tools used to create networks from microarray data and describe multiple methods on how to analyze a single network or a group of networks. The described methods range from topological metrics, functional group identification to data integration strategies, topological pathway analysis as well as graphical models.
Collapse
Affiliation(s)
- Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - Luca Cattelani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- BioMediTech Institute, Tampere University, Tampere, Finland.
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere University, Tampere, Finland.
- Institute of Biotechnology , University of Helsinki, Helsinki, Finland.
| |
Collapse
|
42
|
Correa R, Alonso-Pupo N, Hernández Rodríguez EW. Multi-omics data integration approaches for precision oncology. Mol Omics 2022; 18:469-479. [DOI: 10.1039/d1mo00411e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Next-generation sequencing (NGS) has been pivotal to enhance the molecular characterization of human malignancies, allowing multiple omics data types to be available for cancer researchers and practitioners. In this context,...
Collapse
|
43
|
Demirel HC, Arici MK, Tuncbag N. Computational approaches leveraging integrated connections of multi-omic data toward clinical applications. Mol Omics 2021; 18:7-18. [PMID: 34734935 DOI: 10.1039/d1mo00158b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In line with the advances in high-throughput technologies, multiple omic datasets have accumulated to study biological systems and diseases coherently. No single omics data type is capable of fully representing cellular activity. The complexity of the biological processes arises from the interactions between omic entities such as genes, proteins, and metabolites. Therefore, multi-omic data integration is crucial but challenging. The impact of the molecular alterations in multi-omic data is not local in the neighborhood of the altered gene or protein; rather, the impact diffuses in the network and changes the functionality of multiple signaling pathways and regulation of the gene expression. Additionally, multi-omic data is high-dimensional and has background noise. Several integrative approaches have been developed to accurately interpret the multi-omic datasets, including machine learning, network-based methods, and their combination. In this review, we overview the most recent integrative approaches and tools with a focus on network-based methods. We then discuss these approaches according to their specific applications, from disease-network and biomarker identification to patient stratification, drug discovery, and repurposing.
Collapse
Affiliation(s)
- Habibe Cansu Demirel
- Graduate School of Informatics, Middle East Technical University, Ankara, 06800, Turkey
| | - Muslum Kaan Arici
- Graduate School of Informatics, Middle East Technical University, Ankara, 06800, Turkey.,Foot and Mouth Diseases Institute, Ministry of Agriculture and Forestry, Ankara, 06044, Turkey
| | - Nurcan Tuncbag
- Chemical and Biological Engineering, College of Engineering, Koc University, Istanbul, 34450, Turkey.,School of Medicine, Koc University, Istanbul, 34450, Turkey.,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
| |
Collapse
|
44
|
Shi K, Lin W, Zhao XM. Identifying Molecular Biomarkers for Diseases With Machine Learning Based on Integrative Omics. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:2514-2525. [PMID: 32305934 DOI: 10.1109/tcbb.2020.2986387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Molecular biomarkers are certain molecules or set of molecules that can be of help for diagnosis or prognosis of diseases or disorders. In the past decades, thanks to the advances in high-throughput technologies, a huge amount of molecular 'omics' data, e.g., transcriptomics and proteomics, have been accumulated. The availability of these omics data makes it possible to screen biomarkers for diseases or disorders. Accordingly, a number of computational approaches have been developed to identify biomarkers by exploring the omics data. In this review, we present a comprehensive survey on the recent progress of identification of molecular biomarkers with machine learning approaches. Specifically, we categorize the machine learning approaches into supervised, un-supervised and recommendation approaches, where the biomarkers including single genes, gene sets and small gene networks. In addition, we further discuss potential problems underlying bio-medical data that may pose challenges for machine learning, and provide possible directions for future biomarker identification.
