1
|
Cheng H, Liang M, Gao Y, Zhao W, Guo WF. Multiomics with Evolutionary Computation to Identify Molecular and Module Biomarkers for Early Diagnosis and Treatment of Complex Disease. Genes (Basel) 2025; 16:244. [PMID: 40149396 PMCID: PMC11942451 DOI: 10.3390/genes16030244] [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: 02/02/2025] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
It is important to identify disease biomarkers (DBs) for early diagnosis and treatment of complex diseases in personalized medicine. However, existing methods integrating intelligence technologies and multiomics to predict key biomarkers are limited by the complex dynamic characteristics of omics data, making it difficult to meet the high-precision requirements for biomarker characterization in large dimensions. This study reviewed current analysis methods of evolutionary computation (EC) by considering the essential characteristics of DB identification problems and the advantages of EC, aiming to explore the complex dynamic characteristics of multiomics. In this study, EC-based biomarker identification strategies were summarized as evolutionary algorithms, swarm intelligence and other EC methods for molecular and module DB identification, respectively. Finally, we pointed out the challenges in current research and future research directions. This study can enrich the application of EC theory and promote interdisciplinary integration between EC and bioinformatics.
Collapse
Affiliation(s)
- Han Cheng
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (H.C.); (M.L.); (Y.G.); (W.Z.)
| | - Mengyu Liang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (H.C.); (M.L.); (Y.G.); (W.Z.)
| | - Yiwen Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (H.C.); (M.L.); (Y.G.); (W.Z.)
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (H.C.); (M.L.); (Y.G.); (W.Z.)
| | - Wei-Feng Guo
- School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
2
|
Zhang H, Lin C, Chen Y, Shen X, Wang R, Chen Y, Lyu J. Enhancing Molecular Network-Based Cancer Driver Gene Prediction Using Machine Learning Approaches: Current Challenges and Opportunities. J Cell Mol Med 2025; 29:e70351. [PMID: 39804102 PMCID: PMC11726689 DOI: 10.1111/jcmm.70351] [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: 11/12/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Cancer is a complex disease driven by mutations in the genes that play critical roles in cellular processes. The identification of cancer driver genes is crucial for understanding tumorigenesis, developing targeted therapies and identifying rational drug targets. Experimental identification and validation of cancer driver genes are time-consuming and costly. Studies have demonstrated that interactions among genes are associated with similar phenotypes. Therefore, identifying cancer driver genes using molecular network-based approaches is necessary. Molecular network-based random walk-based approaches, which integrate mutation data with protein-protein interaction networks, have been widely employed in predicting cancer driver genes and demonstrated robust predictive potential. However, recent advancements in deep learning, particularly graph-based models, have provided novel opportunities for enhancing the prediction of cancer driver genes. This review aimed to comprehensively explore how machine learning methodologies, particularly network propagation, graph neural networks, autoencoders, graph embeddings, and attention mechanisms, improve the scalability and interpretability of molecular network-based cancer gene prediction.
Collapse
Affiliation(s)
- Hao Zhang
- Postgraduate Training Base Alliance of Wenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of Biophysics, Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Chaohuan Lin
- Postgraduate Training Base Alliance of Wenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of Biophysics, Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Ying'ao Chen
- Wenzhou Key Laboratory of Biophysics, Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | | | - Ruizhe Wang
- Wenzhou Longwan High SchoolWenzhouZhejiangChina
| | - Yiqi Chen
- Wenzhou Longwan High SchoolWenzhouZhejiangChina
| | - Jie Lyu
- Postgraduate Training Base Alliance of Wenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of Biophysics, Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| |
Collapse
|
3
|
Liang J, Hu Z, Bi Y, Cheng H, Guo WF. Multimodal multiobjective optimization with structural network control principles to optimize personalized drug targets for drug discovery of individual patients. Brief Bioinform 2024; 26:bbaf007. [PMID: 39835535 PMCID: PMC11747759 DOI: 10.1093/bib/bbaf007] [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: 09/05/2024] [Revised: 12/05/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
Structural network control principles provided novel and efficient clues for the optimization of personalized drug targets (PDTs) related to state transitions of individual patients. However, most existing methods focus on one subnetwork or module as drug targets through the identification of the minimal set of driver nodes and ignore the state transition capabilities of other modules with different configurations of drug targets [i.e. multimodal drug targets (MDTs)] embedding the knowledge of previous drug targets (i.e. multiobjective optimization). Therefore, a novel multimodal multiobjective evolutionary optimization framework (called MMONCP) is proposed to optimize PDTs with network control principles. The key points of MMONCP are that a constrained multimodal multiobjective optimization problem is formed with discrete constraints on the decision space and multimodality characteristics, and a novel evolutionary algorithm denoted as CMMOEA-GLS-WSCD is designed by combining a global and local search strategy and a weighting-based special crowding distance strategy to balance the diversity of both objective and decision space. The experimental results on three cancer genomics data from The Cancer Genome Atlas indicate that MMONCP achieves a higher performance including algorithm convergence and diversity, the fraction of identified MDTs, and the area under the curve score than advanced algorithms. Additionally, MMONCP can detect the early state from the difference between the target activity and toxicity of MDTs and provide early treatment options for cancer treatment in precision medicine.
Collapse
Affiliation(s)
- Jing Liang
- School of Electrical and Information Engineering, Zhengzhou University, No. 100, Science Avenue, Hightech District, Zhengzhou City 450001, Henan Province, China
- State Key Laboratory of Intelligent Agricultural Power Equipment, No. 39, Xiyuan Road, Jianxi District, Luoyang City 471039, Henan Province, China
| | - Zhuo Hu
- School of Electrical and Information Engineering, Zhengzhou University, No. 100, Science Avenue, Hightech District, Zhengzhou City 450001, Henan Province, China
| | - Ying Bi
- School of Electrical and Information Engineering, Zhengzhou University, No. 100, Science Avenue, Hightech District, Zhengzhou City 450001, Henan Province, China
| | - Han Cheng
- School of Life Sciences, Zhengzhou University, No. 100, Science Avenue, High-tech District, Zhengzhou City 450001, Henan Province, China
| | - Wei-Feng Guo
- School of Electrical and Information Engineering, Zhengzhou University, No. 100, Science Avenue, Hightech District, Zhengzhou City 450001, Henan Province, China
- State Key Laboratory of Intelligent Agricultural Power Equipment, No. 39, Xiyuan Road, Jianxi District, Luoyang City 471039, Henan Province, China
| |
Collapse
|
4
|
Zhang T, Zhang SW, Xie MY, Li Y. Identifying cooperating cancer driver genes in individual patients through hypergraph random walk. J Biomed Inform 2024; 157:104710. [PMID: 39159864 DOI: 10.1016/j.jbi.2024.104710] [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] [Received: 04/27/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE Identifying cancer driver genes, especially rare or patient-specific cancer driver genes, is a primary goal in cancer therapy. Although researchers have proposed some methods to tackle this problem, these methods mostly identify cancer driver genes at single gene level, overlooking the cooperative relationship among cancer driver genes. Identifying cooperating cancer driver genes in individual patients is pivotal for understanding cancer etiology and advancing the development of personalized therapies. METHODS Here, we propose a novel Personalized Cooperating cancer Driver Genes (PCoDG) method by using hypergraph random walk to identify the cancer driver genes that cooperatively drive individual patient cancer progression. By leveraging the powerful ability of hypergraph in representing multi-way relationships, PCoDG first employs the personalized hypergraph to depict the complex interactions among mutated genes and differentially expressed genes of an individual patient. Then, a hypergraph random walk algorithm based on hyperedge similarity is utilized to calculate the importance scores of mutated genes, integrating these scores with signaling pathway data to identify the cooperating cancer driver genes in individual patients. RESULTS The experimental results on three TCGA cancer datasets (i.e., BRCA, LUAD, and COADREAD) demonstrate the effectiveness of PCoDG in identifying personalized cooperating cancer driver genes. These genes identified by PCoDG not only offer valuable insights into patient stratification correlating with clinical outcomes, but also provide an useful reference resource for tailoring personalized treatments. CONCLUSION We propose a novel method that can effectively identify cooperating cancer driver genes for individual patients, thereby deepening our understanding of the cooperative relationship among personalized cancer driver genes and advancing the development of precision oncology.
Collapse
Affiliation(s)
- Tong Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China; School of Electrical and Mechanical Engineering, Pingdingshan University, Pingdingshan 467000, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Ming-Yu Xie
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yan Li
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
5
|
Tokuhara Y, Akutsu T, Schwartz JM, Nacher JC. A practically efficient algorithm for identifying critical control proteins in directed probabilistic biological networks. NPJ Syst Biol Appl 2024; 10:87. [PMID: 39134558 PMCID: PMC11319667 DOI: 10.1038/s41540-024-00411-y] [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: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
Network controllability is unifying the traditional control theory with the structural network information rooted in many large-scale biological systems of interest, from intracellular networks in molecular biology to brain neuronal networks. In controllability approaches, the set of minimum driver nodes is not unique, and critical nodes are the most important control elements because they appear in all possible solution sets. On the other hand, a common but largely unexplored feature in network control approaches is the probabilistic failure of edges or the uncertainty in the determination of interactions between molecules. This is particularly true when directed probabilistic interactions are considered. Until now, no efficient algorithm existed to determine critical nodes in probabilistic directed networks. Here we present a probabilistic control model based on a minimum dominating set framework that integrates the probabilistic nature of directed edges between molecules and determines the critical control nodes that drive the entire network functionality. The proposed algorithm, combined with the developed mathematical tools, offers practical efficiency in determining critical control nodes in large probabilistic networks. The method is then applied to the human intracellular signal transduction network revealing that critical control nodes are associated with important biological features and perturbed sets of genes in human diseases, including SARS-CoV-2 target proteins and rare disorders. We believe that the proposed methodology can be useful to investigate multiple biological systems in which directed edges are probabilistic in nature, both in natural systems or when determined with large uncertainties in-silico.
