1
|
Wang Z, Benhammouda H, Chen B. Identifying Cancer Stage-Related Biomarkers for Lung Adenocarcinoma by Integrating Both Node and Edge Features. Genes (Basel) 2025; 16:261. [PMID: 40149413 PMCID: PMC11941903 DOI: 10.3390/genes16030261] [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: 01/31/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
Background: In order to characterize phenotypes and diseases, genetic factors and their interactions in biological systems must be considered. Although genes or node features are the core units of genetic information, their connections, also known as edge features, are composed of a network of gene interactions. These components are crucial for understanding the molecular basis of disease and phenotype development. Existing research typically utilizes node biomarkers composed of individual genes or proteins for the binary classification of cancer. However, due to significant heterogeneity among patients, these methods cannot adapt to the subtle changes required for precise cancer staging, and relying solely on node biomarkers often leads to poor accuracy in classifying cancer staging. Methods: In this study, a computational framework was developed to diagnose lung adenocarcinoma, integrating node and edge features such as correlation, covariance, and residuals. The proposed method allows for precise diagnosis in the case of a single sample, which can identify the minimum feature set that effectively distinguishes cancer staging. Results: The advantages of the proposed method are: (i) it can diagnose each individual test sample, promoting personalized treatment; (ii) integrating node and edge features can improve diagnostic accuracy, indicating that each type of feature can capture unique aspects of the disease; (iii) it significantly reduces the number of features required to accurately classify the four stages of cancer, thereby achieving optimal cross-validation accuracy. Conclusions: This streamlined and effective feature set highlights the potential of our approach in advancing personalized medicine and improving clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Zige Wang
- School of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China;
| | - Hamza Benhammouda
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Bolin Chen
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China;
| |
Collapse
|
2
|
Saha E, Fanfani V, Mandros P, Ben Guebila M, Fischer J, Shutta KH, DeMeo DL, Lopes-Ramos CM, Quackenbush J. Bayesian inference of sample-specific coexpression networks. Genome Res 2024; 34:1397-1410. [PMID: 39134413 PMCID: PMC11529861 DOI: 10.1101/gr.279117.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024]
Abstract
Gene regulatory networks (GRNs) are effective tools for inferring complex interactions between molecules that regulate biological processes and hence can provide insights into drivers of biological systems. Inferring coexpression networks is a critical element of GRN inference, as the correlation between expression patterns may indicate that genes are coregulated by common factors. However, methods that estimate coexpression networks generally derive an aggregate network representing the mean regulatory properties of the population and so fail to fully capture population heterogeneity. Bayesian optimized networks obtained by assimilating omic data (BONOBO) is a scalable Bayesian model for deriving individual sample-specific coexpression matrices that recognizes variations in molecular interactions across individuals. For each sample, BONOBO assumes a Gaussian distribution on the log-transformed centered gene expression and a conjugate prior distribution on the sample-specific coexpression matrix constructed from all other samples in the data. Combining the sample-specific gene coexpression with the prior distribution, BONOBO yields a closed-form solution for the posterior distribution of the sample-specific coexpression matrices, thus allowing the analysis of large data sets. We demonstrate BONOBO's utility in several contexts, including analyzing gene regulation in yeast transcription factor knockout studies, the prognostic significance of miRNA-mRNA interaction in human breast cancer subtypes, and sex differences in gene regulation within human thyroid tissue. We find that BONOBO outperforms other methods that have been used for sample-specific coexpression network inference and provides insight into individual differences in the drivers of biological processes.