Collapse
|
45
|
de Schaetzen van Brienen L, Miclotte G, Larmuseau M, Van den Eynden J, Marchal K. Network-Based Analysis to Identify Drivers of Metastatic Prostate Cancer Using GoNetic. Cancers (Basel) 2021; 13:5291. [PMID: 34771455 PMCID: PMC8582433 DOI: 10.3390/cancers13215291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Most known driver genes of metastatic prostate cancer are frequently mutated. To dig into the long tail of rarely mutated drivers, we performed network-based driver identification on the Hartwig Medical Foundation metastatic prostate cancer data set (HMF cohort). Hereto, we developed GoNetic, a method based on probabilistic pathfinding, to identify recurrently mutated subnetworks. In contrast to most state-of-the-art network-based methods, GoNetic can leverage sample-specific mutational information and the weights of the underlying prior network. When applied to the HMF cohort, GoNetic successfully recovered known primary and metastatic drivers of prostate cancer that are frequently mutated in the HMF cohort (TP53, RB1, and CTNNB1). In addition, the identified subnetworks contain frequently mutated genes, reflect processes related to metastatic prostate cancer, and contain rarely mutated driver candidates. To further validate these rarely mutated genes, we assessed whether the identified genes were more mutated in metastatic than in primary samples using an independent cohort. Then we evaluated their association with tumor evolution and with the lymph node status of the patients. This resulted in forwarding several novel putative driver genes for metastatic prostate cancer, some of which might be prognostic for disease evolution.
Collapse
Affiliation(s)
- Louise de Schaetzen van Brienen
- Department of Plant Biotechnology and Bioinformatics, Faculty of Sciences, Ghent University, 9052 Ghent, Belgium; (L.d.S.v.B.); (G.M.); (M.L.)
- Department of Information Technology, Faculty of Engineering and Architecture, Ghent University-IMEC, 9052 Ghent, Belgium
| | - Giles Miclotte
- Department of Plant Biotechnology and Bioinformatics, Faculty of Sciences, Ghent University, 9052 Ghent, Belgium; (L.d.S.v.B.); (G.M.); (M.L.)
- Department of Information Technology, Faculty of Engineering and Architecture, Ghent University-IMEC, 9052 Ghent, Belgium
| | - Maarten Larmuseau
- Department of Plant Biotechnology and Bioinformatics, Faculty of Sciences, Ghent University, 9052 Ghent, Belgium; (L.d.S.v.B.); (G.M.); (M.L.)
- Department of Information Technology, Faculty of Engineering and Architecture, Ghent University-IMEC, 9052 Ghent, Belgium
| | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Kathleen Marchal
- Department of Plant Biotechnology and Bioinformatics, Faculty of Sciences, Ghent University, 9052 Ghent, Belgium; (L.d.S.v.B.); (G.M.); (M.L.)
- Department of Information Technology, Faculty of Engineering and Architecture, Ghent University-IMEC, 9052 Ghent, Belgium
| |
Collapse
|
46
|
Csabai L, Fazekas D, Kadlecsik T, Szalay-Bekő M, Bohár B, Madgwick M, Módos D, Ölbei M, Gul L, Sudhakar P, Kubisch J, Oyeyemi OJ, Liska O, Ari E, Hotzi B, Billes VA, Molnár E, Földvári-Nagy L, Csályi K, Demeter A, Pápai N, Koltai M, Varga M, Lenti K, Farkas IJ, Türei D, Csermely P, Vellai T, Korcsmáros T. SignaLink3: a multi-layered resource to uncover tissue-specific signaling networks. Nucleic Acids Res 2021; 50:D701-D709. [PMID: 34634810 PMCID: PMC8728204 DOI: 10.1093/nar/gkab909] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/26/2022] Open
Abstract
Signaling networks represent the molecular mechanisms controlling a cell's response to various internal or external stimuli. Most currently available signaling databases contain only a part of the complex network of intertwining pathways, leaving out key interactions or processes. Hence, we have developed SignaLink3 (http://signalink.org/), a value-added knowledge-base that provides manually curated data on signaling pathways and integrated data from several types of databases (interaction, regulation, localisation, disease, etc.) for humans, and three major animal model organisms. SignaLink3 contains over 400 000 newly added human protein-protein interactions resulting in a total of 700 000 interactions for Homo sapiens, making it one of the largest integrated signaling network resources. Next to H. sapiens, SignaLink3 is the only current signaling network resource to provide regulatory information for the model species Caenorhabditis elegans and Danio rerio, and the largest resource for Drosophila melanogaster. Compared to previous versions, we have integrated gene expression data as well as subcellular localization of the interactors, therefore uniquely allowing tissue-, or compartment-specific pathway interaction analysis to create more accurate models. Data is freely available for download in widely used formats, including CSV, PSI-MI TAB or SQL.