Collapse
Affiliation(s)
- Yusuke Tokuhara
- Department of Information Science, Faculty of Science, Toho University, Funabashi, Chiba, Japan
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto, Uji, Japan
| | | | - Jose C Nacher
- Department of Information Science, Faculty of Science, Toho University, Funabashi, Chiba, Japan.
| |
Collapse
|
6
|
Someya W, Akutsu T, Nacher JC. Target control of linear directed networks based on the path cover problem. Sci Rep 2024; 14:16881. [PMID: 39043768 PMCID: PMC11266607 DOI: 10.1038/s41598-024-67442-7] [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: 04/03/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Securing complete control of complex systems comprised of tens of thousands of interconnected nodes holds immense significance across various fields, spanning from cell biology and brain science to human-engineered systems. However, depending on specific functional requirements, it can be more practical and efficient to focus on a pre-defined subset of nodes for control, a concept known as target control. While some methods have been proposed to find the smallest driver node set for target control, they either rely on heuristic approaches based on k-walk theory, lacking a guarantee of optimal solutions, or they are overly complex and challenging to implement in real-world networks. To address this challenge, we introduce a simple and elegant algorithm, inspired by the path cover problem, which efficiently identifies the nodes required to control a target node set within polynomial time. To practically apply the algorithm in real-world systems, we have selected several networks in which a specific set of nodes with functional significance can be designated as a target control set. The analysed systems include the complete connectome of the nematode worm C. elegans, the recently disclosed connectome of the Drosophila larval brain, as well as dozens of genome-wide metabolic networks spanning major plant lineages. The target control analysis shed light on distinctions between neural systems in nematode worms and larval brain insects, particularly concerning the number of nodes necessary to regulate specific functional systems. Furthermore, our analysis uncovers evolutionary trends within plant lineages, notably when examining the proportion of nodes required to control functional pathways.
Collapse
Affiliation(s)
- Wataru Someya
- Department of Information Science, Faculty of Science, Toho University, Funabashi, Chiba, 274-8510, Japan
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto, Uji, 611-0011, Japan
| | - Jose C Nacher
- Department of Information Science, Faculty of Science, Toho University, Funabashi, Chiba, 274-8510, Japan.
| |
Collapse
|
7
|
Wei PJ, Zhu AD, Cao R, Zheng C. Personalized Driver Gene Prediction Using Graph Convolutional Networks with Conditional Random Fields. BIOLOGY 2024; 13:184. [PMID: 38534453 DOI: 10.3390/biology13030184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/03/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
Cancer is a complex and evolutionary disease mainly driven by the accumulation of genetic variations in genes. Identifying cancer driver genes is important. However, most related studies have focused on the population level. Cancer is a disease with high heterogeneity. Thus, the discovery of driver genes at the individual level is becoming more valuable but is a great challenge. Although there have been some computational methods proposed to tackle this challenge, few can cover all patient samples well, and there is still room for performance improvement. In this study, to identify individual-level driver genes more efficiently, we propose the PDGCN method. PDGCN integrates multiple types of data features, including mutation, expression, methylation, copy number data, and system-level gene features, along with network structural features extracted using Node2vec in order to construct a sample-gene interaction network. Prediction is performed using a graphical convolutional neural network model with a conditional random field layer, which is able to better combine the network structural features with biological attribute features. Experiments on the ACC (Adrenocortical Cancer) and KICH (Kidney Chromophobe) datasets from TCGA (The Cancer Genome Atlas) demonstrated that the method performs better compared to other similar methods. It can identify not only frequently mutated driver genes, but also rare candidate driver genes and novel biomarker genes. The results of the survival and enrichment analyses of these detected genes demonstrate that the method can identify important driver genes at the individual level.
Collapse
Affiliation(s)
- Pi-Jing Wei
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Hefei 230601, China
| | - An-Dong Zhu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Hefei 230601, China
| | - Ruifen Cao
- School of Computer Science and Technology, Anhui University, 111 Jiulong Road, Hefei 230601, China
| | - Chunhou Zheng
- School of Artificial Intelligence, Anhui University, 111 Jiulong Road, Hefei 230601, China
| |
Collapse
|
8
|
Deschildre J, Vandemoortele B, Loers JU, De Preter K, Vermeirssen V. Evaluation of single-sample network inference methods for precision oncology. NPJ Syst Biol Appl 2024; 10:18. [PMID: 38360881 PMCID: PMC10869342 DOI: 10.1038/s41540-024-00340-w] [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: 07/11/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
A major challenge in precision oncology is to detect targetable cancer vulnerabilities in individual patients. Modeling high-throughput omics data in biological networks allows identifying key molecules and processes of tumorigenesis. Traditionally, network inference methods rely on many samples to contain sufficient information for learning, resulting in aggregate networks. However, to implement patient-tailored approaches in precision oncology, we need to interpret omics data at the level of individual patients. Several single-sample network inference methods have been developed that infer biological networks for an individual sample from bulk RNA-seq data. However, only a limited comparison of these methods has been made and many methods rely on 'normal tissue' samples as reference, which are not always available. Here, we conducted an evaluation of the single-sample network inference methods SSN, LIONESS, SWEET, iENA, CSN and SSPGI using transcriptomic profiles of lung and brain cancer cell lines from the CCLE database. The methods constructed functional gene networks with distinct network characteristics. Hub gene analyses revealed different degrees of subtype-specificity across methods. Single-sample networks were able to distinguish between tumor subtypes, as exemplified by node strength clustering, enrichment of known subtype-specific driver genes among hubs and differential node strength. We also showed that single-sample networks correlated better to other omics data from the same cell line as compared to aggregate networks. We conclude that single-sample network inference methods can reflect sample-specific biology when 'normal tissue' samples are absent and we point out peculiarities of each method.
Collapse
Affiliation(s)
- Joke Deschildre
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Boris Vandemoortele
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jens Uwe Loers
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katleen De Preter
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Lab of Translational Onco-genomics and Bio-informatics, Center for Medical Biotechnology (VIB-UGent), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Vanessa Vermeirssen
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
9
|
Huang Y, Chen F, Sun H, Zhong C. Exploring gene-patient association to identify personalized cancer driver genes by linear neighborhood propagation. BMC Bioinformatics 2024; 25:34. [PMID: 38254011 PMCID: PMC10804660 DOI: 10.1186/s12859-024-05662-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/18/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Driver genes play a vital role in the development of cancer. Identifying driver genes is critical for diagnosing and understanding cancer. However, challenges remain in identifying personalized driver genes due to tumor heterogeneity of cancer. Although many computational methods have been developed to solve this problem, few efforts have been undertaken to explore gene-patient associations to identify personalized driver genes. RESULTS Here we propose a method called LPDriver to identify personalized cancer driver genes by employing linear neighborhood propagation model on individual genetic data. LPDriver builds personalized gene network based on the genetic data of individual patients, extracts the gene-patient associations from the bipartite graph of the personalized gene network and utilizes a linear neighborhood propagation model to mine gene-patient associations to detect personalized driver genes. The experimental results demonstrate that as compared to the existing methods, our method shows competitive performance and can predict cancer driver genes in a more accurate way. Furthermore, these results also show that besides revealing novel driver genes that have been reported to be related with cancer, LPDriver is also able to identify personalized cancer driver genes for individual patients by their network characteristics even if the mutation data of genes are hidden. CONCLUSIONS LPDriver can provide an effective approach to predict personalized cancer driver genes, which could promote the diagnosis and treatment of cancer. The source code and data are freely available at https://github.com/hyr0771/LPDriver .
Collapse
Affiliation(s)
- Yiran Huang
- School of Computer, Electronics and Information, Guangxi University, Nanning, 530004, China
- Key Laboratory of Parallel, Distributed and Intelligent Computing in Guangxi Universities and Colleges, Guangxi University, Nanning, 530004, China
- Guangxi Key Laboratory of Multimedia Communications and Network Technology, Guangxi University, Nanning, 530004, China
| | - Fuhao Chen
- School of Computer, Electronics and Information, Guangxi University, Nanning, 530004, China
| | - Hongtao Sun
- School of Computer, Electronics and Information, Guangxi University, Nanning, 530004, China
| | - Cheng Zhong
- School of Computer, Electronics and Information, Guangxi University, Nanning, 530004, China.
- Key Laboratory of Parallel, Distributed and Intelligent Computing in Guangxi Universities and Colleges, Guangxi University, Nanning, 530004, China.
- Guangxi Key Laboratory of Multimedia Communications and Network Technology, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
10
|
Someya W, Akutsu T, Schwartz JM, Nacher JC. Measuring criticality in control of complex biological networks. NPJ Syst Biol Appl 2024; 10:9. [PMID: 38245555 PMCID: PMC10799883 DOI: 10.1038/s41540-024-00333-9] [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: 03/02/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Recent controllability analyses have demonstrated that driver nodes tend to be associated to genes related to important biological functions as well as human diseases. While researchers have focused on identifying critical nodes, intermittent nodes have received much less attention. Here, we propose a new efficient algorithm based on the Hamming distance for computing the importance of intermittent nodes using a Minimum Dominating Set (MDS)-based control model. We refer to this metric as criticality. The application of the proposed algorithm to compute criticality under the MDS control framework allows us to unveil the biological importance and roles of the intermittent nodes in different network systems, from cellular level such as signaling pathways and cell-cell interactions such as cytokine networks, to the complete nervous system of the nematode worm C. elegans. Taken together, the developed computational tools may open new avenues for investigating the role of intermittent nodes in many biological systems of interest in the context of network control.
Collapse
Affiliation(s)
- Wataru Someya
- Department of Information Science, Faculty of Science, Toho University, Funabashi, Chiba, 274-8510, Japan
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto, Uji, 611-0011, Japan
| | - Jean-Marc Schwartz
- School of Biological Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Jose C Nacher
- Department of Information Science, Faculty of Science, Toho University, Funabashi, Chiba, 274-8510, Japan.
| |
Collapse
|
11
|
Angarita-Rodríguez A, González-Giraldo Y, Rubio-Mesa JJ, Aristizábal AF, Pinzón A, González J. Control Theory and Systems Biology: Potential Applications in Neurodegeneration and Search for Therapeutic Targets. Int J Mol Sci 2023; 25:365. [PMID: 38203536 PMCID: PMC10778851 DOI: 10.3390/ijms25010365] [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: 10/21/2023] [Revised: 12/01/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Control theory, a well-established discipline in engineering and mathematics, has found novel applications in systems biology. This interdisciplinary approach leverages the principles of feedback control and regulation to gain insights into the complex dynamics of cellular and molecular networks underlying chronic diseases, including neurodegeneration. By modeling and analyzing these intricate systems, control theory provides a framework to understand the pathophysiology and identify potential therapeutic targets. Therefore, this review examines the most widely used control methods in conjunction with genomic-scale metabolic models in the steady state of the multi-omics type. According to our research, this approach involves integrating experimental data, mathematical modeling, and computational analyses to simulate and control complex biological systems. In this review, we find that the most significant application of this methodology is associated with cancer, leaving a lack of knowledge in neurodegenerative models. However, this methodology, mainly associated with the Minimal Dominant Set (MDS), has provided a starting point for identifying therapeutic targets for drug development and personalized treatment strategies, paving the way for more effective therapies.
Collapse
Affiliation(s)
- Andrea Angarita-Rodríguez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Edf. Carlos Ortiz, Oficina 107, Cra. 7 40-62, Bogotá 110231, Colombia; (A.A.-R.); (Y.G.-G.); (A.F.A.)
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Yeimy González-Giraldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Edf. Carlos Ortiz, Oficina 107, Cra. 7 40-62, Bogotá 110231, Colombia; (A.A.-R.); (Y.G.-G.); (A.F.A.)
| | - Juan J. Rubio-Mesa
- Departamento de Estadística, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Andrés Felipe Aristizábal
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Edf. Carlos Ortiz, Oficina 107, Cra. 7 40-62, Bogotá 110231, Colombia; (A.A.-R.); (Y.G.-G.); (A.F.A.)
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Edf. Carlos Ortiz, Oficina 107, Cra. 7 40-62, Bogotá 110231, Colombia; (A.A.-R.); (Y.G.-G.); (A.F.A.)
| |
Collapse
|
12
|
Xu J, Pang B, Lan Y, Dou R, Wang S, Kang S, Zhang W, Liu Y, Zhang Y, Ping Y. Identifying the personalized driver gene sets maximally contributing to abnormality of transcriptome phenotype in glioblastoma multiforme individuals. Mol Oncol 2023; 17:2472-2490. [PMID: 37491836 PMCID: PMC10620122 DOI: 10.1002/1878-0261.13499] [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: 12/02/2022] [Revised: 06/21/2023] [Accepted: 07/24/2023] [Indexed: 07/27/2023] Open
Abstract
High heterogeneity in genome and phenotype of cancer populations made it difficult to apply population-based common driver genes to the diagnosis and treatment of cancer individuals. Characterizing and identifying the personalized driver mechanism for glioblastoma multiforme (GBM) individuals were pivotal for the realization of precision medicine. We proposed an integrative method to identify the personalized driver gene sets by integrating the profiles of gene expression and genetic alterations in cancer individuals. This method coupled genetic algorithm and random walk to identify the optimal gene sets that could explain abnormality of transcriptome phenotype to the maximum extent. The personalized driver gene sets were identified for 99 GBM individuals using our method. We found that genomic alterations in between one and seven driver genes could maximally and cumulatively explain the dysfunction of cancer hallmarks across GBM individuals. The driver gene sets were distinct even in GBM individuals with significantly similar transcriptomic phenotypes. Our method identified MCM4 with rare genetic alterations as previously unknown oncogenic genes, the high expression of which were significantly associated with poor GBM prognosis. The functional experiments confirmed that knockdown of MCM4 could significantly inhibit proliferation, invasion, migration, and clone formation of the GBM cell lines U251 and U118MG, and overexpression of MCM4 significantly promoted the proliferation, invasion, migration, and clone formation of the GBM cell line U87MG. Our method could dissect the personalized driver genetic alteration sets that are pivotal for developing targeted therapy strategies and precision medicine. Our method could be extended to identify key drivers from other levels and could be applied to more cancer types.
Collapse
Affiliation(s)
- Jinyuan Xu
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Bo Pang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Yujia Lan
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Renjie Dou
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Shuai Wang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Shaobo Kang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Wanmei Zhang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Yuanyuan Liu
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Yijing Zhang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| | - Yanyan Ping
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityChina
| |
Collapse
|
13
|
Gillman R, Field MA, Schmitz U, Karamatic R, Hebbard L. Identifying cancer driver genes in individual tumours. Comput Struct Biotechnol J 2023; 21:5028-5038. [PMID: 37867967 PMCID: PMC10589724 DOI: 10.1016/j.csbj.2023.10.019] [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: 07/28/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/24/2023] Open
Abstract
Cancer is a heterogeneous disease with a strong genetic component making it suitable for precision medicine approaches aimed at identifying the underlying molecular drivers within a tumour. Large scale population-level cancer sequencing consortia have identified many actionable mutations common across both cancer types and sub-types, resulting in an increasing number of successful precision medicine programs. Nonetheless, such approaches fail to consider the effects of mutations unique to an individual patient and may miss rare driver mutations, necessitating personalised approaches to driver-gene prioritisation. One approach is to quantify the functional importance of individual mutations in a single tumour based on how they affect the expression of genes in a gene interaction network (GIN). These GIN-based approaches can be broadly divided into those that utilise an existing reference GIN and those that construct de novo patient-specific GINs. These single-tumour approaches have several limitations that likely influence their results, such as use of reference cohort data, network choice, and approaches to mathematical approximation, and more research is required to evaluate the in vitro and in vivo applicability of their predictions. This review examines the current state of the art methods that identify driver genes in single tumours with a focus on GIN-based driver prioritisation.
Collapse
Affiliation(s)
- Rhys Gillman
- Department of Biomedical Sciences and Molecular and Cell Biology, College of Public Health, Medical, and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, Queensland, Australia
| | - Matt A. Field
- Department of Biomedical Sciences and Molecular and Cell Biology, College of Public Health, Medical, and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, Queensland, Australia
- Immunogenomics Lab, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Ulf Schmitz
- Department of Biomedical Sciences and Molecular and Cell Biology, College of Public Health, Medical, and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, Queensland, Australia
| | - Rozemary Karamatic
- Gastroenterology and Hepatology, Townsville University Hospital, PO Box 670, Townsville, Queensland 4810, Australia
- College of Medicine and Dentistry, Division of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Lionel Hebbard
- Department of Biomedical Sciences and Molecular and Cell Biology, College of Public Health, Medical, and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, Queensland, Australia
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
- Australian Institute for Tropical Health and Medicine, Townsville, Queensland, Australia
| |
Collapse
|
14
|
Liang J, Li ZW, Sun ZN, Bi Y, Cheng H, Zeng T, Guo WF. Latent space search based multimodal optimization with personalized edge-network biomarker for multi-purpose early disease prediction. Brief Bioinform 2023; 24:bbad364. [PMID: 37833844 DOI: 10.1093/bib/bbad364] [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] [Received: 05/29/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Considering that cancer is resulting from the comutation of several essential genes of individual patients, researchers have begun to focus on identifying personalized edge-network biomarkers (PEBs) using personalized edge-network analysis for clinical practice. However, most of existing methods ignored the optimization of PEBs when multimodal biomarkers exist in multi-purpose early disease prediction (MPEDP). To solve this problem, this study proposes a novel model (MMPDENB-RBM) that combines personalized dynamic edge-network biomarkers (PDENB) theory, multimodal optimization strategy and latent space search scheme to identify biomarkers with different configurations of PDENB modules (i.e. to effectively identify multimodal PDENBs). The application to the three largest cancer omics datasets from The Cancer Genome Atlas database (i.e. breast invasive carcinoma, lung squamous cell carcinoma and lung adenocarcinoma) showed that the MMPDENB-RBM model could more effectively predict critical cancer state compared with other advanced methods. And, our model had better convergence, diversity and multimodal property as well as effective optimization ability compared with the other state-of-art methods. Particularly, multimodal PDENBs identified were more enriched with different functional biomarkers simultaneously, such as tissue-specific synthetic lethality edge-biomarkers including cancer driver genes and disease marker genes. Importantly, as our aim, these multimodal biomarkers can perform diverse biological and biomedical significances for drug target screen, survival risk assessment and novel biomedical sight as the expected multi-purpose of personalized early disease prediction. In summary, the present study provides multimodal property of PDENBs, especially the therapeutic biomarkers with more biological significances, which can help with MPEDP of individual cancer patients.
Collapse
Affiliation(s)
- Jing Liang
- School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Intelligent Agricultural Power Equipment, Zhengzhou University, Luoyang 471000, China
| | - Zong-Wei Li
- School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Ze-Ning Sun
- School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Ying Bi
- School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Han Cheng
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tao Zeng
- Guangzhou National Laboratory, Guangzhou 510005, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, 510005, Guangzhou Medical University
| | - Wei-Feng Guo
- School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Intelligent Agricultural Power Equipment, Zhengzhou University, Luoyang 471000, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center,Guangzhou 7510060, China
| |
Collapse
|
15
|
Li Y, Zhang SW, Xie MY, Zhang T. PhenoDriver: interpretable framework for studying personalized phenotype-associated driver genes in breast cancer. Brief Bioinform 2023; 24:bbad291. [PMID: 37738403 DOI: 10.1093/bib/bbad291] [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] [Received: 04/02/2023] [Revised: 07/12/2023] [Accepted: 07/27/2023] [Indexed: 09/24/2023] Open
Abstract
Identifying personalized cancer driver genes and further revealing their oncogenic mechanisms is critical for understanding the mechanisms of cell transformation and aiding clinical diagnosis. Almost all existing methods primarily focus on identifying driver genes at the cohort or individual level but fail to further uncover their underlying oncogenic mechanisms. To fill this gap, we present an interpretable framework, PhenoDriver, to identify personalized cancer driver genes, elucidate their roles in cancer development and uncover the association between driver genes and clinical phenotypic alterations. By analyzing 988 breast cancer patients, we demonstrate the outstanding performance of PhenoDriver in identifying breast cancer driver genes at the cohort level compared to other state-of-the-art methods. Otherwise, our PhenoDriver can also effectively identify driver genes with both recurrent and rare mutations in individual patients. We further explore and reveal the oncogenic mechanisms of some known and unknown breast cancer driver genes (e.g. TP53, MAP3K1, HTT, etc.) identified by PhenoDriver, and construct their subnetworks for regulating clinical abnormal phenotypes. Notably, most of our findings are consistent with existing biological knowledge. Based on the personalized driver profiles, we discover two existing and one unreported breast cancer subtypes and uncover their molecular mechanisms. These results intensify our understanding for breast cancer mechanisms, guide therapeutic decisions and assist in the development of targeted anticancer therapies.