Collapse
Affiliation(s)
- Luca Csabai
- Earlham Institute, Norwich NR4 7UZ, UK.,Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Dávid Fazekas
- Earlham Institute, Norwich NR4 7UZ, UK.,Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Tamás Kadlecsik
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | | | - Balázs Bohár
- Earlham Institute, Norwich NR4 7UZ, UK.,Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Matthew Madgwick
- Earlham Institute, Norwich NR4 7UZ, UK.,Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, NR4 7UQ, UK
| | - Dezső Módos
- Earlham Institute, Norwich NR4 7UZ, UK.,Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, NR4 7UQ, UK
| | - Márton Ölbei
- Earlham Institute, Norwich NR4 7UZ, UK.,Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, NR4 7UQ, UK
| | - Lejla Gul
- Earlham Institute, Norwich NR4 7UZ, UK
| | - Padhmanand Sudhakar
- Earlham Institute, Norwich NR4 7UZ, UK.,Translational Research in GastroIntestinal Disorders, Leuven BE-3000, Belgium
| | - János Kubisch
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | | | - Orsolya Liska
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,HCEMM-BRC Metabolic Systems Biology Lab, Szeged H-6726, Hungary.,Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged H-6726, Hungary.,Doctoral School in Biology, University of Szeged, Szeged H-6720 Hungary
| | - Eszter Ari
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,HCEMM-BRC Metabolic Systems Biology Lab, Szeged H-6726, Hungary.,Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged H-6726, Hungary
| | - Bernadette Hotzi
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Viktor A Billes
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,ELKH/MTA-ELTE Genetics Research Group, Budapest H-1117, Hungary
| | - Eszter Molnár
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - László Földvári-Nagy
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,Department of Morphology and Physiology, Semmelweis University, Budapest H-1088, Hungary
| | - Kitti Csályi
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Amanda Demeter
- Earlham Institute, Norwich NR4 7UZ, UK.,Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Nóra Pápai
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,Institute of Molecular Biotechnology, Vienna A-1030, Austria
| | - Mihály Koltai
- Centre for the Mathematical Modelling of Infectious Diseases (CMMID), London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Máté Varga
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Katalin Lenti
- Department of Morphology and Physiology, Semmelweis University, Budapest H-1088, Hungary
| | - Illés J Farkas
- Citibank Europe plc Hungarian Branch Office, Budapest H-1133, Hungary
| | - Dénes Türei
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Péter Csermely
- Department of Molecular Biology, Semmelweis University, Budapest H-1094, Hungary
| | - Tibor Vellai
- Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,ELKH/MTA-ELTE Genetics Research Group, Budapest H-1117, Hungary
| | - Tamás Korcsmáros
- Earlham Institute, Norwich NR4 7UZ, UK.,Department of Genetics, ELTE Eötvös Loránd University, Budapest H-1117, Hungary.,Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, NR4 7UQ, UK
| |
Collapse
|
47
|
Zheng F, Kelly MR, Ramms DJ, Heintschel ML, Tao K, Tutuncuoglu B, Lee JJ, Ono K, Foussard H, Chen M, Herrington KA, Silva E, Liu S, Chen J, Churas C, Wilson N, Kratz A, Pillich RT, Patel DN, Park J, Kuenzi B, Yu MK, Licon K, Pratt D, Kreisberg JF, Kim M, Swaney DL, Nan X, Fraley SI, Gutkind JS, Krogan NJ, Ideker T. Interpretation of cancer mutations using a multiscale map of protein systems. Science 2021; 374:eabf3067. [PMID: 34591613 PMCID: PMC9126298 DOI: 10.1126/science.abf3067] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A major goal of cancer research is to understand how mutations distributed across diverse genes affect common cellular systems, including multiprotein complexes and assemblies. Two challenges—how to comprehensively map such systems and how to identify which are under mutational selection—have hindered this understanding. Accordingly, we created a comprehensive map of cancer protein systems integrating both new and published multi-omic interaction data at multiple scales of analysis. We then developed a unified statistical model that pinpoints 395 specific systems under mutational selection across 13 cancer types. This map, called NeST (Nested Systems in Tumors), incorporates canonical processes and notable discoveries, including a PIK3CA-actomyosin complex that inhibits phosphatidylinositol 3-kinase signaling and recurrent mutations in collagen complexes that promote tumor proliferation. These systems can be used as clinical biomarkers and implicate a total of 548 genes in cancer evolution and progression. This work shows how disparate tumor mutations converge on protein assemblies at different scales.