Collapse
Affiliation(s)
- Yan Li
- School of Automation from Northwestern Polytechnical University, China
| | - Shao-Wu Zhang
- School of Automation from Northwestern Polytechnical University, China
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, China
| | - Ming-Yu Xie
- School of Automation from Northwestern Polytechnical University, China
| | - Tong Zhang
- School of Automation from Northwestern Polytechnical University, China
| |
Collapse
|
16
|
Meng P, Wang G, Guo H, Jiang T. Identifying cancer driver genes using a two-stage random walk with restart on a gene interaction network. Comput Biol Med 2023; 158:106810. [PMID: 37011433 DOI: 10.1016/j.compbiomed.2023.106810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
Cancer development and progression are significantly influenced by cancer driver genes. Understanding cancer driver genes and their mechanisms of action is essential for developing effective cancer treatments. As a result, identifying driver genes is important for drug development, cancer diagnosis, and treatment. Here, we present an algorithm to discover driver genes based on the two-stage random walk with restart (RWR), and the modified method for calculating the transition probability matrix in random walk algorithm. First, we performed the first stage of RWR on the whole gene interaction network, in which we employ a new method for calculating the transition probability matrix and extracted the subnetwork based on nodes that had a high correlation with the seed nodes. The subnetwork was then applied to the second stage of RWR and the nodes were re-ranked in the subnetwork. Our approach outperformed existing methods in identifying driver genes. The outcome of the effect of three gene interaction networks, two rounds of random walk, and the seed nodes' sensitivity were all compared at the same time. In addition, we identified several potential driver genes, some of which are involved in driving cancer development. Overall, our method is efficient in various cancer types, significantly outperforms existing methods, and can identify possible driver genes.
Collapse
|
17
|
Chen HH, Hsueh CW, Lee CH, Hao TY, Tu TY, Chang LY, Lee JC, Lin CY. SWEET: a single-sample network inference method for deciphering individual features in disease. Brief Bioinform 2023; 24:7017366. [PMID: 36719112 PMCID: PMC10025435 DOI: 10.1093/bib/bbad032] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/05/2023] [Accepted: 01/14/2023] [Indexed: 02/01/2023] Open
Abstract
Recently, extracting inherent biological system information (e.g. cellular networks) from genome-wide expression profiles for developing personalized diagnostic and therapeutic strategies has become increasingly important. However, accurately constructing single-sample networks (SINs) to capture individual characteristics and heterogeneity in disease remains challenging. Here, we propose a sample-specific-weighted correlation network (SWEET) method to model SINs by integrating the genome-wide sample-to-sample correlation (i.e. sample weights) with the differential network between perturbed and aggregate networks. For a group of samples, the genome-wide sample weights can be assessed without prior knowledge of intrinsic subpopulations to address the network edge number bias caused by sample size differences. Compared with the state-of-the-art SIN inference methods, the SWEET SINs in 16 cancers more likely fit the scale-free property, display higher overlap with the human interactomes and perform better in identifying three types of cancer-related genes. Moreover, integrating SWEET SINs with a network proximity measure facilitates characterizing individual features and therapy in diseases, such as somatic mutation, mut-driver and essential genes. Biological experiments further validated two candidate repurposable drugs, albendazole for head and neck squamous cell carcinoma (HNSCC) and lung adenocarcinoma (LUAD) and encorafenib for HNSCC. By applying SWEET, we also identified two possible LUAD subtypes that exhibit distinct clinical features and molecular mechanisms. Overall, the SWEET method complements current SIN inference and analysis methods and presents a view of biological systems at the network level to offer numerous clues for further investigation and clinical translation in network medicine and precision medicine.
Collapse
Affiliation(s)
- Hsin-Hua Chen
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Chun-Wei Hsueh
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Chia-Hwa Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ting-Yi Hao
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Tzu-Ying Tu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Lan-Yun Chang
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Jih-Chin Lee
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 110, Taiwan
| | - Chun-Yu Lin
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Institute of Data Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
18
|
Cheng X, Amanullah M, Liu W, Liu Y, Pan X, Zhang H, Xu H, Liu P, Lu Y. WMDS.net: a network control framework for identifying key players in transcriptome programs. Bioinformatics 2023; 39:7023921. [PMID: 36727489 PMCID: PMC9925106 DOI: 10.1093/bioinformatics/btad071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Mammalian cells can be transcriptionally reprogramed to other cellular phenotypes. Controllability of such complex transitions in transcriptional networks underlying cellular phenotypes is an inherent biological characteristic. This network controllability can be interpreted by operating a few key regulators to guide the transcriptional program from one state to another. Finding the key regulators in the transcriptional program can provide key insights into the network state transition underlying cellular phenotypes. RESULTS To address this challenge, here, we proposed to identify the key regulators in the transcriptional co-expression network as a minimum dominating set (MDS) of driver nodes that can fully control the network state transition. Based on the theory of structural controllability, we developed a weighted MDS network model (WMDS.net) to find the driver nodes of differential gene co-expression networks. The weight of WMDS.net integrates the degree of nodes in the network and the significance of gene co-expression difference between two physiological states into the measurement of node controllability of the transcriptional network. To confirm its validity, we applied WMDS.net to the discovery of cancer driver genes in RNA-seq datasets from The Cancer Genome Atlas. WMDS.net is powerful among various cancer datasets and outperformed the other top-tier tools with a better balance between precision and recall. AVAILABILITY AND IMPLEMENTATION https://github.com/chaofen123/WMDS.net. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xiang Cheng
- Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310006, China.,Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| | - Md Amanullah
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China.,Department of Respiratory Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Weigang Liu
- Department of Respiratory Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yi Liu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China.,Department of Respiratory Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaoqing Pan
- Department of Mathematics, Shanghai Normal University, Xuhui 200234, China
| | - Honghe Zhang
- Department of Pathology, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Haiming Xu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| | - Pengyuan Liu
- Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310006, China.,Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cancer Center, Zhejiang University, Hangzhou 310029, China
| | - Yan Lu
- Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310006, China.,Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China.,Cancer Center, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
19
|
Guan S, Yu YN, Li B, Gu H, Chen L, Wang N, Wang B, Liu X, Liu J, Wang Z. Discovery of Drug-Responsive Phenomic Alteration-Related Driver Genes in the Treatment of Coronary Heart Disease. Pharmgenomics Pers Med 2023; 16:201-217. [PMID: 36945217 PMCID: PMC10024908 DOI: 10.2147/pgpm.s398522] [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: 11/24/2022] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Background The Xueyu Zheng (XYZ) phenome is central to coronary heart disease (CHD), but efforts to detect genetic associations in the XYZ phenome have been disappointing. Methods The phenomic alteration-related genes (PARGs) for the XYZ phenome were screened using |ρ| > 0.4 and p < 0.05 after treatment with Danhong injection at day 14 and day 30. Then, the driver genes for the Protein-Protein Interaction (PPI) networks of the PARGs established using STRING 11.0 were detected using a personalized network control algorithm (PNC). Finally, the molecular correlations of the driver genes with the XYZ phenome were analyzed with the Gene Ontology (GO) biological processes and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways from a holistic viewpoint. Results A total of 525 and 309 PARGs in the XYZ phenome at day 14 and day 30 were identified. These genes were separately enriched in 48 and 35 pathways. Furthermore, five driver genes were detected. These genes were mainly correlated with endoplasmic reticulum stress-mediated apoptosis and autophagy regulation, which could suppress atherosclerosis progression. Conclusion Our study detected the drug-responsive PARGs of the XYZ phenome in CHD and provides an exemplary strategy to investigate the genetic associations among this common phenome and its component symptoms in patients with CHD. Trial Registration ClinicalTrials.gov, NCT01681316; registered on September 7, 2012.
Collapse
Affiliation(s)
- Shuang Guan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Ya-Nan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Hao Gu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Lin Chen
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Nian Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Bo Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xi Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
- Correspondence: Zhong Wang; Jun Liu, Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimennei, Beijing, People’s Republic of China, Email ;
| |
Collapse
|
20
|
Zhang SW, Xu JY, Zhang T. DGMP: Identifying Cancer Driver Genes by Jointing DGCN and MLP from Multi-omics Genomic Data. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:928-938. [PMID: 36464123 PMCID: PMC10025764 DOI: 10.1016/j.gpb.2022.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 12/03/2022]
Abstract
Identification of cancer driver genes plays an important role in precision oncology research, which is helpful to understand cancer initiation and progression. However, most existing computational methods mainly used the protein-protein interaction (PPI) networks, or treated the directed gene regulatory networks (GRNs) as the undirected gene-gene association networks to identify the cancer driver genes, which will lose the unique structure regulatory information in the directed GRNs, and then affect the outcome of the cancer driver gene identification. Here, based on the multi-omics pan-cancer data (i.e., gene expression, mutation, copy number variation, and DNA methylation), we propose a novel method (called DGMP) to identify cancer driver genes by jointing directed graph convolutional network (DGCN) and multilayer perceptron (MLP). DGMP learns the multi-omics features of genes as well as the topological structure features in GRN with the DGCN model and uses MLP to weigh more on gene features for mitigating the bias toward the graph topological features in the DGCN learning process. The results on three GRNs show that DGMP outperforms other existing state-of-the-art methods. The ablation experimental results on the DawnNet network indicate that introducing MLP into DGCN can offset the performance degradation of DGCN, and jointing MLP and DGCN can effectively improve the performance of identifying cancer driver genes. DGMP can identify not only the highly mutated cancer driver genes but also the driver genes harboring other kinds of alterations (e.g., differential expression and aberrant DNA methylation) or genes involved in GRNs with other cancer genes. The source code of DGMP can be freely downloaded from https://github.com/NWPU-903PR/DGMP.