Collapse
Affiliation(s)
- Fan Zheng
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Marcus R. Kelly
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Dana J. Ramms
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Marissa L. Heintschel
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kai Tao
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Beril Tutuncuoglu
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - John J. Lee
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Keiichiro Ono
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Helene Foussard
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Michael Chen
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kari A. Herrington
- Department of Biochemistry and Biophysics Center for Advanced Light Microscopy at UCSF, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Erica Silva
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Sophie Liu
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jing Chen
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher Churas
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicholas Wilson
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Anton Kratz
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Rudolf T. Pillich
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Devin N. Patel
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Jisoo Park
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Brent Kuenzi
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Michael K. Yu
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Katherine Licon
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Dexter Pratt
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jason F. Kreisberg
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Minkyu Kim
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Danielle L. Swaney
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Xiaolin Nan
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - J. Silvio Gutkind
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Nevan J. Krogan
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| |
Collapse
|
48
|
Zhang Q, Huang X. The modulatory properties of Astragalus membranaceus treatment on endometrial cancer: an integrated pharmacological method. PeerJ 2021; 9:e11995. [PMID: 34513331 PMCID: PMC8395571 DOI: 10.7717/peerj.11995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Astragalus membranaceus is a traditional Chinese medicine and has been used for adjuvant clinical therapy for a variety of cancers. However, the mechanism of its action on endometrial carcinoma is unclear. Based on the Gene Expression Omnibus (GEO) database, the Cancer Genome Atlas (TCGA) database, and the Traditional Chinese Medicine System Pharmacology Database (TCMSP™), the drug and target compounds were initially screened to construct a common network module. Twenty active compounds in Astragalus membranaceus were successfully identified, which hit by 463 potential targets related to endometrial cancer. Eight of the more highly predictive compounds (such as Jaranol, Bifendate, Isorhamnetin, Calycosin, 7-O-methylisomucronulatol, Formononetin, Kaempferol, Quercetin) were involved in DNA integrity checkpoint, cyclin-dependent protein kinase holoenzyme complex, and histone kinase activity. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway confirmed that Astragalus membranaceus might play a role in the treatment of endometrial cancer through p53 signalling pathway, transcriptional misregulation in cancer, and endometrial cancer signalling pathway. Drug-target-pathway networks were constructed using Cytoscape to provide a visual perspective. In addition, we verified that formononetin inhibited the proliferation of endometrial cancer cells through cell viability tests and clone formation tests. And qPCR and western blot found that formononetin exerts anti-cancer effects by promoting the expression of estrogen receptor beta (ERβ) and p53. Based on a systematic network pharmacology approach, our works successfully predict the active ingredients and potential targets of Astragalus membranaceus for application to endometrial cancer and helps to illustrate mechanism of action on a comprehensive level.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Gynecology, Hebei Medical University Second Affiliated Hospital, Shijiazhuang, China
| | - Xianghua Huang
- Department of Gynecology, Hebei Medical University Second Affiliated Hospital, Shijiazhuang, China
| |
Collapse
|
49
|
Montazeri H, Coto-Llerena M, Bianco G, Zangene E, Taha-Mehlitz S, Paradiso V, Srivatsa S, de Weck A, Roma G, Lanzafame M, Bolli M, Beerenwinkel N, von Flüe M, Terracciano L, Piscuoglio S, Ng CKY. Systematic identification of novel cancer genes through analysis of deep shRNA perturbation screens. Nucleic Acids Res 2021; 49:8488-8504. [PMID: 34313788 PMCID: PMC8421231 DOI: 10.1093/nar/gkab627] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 11/30/2022] Open
Abstract
Systematic perturbation screens provide comprehensive resources for the elucidation of cancer driver genes. The perturbation of many genes in relatively few cell lines in such functional screens necessitates the development of specialized computational tools with sufficient statistical power. Here we developed APSiC (Analysis of Perturbation Screens for identifying novel Cancer genes) to identify genetic drivers and effectors in perturbation screens even with few samples. Applying APSiC to the shRNA screen Project DRIVE, APSiC identified well-known and novel putative mutational and amplified cancer genes across all cancer types and in specific cancer types. Additionally, APSiC discovered tumor-promoting and tumor-suppressive effectors, respectively, for individual cancer types, including genes involved in cell cycle control, Wnt/β-catenin and hippo signalling pathways. We functionally demonstrated that LRRC4B, a putative novel tumor-suppressive effector, suppresses proliferation by delaying cell cycle and modulates apoptosis in breast cancer. We demonstrate APSiC is a robust statistical framework for discovery of novel cancer genes through analysis of large-scale perturbation screens. The analysis of DRIVE using APSiC is provided as a web portal and represents a valuable resource for the discovery of novel cancer genes.