Collapse
Affiliation(s)
- Shao-Wu Zhang
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Jing-Yu Xu
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tong Zhang
- MOE Key Laboratory of Information Fusion Technology, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
21
|
Zhang SW, Wang ZN, Li Y, Guo WF. Prioritization of cancer driver gene with prize-collecting steiner tree by introducing an edge weighted strategy in the personalized gene interaction network. BMC Bioinformatics 2022; 23:341. [PMID: 35974311 PMCID: PMC9380343 DOI: 10.1186/s12859-022-04802-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background Cancer is a heterogeneous disease in which tumor genes cooperate as well as adapt and evolve to the changing conditions for individual patients. It is a meaningful task to discover the personalized cancer driver genes that can provide diagnosis and target drug for individual patients. However, most of existing methods mainly ranks potential personalized cancer driver genes by considering the patient-specific nodes information on the gene/protein interaction network. These methods ignore the personalized edge weight information in gene interaction network, leading to false positive results. Results In this work, we presented a novel algorithm (called PDGPCS) to predict the Personalized cancer Driver Genes based on the Prize-Collecting Steiner tree model by considering the personalized edge weight information. PDGPCS first constructs the personalized weighted gene interaction network by integrating the personalized gene expression data and prior known gene/protein interaction network knowledge. Then the gene mutation data and pathway data are integrated to quantify the impact of each mutant gene on every dysregulated pathway with the prize-collecting Steiner tree model. Finally, according to the mutant gene’s aggregated impact score on all dysregulated pathways, the mutant genes are ranked for prioritizing the personalized cancer driver genes. Experimental results on four TCGA cancer datasets show that PDGPCS has better performance than other personalized driver gene prediction methods. In addition, we verified that the personalized edge weight of gene interaction network can improve the prediction performance. Conclusions PDGPCS can more accurately identify the personalized driver genes and takes a step further toward personalized medicine and treatment. The source code of PDGPCS can be freely downloaded from https://github.com/NWPU-903PR/PDGPCS. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04802-y.
Collapse
Affiliation(s)
- Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Zhen-Nan Wang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yan Li
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
22
|
Zhu Y, Zhang H, Yang Y, Zhang C, Ou-Yang L, Bai L, Deng M, Yi M, Liu S, Wang C. Discovery of pan-cancer related genes via integrative network analysis. Brief Funct Genomics 2022; 21:325-338. [PMID: 35760070 DOI: 10.1093/bfgp/elac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/14/2022] [Accepted: 05/25/2022] [Indexed: 01/02/2023] Open
Abstract
Identification of cancer-related genes is helpful for understanding the pathogenesis of cancer, developing targeted drugs and creating new diagnostic and therapeutic methods. Considering the complexity of the biological laboratory methods, many network-based methods have been proposed to identify cancer-related genes at the global perspective with the increasing availability of high-throughput data. Some studies have focused on the tissue-specific cancer networks. However, cancers from different tissues may share common features, and those methods may ignore the differences and similarities across cancers during the establishment of modeling. In this work, in order to make full use of global information of the network, we first establish the pan-cancer network via differential network algorithm, which not only contains heterogeneous data across multiple cancer types but also contains heterogeneous data between tumor samples and normal samples. Second, the node representation vectors are learned by network embedding. In contrast to ranking analysis-based methods, with the help of integrative network analysis, we transform the cancer-related gene identification problem into a binary classification problem. The final results are obtained via ensemble classification. We further applied these methods to the most commonly used gene expression data involving six tissue-specific cancer types. As a result, an integrative pan-cancer network and several biologically meaningful results were obtained. As examples, nine genes were ultimately identified as potential pan-cancer-related genes. Most of these genes have been reported in published studies, thus showing our method's potential for application in identifying driver gene candidates for further biological experimental verification.
Collapse
Affiliation(s)
- Yuan Zhu
- School of Automation, China University of Geosciences, Lumo Road, 430074, Wuhan, China
- Hubei Key Laboratory of Advanced Control and Intelligent Automation for Complex Systems, Lumo Road, 430074, Wuhan, China
- Engineering Research Center of Intelligent Technology for Geo-Exploration, Lumo Road, 430074, Wuhan, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence(Fudan University), Ministry of Education, Handan Road, 200433, Shanghai, China
| | - Houwang Zhang
- Electrical Engineering, City University of HongKong, Kowloon, 999077, HongKong, China
| | - Yuanhang Yang
- School of Mathematics and Physics, China University of Geosciences, Lumo Road, 430074, Wuhan, China
| | - Chaoyang Zhang
- School of Computing Sciences and Computer Engineering, The University of Southern Mississippi, Hattiesburg, USA
| | - Le Ou-Yang
- Guangdong Key Laboratory of Intelligent Information Processing and Shenzhen Key Laboratory of Media Security, Shenzhen University, Nanhai Avenue, 518060, Shenzhen, China
| | - Litai Bai
- School of Automation, China University of Geosciences, Lumo Road, 430074, Wuhan, China
- Hubei Key Laboratory of Advanced Control and Intelligent Automation for Complex Systems, Lumo Road, 430074, Wuhan, China
- Engineering Research Center of Intelligent Technology for Geo-Exploration, Lumo Road, 430074, Wuhan, China
| | - Minghua Deng
- School of Mathematical Sciences, Peking University, No.5 Yiheyuan Road, 100871, Beijing, China
| | - Ming Yi
- School of Mathematics and Physics, China University of Geosciences, Lumo Road, 430074, Wuhan, China
| | - Song Liu
- School of Automation, China University of Geosciences, Lumo Road, 430074, Wuhan, China
- Hubei Key Laboratory of Advanced Control and Intelligent Automation for Complex Systems, Lumo Road, 430074, Wuhan, China
- Engineering Research Center of Intelligent Technology for Geo-Exploration, Lumo Road, 430074, Wuhan, China
| | - Chao Wang
- Hepatic Surgery Center, Institute of Hepato-Pancreato-Biliary Surgery, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, 430030, Wuhan, China
| |
Collapse
|
23
|
Liang J, Li ZW, Yue CT, Hu Z, Cheng H, Liu ZX, Guo WF. Multi-modal optimization to identify personalized biomarkers for disease prediction of individual patients with cancer. Brief Bioinform 2022; 23:6647504. [PMID: 35858208 DOI: 10.1093/bib/bbac254] [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: 04/25/2022] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 11/14/2022] Open
Abstract
Finding personalized biomarkers for disease prediction of patients with cancer remains a massive challenge in precision medicine. Most methods focus on one subnetwork or module as a network biomarker; however, this ignores the early warning capabilities of other modules with different configurations of biomarkers (i.e. multi-modal personalized biomarkers). Identifying such modules would not only predict disease but also provide effective therapeutic drug target information for individual patients. To solve this problem, we developed a novel model (denoted multi-modal personalized dynamic network biomarkers (MMPDNB)) based on a multi-modal optimization mechanism and personalized dynamic network biomarker (PDNB) theory, which can provide multiple modules of personalized biomarkers and unveil their multi-modal properties. Using the genomics data of patients with breast or lung cancer from The Cancer Genome Atlas database, we validated the effectiveness of the MMPDNB model. The experimental results showed that compared with other advanced methods, MMPDNB can more effectively predict the critical state with the highest early warning signal score during cancer development. Furthermore, MMPDNB more significantly identified PDNBs containing driver and biomarker genes specific to cancer tissues. More importantly, we validated the biological significance of multi-modal PDNBs, which could provide effective drug targets of individual patients as well as markers for predicting early warning signals of the critical disease state. In conclusion, multi-modal optimization is an effective method to identify PDNBs and offers a new perspective for understanding tumor heterogeneity in cancer precision medicine.
Collapse
Affiliation(s)
- Jing Liang
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zong-Wei Li
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Cai-Tong Yue
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zhuo Hu
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Han Cheng
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
24
|
Liu Y, Li A, Liu J, Meng G, Wang M. TSDLPP: A Novel Two-Stage Deep Learning Framework For Prognosis Prediction Based on Whole Slide Histopathological Images. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2523-2532. [PMID: 33989155 DOI: 10.1109/tcbb.2021.3080295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Recently, digital pathology image-based prognosis prediction has become a hot topic in healthcare research to make early decisions on therapy and improve the treatment quality of patients. Therefore, there has been a recent surge of interest in designing deep learning method solving the problem of prognosis prediction with digital pathology images. However, whole slide histopathological images (WSIs) based prognosis prediction is still a challenge due to the large size of pathological images, the heterogeneity of tumors and the high cost of region of interests (ROIs) labeling. In this study, we design a novel two-stage deep learning framework for prognosis prediction (TSDLPP) based on WSIs. Our proposed framework consists of two-stage paradigms: 1) training tissue decomposition network (TDNet) to divide WSIs into cancerous and non-cancerous regions, 2) integrating general prognosis-related densely connected CNN (GPR-DCCNN) and morphology-specific prognosis-related densely connected CNNs (MSPR-DCCNNs) to extract different level features of pathological images. In the end, we apply TSDLPP to the prognosis prediction of breast cancer using The Cancer Genome Atlas (TCGA) datasets. Experiment results demonstrate that TSDLPP obtains superior performance of prognosis prediction compared with the existing state-of-arts methods.
Collapse
|
25
|
Yan J, Hu Z, Li ZW, Sun S, Guo WF. Network Control Models With Personalized Genomics Data for Understanding Tumor Heterogeneity in Cancer. Front Oncol 2022; 12:891676. [PMID: 35712516 PMCID: PMC9195174 DOI: 10.3389/fonc.2022.891676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022] Open
Abstract
Due to rapid development of high-throughput sequencing and biotechnology, it has brought new opportunities and challenges in developing efficient computational methods for exploring personalized genomics data of cancer patients. Because of the high-dimension and small sample size characteristics of these personalized genomics data, it is difficult for excavating effective information by using traditional statistical methods. In the past few years, network control methods have been proposed to solve networked system with high-dimension and small sample size. Researchers have made progress in the design and optimization of network control principles. However, there are few studies comprehensively surveying network control methods to analyze the biomolecular network data of individual patients. To address this problem, here we comprehensively surveyed complex network control methods on personalized omics data for understanding tumor heterogeneity in precision medicine of individual patients with cancer.