Collapse
Affiliation(s)
- Hesam Montazeri
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Mairene Coto-Llerena
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Visceral Surgery and Precision Medicine Research laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gaia Bianco
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Visceral Surgery and Precision Medicine Research laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ehsan Zangene
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Stephanie Taha-Mehlitz
- Visceral Surgery and Precision Medicine Research laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Viola Paradiso
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Sumana Srivatsa
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Antoine de Weck
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Guglielmo Roma
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Manuela Lanzafame
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Martin Bolli
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Markus von Flüe
- Clarunis, Department of Visceral Surgery, University Centre for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital Basel, Switzerland
| | - Luigi M Terracciano
- Department of Pathology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Salvatore Piscuoglio
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Visceral Surgery and Precision Medicine Research laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Charlotte K Y Ng
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
50
|
Ye C, Wang H, Li Z, Xia C, Yuan S, Yan R, Yang X, Ma T, Wen X, Yang D. Comprehensive data analysis of genomics, epigenomics, and transcriptomics to identify specific biomolecular markers for prostate adenocarcinoma. Transl Androl Urol 2021; 10:3030-3045. [PMID: 34430406 PMCID: PMC8350225 DOI: 10.21037/tau-21-576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Background Multiomics data analysis based on high-throughput sequencing technology has become a hotspot in tumor investigation. The present study aimed to explore prognostic biomarkers via investigating DNA copy number variation (CNV) and methylation variation (MET) data in prostate cancer. Methods We obtained the messenger RNA (mRNA) expression, CNV, and methylated data of prostate adenocarcinoma (PRAD) samples via The Cancer Genome Atlas (TCGA)-PRAD cohort. We calculated and assessed the associations between CNV and RNA sequencing (RNA-seq), and between MET and RNA-seq via Pearson correlation coefficients. We then used the "iCluster" package to perform multigroup cluster analysis with CNVcor gene CNV data, METcor gene methylation data, and CNVcor and METcor gene mRNA data. The univariate Cox analysis was used to screen significant hub genes, and multivariate Cox analysis was used to construct risk a model. The nomogram was constructed based on "rms" package, and the immune infiltrating patterns were compared between high- and low-risk groups. Results A total of 477 PRAD samples with complete CNV, methylation, mRNA, and matched clinical information were included in our study. A list of 10,073 CNVcor genes and 9841 METcor genes were confirmed with a significance level of P<0.01. We found that CNVcor is more likely to appear on chromosome (chr)8, chr17, and chr10, while METcor is more likely to appear on chr1, chr19, and chr17. Based on the core genes, we finally classified the samples into 4 subtypes, incorporating iC1 (iCluster) (92 samples), iC2 (79 samples), iC3 (165 samples), and iC4 (141 samples). Furthermore, we constructed the prognostic model for PRAD based on the 5 genes (IER3, AOX1, PRKCDBP, UBD, and FBLN5). Nomograms incorporating risk score and other clinical variables were further constructed, and these nomograms exhibited superior predictive ability. We further compared the differential immune infiltrating patterns in 2 risk groups and found significantly low levels of infiltrating cluster of differentiation (CD)8+ T cells in high-risk samples. Conclusions Our study integrated the multi-omics data to elucidate the molecular features of PRAD and pivotal genes for predicting prognosis.
Collapse
Affiliation(s)
- Chunwei Ye
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhipeng Li
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chengxing Xia
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shunhui Yuan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruping Yan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaofang Yang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tao Ma
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingqiao Wen
- Department of Urology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Delin Yang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|