Collapse
Affiliation(s)
- Jipeng Yan
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhuo Hu
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
| | - Zong-Wei Li
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Wei-Feng Guo, ; Shiren Sun,
| | - Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Wei-Feng Guo, ; Shiren Sun,
| |
Collapse
|
26
|
Erten C, Houdjedj A, Kazan H, Taleb Bahmed AA. PersonaDrive: A Method for the Identification and Prioritization of Personalized Cancer Drivers. Bioinformatics 2022; 38:3407-3414. [PMID: 35579340 DOI: 10.1093/bioinformatics/btac329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION A major challenge in cancer genomics is to distinguish the driver mutations that are causally linked to cancer from passenger mutations that do not contribute to cancer development. The majority of existing methods provide a single driver gene list for the entire cohort of patients. However, since mutation profiles of patients from the same cancer type show a high degree of heterogeneity, a more ideal approach is to identify patient-specific drivers. RESULTS We propose a novel method that integrates genomic data, biological pathways, and protein connectivity information for personalized identification of driver genes. The method is formulated on a personalized bipartite graph for each patient. Our approach provides a personalized ranking of the mutated genes of a patient based on the sum of weighted 'pairwise pathway coverage' scores across all the samples, where appropriate pairwise patient similarity scores are used as weights to normalize these coverage scores. We compare our method against three state-of-the-art patient-specific cancer gene prioritization methods. The comparisons are with respect to a novel evaluation method that takes into account the personalized nature of the problem. We show that our approach outperforms the existing alternatives for both the TCGA and the cell line data. Additionally, we show that the KEGG/Reactome pathways enriched in our ranked genes and those that are enriched in cell lines' reference sets overlap significantly when compared to the overlaps achieved by the rankings of the alternative methods. Our findings can provide valuable information towards the development of personalized treatments and therapies. AVAILABILITY All the code and data are available at https://github.com/abu-compbio/PersonaDrive (archived at https://doi.org/10.5281/zenodo.6520187). SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Cesim Erten
- Department of Computer Engineering, Antalya Bilim University, Antalya, 07190, Turkey
| | - Aissa Houdjedj
- Department of Computer Engineering, Antalya Bilim University, Antalya, 07190, Turkey.,Department of Computer Engineering, Akdeniz University, Antalya, 07070, Turkey
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Antalya, 07190, Turkey
| | - Ahmed Amine Taleb Bahmed
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Antalya, 07190, Turkey
| |
Collapse
|
27
|
Li B, Wang Y, Gu H, Yu Y, Wang P, Liu J, Zhang Y, Chen Y, Niu Q, Wang B, Liu Q, Guan S, Li Y, Zhang H, Wang Z. Modular Screening Reveals Driver Induced Additive Mechanisms of Baicalin and Jasminoidin on Cerebral Ischemia Therapy. Front Cardiovasc Med 2022; 9:813983. [PMID: 35265682 PMCID: PMC8899124 DOI: 10.3389/fcvm.2022.813983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Combination therapy with increased efficacy and reduced toxicity plays a crucial role in treating complex diseases, such as stroke, but it remains an insurmountable barrier to elucidate the mechanisms of synergistic effects. Here, we present a Driver-induced Modular Screening (DiMS) strategy integrated synergistic module and driver gene identification to elucidate the additive mechanisms of Baicalin (BA) and Jasminoidin (JA) on cerebral ischemia (CI) therapy. Based on anti-ischemia genomic networks BA, JA, and their combination (BJ), we obtained 4, 3, and 9 On-modules of BA, JA, and BJ by modular similarity analysis. Compared with the monotherapy groups, four additive modules (Add-module, BJ_Mod-4, 7, 9, and 13), 15 driver genes of BJ were identified by modular similarity and network control methods, and seven driver proteins (PAQR8, RhoA, EMC10, GGA2, VIPR1, FAM120A, and SEMA3F) were validated by animal experiments. The functional analysis found neuroprotective roles of the Add-modules and driver genes, such as the Neurotrophin signaling pathway and FoxO signaling pathway, which may reflect the additive mechanisms of BJ. Moreover, such a DiMS paradigm provides a new angle to explore the synergistic mechanisms of combination therapy and screen multi-targeted drugs for complex diseases.
Collapse
Affiliation(s)
- Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Wang
- College of Nursing, Chengde Medical University, Chengde, China
| | - Hao Gu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yinying Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiong Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuang Guan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanda Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huamin Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Huamin Zhang
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Zhong Wang
| |
Collapse
|
28
|
Sun R, Zhou Y, Cui Q. Comparative analysis of aneurysm subtypes associated genes based on protein-protein interaction network. BMC Bioinformatics 2021; 22:587. [PMID: 34895131 PMCID: PMC8665538 DOI: 10.1186/s12859-021-04513-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/01/2021] [Indexed: 11/10/2022] Open
Abstract
The arterial aneurysm refers to localized dilation of blood vessel wall and is common in general population. The majority of aneurysm cases remains asymptomatic until a sudden rupture which is usually fatal and of extremely high mortality (~ 50-60%). Therefore, early diagnosis, prevention and management of aneurysm are in urgent need. Unfortunately, current understanding of disease driver genes of various aneurysm subtypes is still limited, and without appropriate biomarkers and drug targets no specialized drug has been developed for aneurysm treatment. In this research, aneurysm subtypes were analyzed based on protein-protein interaction network to better understand aneurysm pathogenesis. By measuring network-based proximity of aneurysm subtypes, we identified a relevant closest relationship between aortic aneurysm and aortic dissection. An improved random walk method was performed to prioritize candidate driver genes of each aneurysm subtype. Thereafter, transcriptomes of 6 human aneurysm subtypes were collected and differential expression genes were identified to further filter potential driver genes. Functional enrichment of above driver genes indicated a general role of ubiquitination and programmed cell death in aneurysm pathogenesis. Especially, we further observed participation of BCL-2-mediated apoptosis pathway and caspase-1 related pyroptosis in the development of cerebral aneurysm and aneurysmal subarachnoid hemorrhage in corresponding transcriptomes.
Collapse
Affiliation(s)
- Ruya Sun
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-Coding RNA Medicine, Peking University Health Science Center Beijing, Beijing, China.
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-Coding RNA Medicine, Peking University Health Science Center Beijing, Beijing, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-Coding RNA Medicine, Peking University Health Science Center Beijing, Beijing, China.
| |
Collapse
|
29
|
Dan Z, Chen Y, Li H, Zeng Y, Xu W, Zhao W, He R, Huang W. The metabolomic landscape of rice heterosis highlights pathway biomarkers for predicting complex phenotypes. PLANT PHYSIOLOGY 2021; 187:1011-1025. [PMID: 34608951 PMCID: PMC8491067 DOI: 10.1093/plphys/kiab273] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/27/2021] [Indexed: 06/13/2023]
Abstract
Understanding the molecular mechanisms underlying complex phenotypes requires systematic analyses of complicated metabolic networks and contributes to improvements in the breeding efficiency of staple cereal crops and diagnostic accuracy for human diseases. Here, we selected rice (Oryza sativa) heterosis as a complex phenotype and investigated the mechanisms of both vegetative and reproductive traits using an untargeted metabolomics strategy. Heterosis-associated analytes were identified, and the overlapping analytes were shown to underlie the association patterns for six agronomic traits. The heterosis-associated analytes of four yield components and plant height collectively contributed to yield heterosis, and the degree of contribution differed among the five traits. We performed dysregulated network analyses of the high- and low-better parent heterosis hybrids and found multiple types of metabolic pathways involved in heterosis. The metabolite levels of the significantly enriched pathways (especially those from amino acid and carbohydrate metabolism) were predictive of yield heterosis (area under the curve = 0.907 with 10 features), and the predictability of these pathway biomarkers was validated with hybrids across environments and populations. Our findings elucidate the metabolomic landscape of rice heterosis and highlight the potential application of pathway biomarkers in achieving accurate predictions of complex phenotypes.
Collapse
Affiliation(s)
- Zhiwu Dan
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yunping Chen
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hui Li
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yafei Zeng
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wuwu Xu
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Weibo Zhao
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ruifeng He
- Institute of Biological Chemistry, Washington State University, Pullman, Washington 99164-6414, USA
| | - Wenchao Huang
- State Key Laboratory of Hybrid Rice, Key Laboratory for Research and Utilization of Heterosis in Indica Rice, the Ministry of Agriculture, College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
30
|
Tang YY, Wei PJ, Zhao JP, Xia J, Cao RF, Zheng CH. Identification of driver genes based on gene mutational effects and network centrality. BMC Bioinformatics 2021; 22:457. [PMID: 34560840 PMCID: PMC8461858 DOI: 10.1186/s12859-021-04377-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 08/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As one of the deadliest diseases in the world, cancer is driven by a few somatic mutations that disrupt the normal growth of cells, and leads to abnormal proliferation and tumor development. The vast majority of somatic mutations did not affect the occurrence and development of cancer; thus, identifying the mutations responsible for tumor occurrence and development is one of the main targets of current cancer treatments. RESULTS To effectively identify driver genes, we adopted a semi-local centrality measure and gene mutation effect function to assess the effect of gene mutations on changes in gene expression patterns. Firstly, we calculated the mutation score for each gene. Secondly, we identified differentially expressed genes (DEGs) in the cohort by comparing the expression profiles of tumor samples and normal samples, and then constructed a local network for each mutation gene using DEGs and mutant genes according to the protein-protein interaction network. Finally, we calculated the score of each mutant gene according to the objective function. The top-ranking mutant genes were selected as driver genes. We name the proposed method as mutations effect and network centrality. CONCLUSIONS Four types of cancer data in The Cancer Genome Atlas were tested. The experimental data proved that our method was superior to the existing network-centric method, as it was able to quickly and easily identify driver genes and rare driver factors.
Collapse
Affiliation(s)
- Yun-Yun Tang
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| | - Pi-Jing Wei
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| | - Jian-Ping Zhao
- College of Mathematics and System Sciences, Xinjiang University, Urumqi, China
| | - Junfeng Xia
- Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Rui-Fen Cao
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China.,Engineering Research Center of Big Data Application in Private Health Medicine, Fujian Province University, Putian, Fujian, China
| | - Chun-Hou Zheng
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China. .,College of Mathematics and System Sciences, Xinjiang University, Urumqi, China.
| |
Collapse
|
31
|
Du XW, Li G, Liu J, Zhang CY, Liu Q, Wang H, Chen TS. Comprehensive analysis of the cancer driver genes in breast cancer demonstrates their roles in cancer prognosis and tumor microenvironment. World J Surg Oncol 2021; 19:273. [PMID: 34507558 PMCID: PMC8434726 DOI: 10.1186/s12957-021-02387-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/31/2021] [Indexed: 12/28/2022] Open
Abstract
Background Breast cancer is the most common malignancy in women. Cancer driver gene-mediated alterations in the tumor microenvironment are critical factors affecting the biological behavior of breast cancer. The purpose of this study was to identify the expression characteristics and prognostic value of cancer driver genes in breast cancer. Methods The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets are used as the training and test sets. Classified according to cancer and paracancerous tissues, we identified differentially expressed cancer driver genes. We further screened prognosis-associated genes, and candidate genes were submitted for the construction of a risk signature. Functional enrichment analysis and transcriptional regulatory networks were performed to search for possible mechanisms by which cancer driver genes affect breast cancer prognosis. Results We identified more than 200 differentially expressed driver genes and 27 prognosis-related genes. High-risk group patients had a lower survival rate compared to the low-risk group (P<0.05), and risk signature showed high specificity and sensitivity in predicting the patient prognosis (AUC 0.790). Multivariate regression analysis suggested that risk scores can independently predict patient prognosis. Further, we found differences in PD-1 expression, immune score, and stromal score among different risk groups. Conclusion Our study confirms the critical prognosis role of cancer driver genes in breast cancer. The cancer driver gene risk signature may provide a novel biomarker for clinical treatment strategy and survival prediction of breast cancer.
Collapse
Affiliation(s)
| | - Gao Li
- Department of Oncology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200000, China
| | - Juan Liu
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chun-Yan Zhang
- Department of Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiong Liu
- Office of Academic Research, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Wang
- Department of Oncology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200000, China.
| | - Ting-Song Chen
- Department of Oncology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200000, China.
| |
Collapse
|
32
|
Kan Y, Jiang L, Tang J, Guo Y, Guo F. A systematic view of computational methods for identifying driver genes based on somatic mutation data. Brief Funct Genomics 2021; 20:333-343. [PMID: 34312663 DOI: 10.1093/bfgp/elab032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 11/13/2022] Open
Abstract
Abnormal changes of driver genes are serious for human health and biomedical research. Identifying driver genes, exactly from enormous genes with mutations, promotes accurate diagnosis and treatment of cancer. A lot of works about uncovering driver genes have been developed over the past decades. By analyzing previous works, we find that computational methods are more efficient than traditional biological experiments when distinguishing driver genes from massive data. In this study, we summarize eight common computational algorithms only using somatic mutation data. We first group these methods into three categories according to mutation features they apply. Then, we conclude a general process of nominating candidate cancer driver genes. Finally, we evaluate three representative methods on 10 kinds of cancer derived from The Cancer Genome Atlas Program and five Chinese projects from the International Cancer Genome Consortium. In addition, we compare results of methods with various parameters. Evaluation is performed from four perspectives, including CGC, OG/TSG, Q-value and QQQuantile-Quantileplot. To sum up, we present algorithms using somatic mutation data in order to offer a systematic view of various mutation features and lay the foundation of methods based on integration of mutation information and other types of data.
Collapse
Affiliation(s)
- Yingxin Kan
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Limin Jiang
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China.,Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jijun Tang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,School of Computational Science and Engineering, University of South Carolina, Columbia, U.S
| | - Yan Guo
- Comprehensive cancer center, Department of Internal Medicine, University of New Mexico, Albuquerque, U.S
| | - Fei Guo
- School of Computer Science and Engineering, Central South University, Changsha, China
| |
Collapse
|
33
|
Zhang T, Zhang SW, Li Y. Identifying Driver Genes for Individual Patients through Inductive Matrix Completion. Bioinformatics 2021; 37:4477-4484. [PMID: 34175939 DOI: 10.1093/bioinformatics/btab477] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/30/2021] [Accepted: 06/25/2021] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION The driver genes play a key role in the evolutionary process of cancer. Effectively identifying these driver genes is crucial to cancer diagnosis and treatment. However, due to the high heterogeneity of cancers, it remains challenging to identify the driver genes for individual patients. Although some computational methods have been proposed to tackle this problem, they seldom consider the fact that the genes functionally similar to the well-established driver genes may likely play similar roles in cancer process, which potentially promotes the driver gene identification. Thus, here we developed a novel approach of IMCDriver to promote the driver gene identification both for cohorts and individual patients. RESULTS IMCDriver first considers the well-established driver genes as prior information, and adopts the using multi-omics data (e.g., somatic mutation, gene expression and protein-protein interaction) to compute the similarity between patients/genes. Then, IMCDriver prioritizes the personalized mutated genes according to their functional similarity to the well-established driver genes via Inductive Matrix Completion. Finally, IMCDriver identifies the highly rank-ordered genes as the personalized driver genes. The results on five cancer datasets from TCGA show that our IMCDriver outperforms other existing state-of-the-art methods both in the cohort and patient-specific driver gene identification. IMCDriver also reveals some novel driver genes that potentially drive cancer development. In addition, even for the driver genes rarely mutated among a population, IMCDriver can still identify them and prioritize them with high priorities. AVAILABILITY Code available at https://github.com/NWPU-903PR/IMCDriver. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tong Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, China Xi'an.,School of Electrical and Mechanical Engineering, Pingdingshan University, Pingdingshan, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, China Xi'an
| | - Yan Li
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, China Xi'an
| |
Collapse
|
34
|
Guo WF, Yu X, Shi QQ, Liang J, Zhang SW, Zeng T. Performance assessment of sample-specific network control methods for bulk and single-cell biological data analysis. PLoS Comput Biol 2021; 17:e1008962. [PMID: 33956788 PMCID: PMC8130943 DOI: 10.1371/journal.pcbi.1008962] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/18/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022] Open
Abstract
In the past few years, a wealth of sample-specific network construction methods and structural network control methods has been proposed to identify sample-specific driver nodes for supporting the Sample-Specific network Control (SSC) analysis of biological networked systems. However, there is no comprehensive evaluation for these state-of-the-art methods. Here, we conducted a performance assessment for 16 SSC analysis workflows by using the combination of 4 sample-specific network reconstruction methods and 4 representative structural control methods. This study includes simulation evaluation of representative biological networks, personalized driver genes prioritization on multiple cancer bulk expression datasets with matched patient samples from TCGA, and cell marker genes and key time point identification related to cell differentiation on single-cell RNA-seq datasets. By widely comparing analysis of existing SSC analysis workflows, we provided the following recommendations and banchmarking workflows. (i) The performance of a network control method is strongly dependent on the up-stream sample-specific network method, and Cell-Specific Network construction (CSN) method and Single-Sample Network (SSN) method are the preferred sample-specific network construction methods. (ii) After constructing the sample-specific networks, the undirected network-based control methods are more effective than the directed network-based control methods. In addition, these data and evaluation pipeline are freely available on https://github.com/WilfongGuo/Benchmark_control.
Collapse
Affiliation(s)
- Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Qian-Qian Shi
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Jing Liang
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
| | - Tao Zeng
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
35
|
Pham VVH, Liu L, Bracken CP, Nguyen T, Goodall GJ, Li J, Le TD. pDriver : A novel method for unravelling personalised coding and miRNA cancer drivers. Bioinformatics 2021; 37:3285-3292. [PMID: 33904576 DOI: 10.1093/bioinformatics/btab262] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
MOTIVATION Unravelling cancer driver genes is important in cancer research. Although computational methods have been developed to identify cancer drivers, most of them detect cancer drivers at population level. However, two patients who have the same cancer type and receive the same treatment may have different outcomes because each patient has a different genome and their disease might be driven by different driver genes. Therefore new methods are being developed for discovering cancer drivers at individual level, but existing personalised methods only focus on coding drivers while microRNAs (miRNAs) have been shown to drive cancer progression as well. Thus, novel methods are required to discover both coding and miRNA cancer drivers at individual level. RESULTS We propose the novel method, pDriver, to discover personalised cancer drivers. pDriver includes two stages: (1) Constructing gene networks for each cancer patient and (2) Discovering cancer drivers for each patient based on the constructed gene networks. To demonstrate the effectiveness of pDriver, we have applied it to five TCGA cancer datasets and compared it with the state-of-the-art methods. The result indicates that pDriver is more effective than other methods. Furthermore, pDriver can also detect miRNA cancer drivers and most of them have been confirmed to be associated with cancer by literature. We further analyse the predicted personalised drivers for breast cancer patients and the result shows that they are significantly enriched in many GO processes and KEGG pathways involved in breast cancer. AVAILABILITY AND IMPLEMENTATION pDriver is available at https://github.com/pvvhoang/pDriver. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Vu V H Pham
- UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Lin Liu
- UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Cameron P Bracken
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA 5000, Australia.,Department of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Thin Nguyen
- Applied Artificial Intelligence Institute, Deakin University, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, an alliance of SA Pathology and University of South Australia, Adelaide, SA 5000, Australia.,Department of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Jiuyong Li
- UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Thuc D Le
- UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| |
Collapse
|
36
|
Guo WF, Zhang SW, Feng YH, Liang J, Zeng T, Chen L. Network controllability-based algorithm to target personalized driver genes for discovering combinatorial drugs of individual patients. Nucleic Acids Res 2021; 49:e37. [PMID: 33434272 PMCID: PMC8053130 DOI: 10.1093/nar/gkaa1272] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/02/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Multiple driver genes in individual patient samples may cause resistance to individual drugs in precision medicine. However, current computational methods have not studied how to fill the gap between personalized driver gene identification and combinatorial drug discovery for individual patients. Here, we developed a novel structural network controllability-based personalized driver genes and combinatorial drug identification algorithm (CPGD), aiming to identify combinatorial drugs for an individual patient by targeting personalized driver genes from network controllability perspective. On two benchmark disease datasets (i.e. breast cancer and lung cancer datasets), performance of CPGD is superior to that of other state-of-the-art driver gene-focus methods in terms of discovery rate among prior-known clinical efficacious combinatorial drugs. Especially on breast cancer dataset, CPGD evaluated synergistic effect of pairwise drug combinations by measuring synergistic effect of their corresponding personalized driver gene modules, which are affected by a given targeting personalized driver gene set of drugs. The results showed that CPGD performs better than existing synergistic combinatorial strategies in identifying clinical efficacious paired combinatorial drugs. Furthermore, CPGD enhanced cancer subtyping by computationally providing personalized side effect signatures for individual patients. In addition, CPGD identified 90 drug combinations candidates from SARS-COV2 dataset as potential drug repurposing candidates for recently spreading COVID-19.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian 710072, China.,School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian 710072, China
| | - Yue-Hua Feng
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian 710072, China
| | - Jing Liang
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Tao Zeng
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy Science, Shanghai 200031, China
| | - Luonan Chen
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy Science, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.,Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
37
|
Zhao W, Yang J, Wu J, Cai G, Zhang Y, Haltom J, Su W, Dong MJ, Chen S, Wu J, Zhou Z, Gu X. CanDriS: posterior profiling of cancer-driving sites based on two-component evolutionary model. Brief Bioinform 2021; 22:6238585. [PMID: 33876217 DOI: 10.1093/bib/bbab131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Current cancer genomics databases have accumulated millions of somatic mutations that remain to be further explored. Due to the over-excess mutations unrelated to cancer, the great challenge is to identify somatic mutations that are cancer-driven. Under the notion that carcinogenesis is a form of somatic-cell evolution, we developed a two-component mixture model: while the ground component corresponds to passenger mutations, the rapidly evolving component corresponds to driver mutations. Then, we implemented an empirical Bayesian procedure to calculate the posterior probability of a site being cancer-driven. Based on these, we developed a software CanDriS (Cancer Driver Sites) to profile the potential cancer-driving sites for thousands of tumor samples from the Cancer Genome Atlas and International Cancer Genome Consortium across tumor types and pan-cancer level. As a result, we identified that approximately 1% of the sites have posterior probabilities larger than 0.90 and listed potential cancer-wide and cancer-specific driver mutations. By comprehensively profiling all potential cancer-driving sites, CanDriS greatly enhances our ability to refine our knowledge of the genetic basis of cancer and might guide clinical medication in the upcoming era of precision medicine. The results were displayed in a database CandrisDB (http://biopharm.zju.edu.cn/candrisdb/).
Collapse
Affiliation(s)
- Wenyi Zhao
- College of Pharmaceutical Sciences & College of Computer Science and Technology, Zhejiang University, China
| | - Jingwen Yang
- MOE Key Laboratory of Contemporary Anthropology, Human Phenome Institute, School of Life Sciences, Fudan University, China
| | - Jingcheng Wu
- College of Pharmaceutical Sciences, Zhejiang University, China
| | - Guoxing Cai
- College of Pharmaceutical Sciences, Zhejiang University, China
| | - Yao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, China
| | - Jeffrey Haltom
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, 12 Iowa 50011, USA
| | - Weijia Su
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, 12 Iowa 50011, USA
| | - Michael J Dong
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, 12 Iowa 50011, USA
| | - Shuqing Chen
- College of Pharmaceutical Sciences, Zhejiang University, China
| | - Jian Wu
- College of Computer Science and Technology & School of Medicine, Zhejiang University, China
| | - Zhan Zhou
- College of Pharmaceutical Sciences, Innovation Institute for Artificial Intelligence in Medicine, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, China
| | - Xun Gu
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, 12 Iowa 50011, USA
| |
Collapse
|
38
|
Pham VVH, Liu L, Bracken C, Goodall G, Li J, Le TD. Computational methods for cancer driver discovery: A survey. Am J Cancer Res 2021; 11:5553-5568. [PMID: 33859763 PMCID: PMC8039954 DOI: 10.7150/thno.52670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/20/2021] [Indexed: 12/21/2022] Open
Abstract
Identifying the genes responsible for driving cancer is of critical importance for directing treatment. Accordingly, multiple computational tools have been developed to facilitate this task. Due to the different methods employed by these tools, different data considered by the tools, and the rapidly evolving nature of the field, the selection of an appropriate tool for cancer driver discovery is not straightforward. This survey seeks to provide a comprehensive review of the different computational methods for discovering cancer drivers. We categorise the methods into three groups; methods for single driver identification, methods for driver module identification, and methods for identifying personalised cancer drivers. In addition to providing a “one-stop” reference of these methods, by evaluating and comparing their performance, we also provide readers the information about the different capabilities of the methods in identifying biologically significant cancer drivers. The biologically relevant information identified by these tools can be seen through the enrichment of discovered cancer drivers in GO biological processes and KEGG pathways and through our identification of a small cancer-driver cohort that is capable of stratifying patient survival.
Collapse
|
39
|
Abstract
Understanding the genetic mechanisms underlying particular adaptations/phenotypes of organisms is one of the core issues of evolutionary biology. The use of genomic data has greatly advanced our understandings on this issue, as well as other aspects of evolutionary biology, including molecular adaptation, speciation, and even conservation of endangered species. Despite the well-recognized advantages, usages of genomic data are still limited to non-mammal vertebrate groups, partly due to the difficulties in assembling large or highly heterozygous genomes. Although this is particularly the case for amphibians, nonetheless, several comparative and population genomic analyses have shed lights into the speciation and adaptation processes of amphibians in a complex landscape, giving a promising hope for a wider application of genomics in the previously believed challenging groups of organisms. At the same time, these pioneer studies also allow us to realize numerous challenges in studying the molecular adaptations and/or phenotypic evolutionary mechanisms of amphibians. In this review, we first summarize the recent progresses in the study of adaptive evolution of amphibians based on genomic data, and then we give perspectives regarding how to effectively identify key pathways underlying the evolution of complex traits in the genomic era, as well as directions for future research.
Collapse
Affiliation(s)
- Yan-Bo Sun
- Laboratory of Ecology and Evolutionary Biology, Yunnan University, Kunming, Yunnan 650091, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| | - Yi Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Kai Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Sam Noble Oklahoma Museum of Natural History and Department of Biology, University of Oklahoma, Norman, Oklahoma 73072, USA
| |
Collapse
|
40
|
Liu X, Maiorino E, Halu A, Glass K, Prasad RB, Loscalzo J, Gao J, Sharma A. Robustness and lethality in multilayer biological molecular networks. Nat Commun 2020; 11:6043. [PMID: 33247151 PMCID: PMC7699651 DOI: 10.1038/s41467-020-19841-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 10/26/2020] [Indexed: 12/27/2022] Open
Abstract
Robustness is a prominent feature of most biological systems. Most previous related studies have been focused on homogeneous molecular networks. Here we propose a comprehensive framework for understanding how the interactions between genes, proteins and metabolites contribute to the determinants of robustness in a heterogeneous biological network. We integrate heterogeneous sources of data to construct a multilayer interaction network composed of a gene regulatory layer, a protein-protein interaction layer, and a metabolic layer. We design a simulated perturbation process to characterize the contribution of each gene to the overall system's robustness, and find that influential genes are enriched in essential and cancer genes. We show that the proposed mechanism predicts a higher vulnerability of the metabolic layer to perturbations applied to genes associated with metabolic diseases. Furthermore, we find that the real network is comparably or more robust than expected in multiple random realizations. Finally, we analytically derive the expected robustness of multilayer biological networks starting from the degree distributions within and between layers. These results provide insights into the non-trivial dynamics occurring in the cell after a genetic perturbation is applied, confirming the importance of including the coupling between different layers of interaction in models of complex biological systems.
Collapse
Affiliation(s)
- Xueming Liu
- Key Laboratory of Imaging Processing and Intelligent Control, School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Enrico Maiorino
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Rashmi B Prasad
- Genomics Diabetes and Endocrinology, Lund University Diabetes Centre, CRC, Malmö, SE, 20502, Sweden
| | - Joseph Loscalzo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jianxi Gao
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA.
| | - Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Gu H, Xu X, Qin P, Wang J. FI-Net: Identification of Cancer Driver Genes by Using Functional Impact Prediction Neural Network. Front Genet 2020; 11:564839. [PMID: 33244318 PMCID: PMC7683798 DOI: 10.3389/fgene.2020.564839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022] Open
Abstract
Identification of driver genes, whose mutations cause the development of tumors, is crucial for the improvement of cancer research and precision medicine. To overcome the problem that the traditional frequency-based methods cannot detect lowly recurrently mutated driver genes, researchers have focused on the functional impact of gene mutations and proposed the function-based methods. However, most of the function-based methods estimate the distribution of the null model through the non-parametric method, which is sensitive to sample size. Besides, such methods could probably lead to underselection or overselection results. In this study, we proposed a method to identify driver genes by using functional impact prediction neural network (FI-net). An artificial neural network as a parametric model was constructed to estimate the functional impact scores for genes, in which multi-omics features were used as the multivariate inputs. Then the estimation of the background distribution and the identification of driver genes were conducted in each cluster obtained by the hierarchical clustering algorithm. We applied FI-net and other 22 state-of-the-art methods to 31 datasets from The Cancer Genome Atlas project. According to the comprehensive evaluation criterion, FI-net was powerful among various datasets and outperformed the other methods in terms of the overlap fraction with Cancer Gene Census and Network of Cancer Genes database, and the consensus in predictions among methods. Furthermore, the results illustrated that FI-net can identify known and potential novel driver genes.
Collapse
Affiliation(s)
- Hong Gu
- Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Xiaolu Xu
- Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Pan Qin
- Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Jia Wang
- Department of Breast Surgery, Institute of Breast Disease, Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
42
|
Wei PJ, Wu FX, Xia J, Su Y, Wang J, Zheng CH. Prioritizing Cancer Genes Based on an Improved Random Walk Method. Front Genet 2020; 11:377. [PMID: 32411180 PMCID: PMC7198854 DOI: 10.3389/fgene.2020.00377] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/26/2020] [Indexed: 12/18/2022] Open
Abstract
Identifying driver genes that contribute to cancer progression from numerous passenger genes, although a central goal, is a major challenge. The protein-protein interaction network provides convenient and reasonable assistance for driver gene discovery. Random walk-based methods have been widely used to prioritize nodes in social or biological networks. However, most studies select the next arriving node uniformly from the random walker's neighbors. Few consider transiting preference according to the degree of random walker's neighbors. In this study, based on the random walk method, we propose a novel approach named Driver_IRW (Driver genes discovery with Improved Random Walk method), to prioritize cancer genes in cancer-related network. The key idea of Driver_IRW is to assign different transition probabilities for different edges of a constructed cancer-related network in accordance with the degree of the nodes' neighbors. Furthermore, the global centrality (here is betweenness centrality) and Katz feedback centrality are incorporated into the framework to evaluate the probability to walk to the seed nodes. Experimental results on four cancer types indicate that Driver_IRW performs more efficiently than some previously published methods for uncovering known cancer-related genes. In conclusion, our method can aid in prioritizing cancer-related genes and complement traditional frequency and network-based methods.
Collapse
Affiliation(s)
- Pi-Jing Wei
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| | - Fang-Xiang Wu
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Computer Sciences, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Junfeng Xia
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Yansen Su
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| | - Jing Wang
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
- College of Computer and Information Engineering, Fuyang Normal University, Fuyang, China
| | - Chun-Hou Zheng
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, College of Computer Science and Technology, Anhui University, Hefei, China
| |
Collapse
|