Collapse
Affiliation(s)
- Enakshi Saha
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Viola Fanfani
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Panagiotis Mandros
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Jonas Fischer
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Katherine H Shutta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Camila M Lopes-Ramos
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA;
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
3
|
Zhang J, Liu L, Wei X, Zhao C, Luo Y, Li J, Le TD. Scanning sample-specific miRNA regulation from bulk and single-cell RNA-sequencing data. BMC Biol 2024; 22:218. [PMID: 39334271 PMCID: PMC11438147 DOI: 10.1186/s12915-024-02020-x] [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: 01/15/2024] [Accepted: 09/24/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND RNA-sequencing technology provides an effective tool for understanding miRNA regulation in complex human diseases, including cancers. A large number of computational methods have been developed to make use of bulk and single-cell RNA-sequencing data to identify miRNA regulations at the resolution of multiple samples (i.e. group of cells or tissues). However, due to the heterogeneity of individual samples, there is a strong need to infer miRNA regulation specific to individual samples to uncover miRNA regulation at the single-sample resolution level. RESULTS Here, we develop a framework, Scan, for scanning sample-specific miRNA regulation. Since a single network inference method or strategy cannot perform well for all types of new data, Scan incorporates 27 network inference methods and two strategies to infer tissue-specific or cell-specific miRNA regulation from bulk or single-cell RNA-sequencing data. Results on bulk and single-cell RNA-sequencing data demonstrate the effectiveness of Scan in inferring sample-specific miRNA regulation. Moreover, we have found that incorporating the prior information of miRNA targets can generally improve the accuracy of miRNA target prediction. In addition, Scan can contribute to construct cell/tissue correlation networks and recover aggregate miRNA regulatory networks. Finally, the comparison results have shown that the performance of network inference methods is likely to be data-specific, and selecting optimal network inference methods is required for more accurate prediction of miRNA targets. CONCLUSIONS Scan provides a useful method to help infer sample-specific miRNA regulation for new data, benchmark new network inference methods and deepen the understanding of miRNA regulation at the resolution of individual samples.
Collapse
Affiliation(s)
- Junpeng Zhang
- School of Engineering, Dali University, Dali, 671003, Yunnan, China.
| | - Lin Liu
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Xuemei Wei
- School of Engineering, Dali University, Dali, 671003, Yunnan, China
| | - Chunwen Zhao
- School of Engineering, Dali University, Dali, 671003, Yunnan, China
| | - Yanbi Luo
- School of Engineering, Dali University, Dali, 671003, Yunnan, China
| | - Jiuyong Li
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Thuc Duy Le
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia.
| |
Collapse
|
4
|
Saha E, Fanfani V, Mandros P, Ben-Guebila M, Fischer J, Hoff-Shutta K, Glass K, DeMeo DL, Lopes-Ramos C, Quackenbush J. Bayesian Optimized sample-specific Networks Obtained By Omics data (BONOBO). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567119. [PMID: 38014256 PMCID: PMC10680741 DOI: 10.1101/2023.11.16.567119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Gene regulatory networks (GRNs) are effective tools for inferring complex interactions between molecules that regulate biological processes and hence can provide insights into drivers of biological systems. Inferring co-expression networks is a critical element of GRN inference as the correlation between expression patterns may indicate that genes are coregulated by common factors. However, methods that estimate co-expression networks generally derive an aggregate network representing the mean regulatory properties of the population and so fail to fully capture population heterogeneity. To address these concerns, we introduce BONOBO (Bayesian Optimized Networks Obtained By assimilating Omics data), a scalable Bayesian model for deriving individual sample-specific co-expression networks by recognizing variations in molecular interactions across individuals. For every sample, BONOBO assumes a Gaussian distribution on the log-transformed centered gene expression and a conjugate prior distribution on the sample-specific co-expression matrix constructed from all other samples in the data. Combining the sample-specific gene expression with the prior distribution, BONOBO yields a closed-form solution for the posterior distribution of the sample-specific co-expression matrices, thus making the method extremely scalable. We demonstrate the utility of BONOBO in several contexts, including analyzing gene regulation in yeast transcription factor knockout studies, prognostic significance of miRNA-mRNA interaction in human breast cancer subtypes, and sex differences in gene regulation within human thyroid tissue. We find that BONOBO outperforms other sample-specific co-expression network inference methods and provides insight into individual differences in the drivers of biological processes.
Collapse
Affiliation(s)
- Enakshi Saha
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Viola Fanfani
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Panagiotis Mandros
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Marouen Ben-Guebila
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jonas Fischer
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Katherine Hoff-Shutta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kimberly Glass
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Dawn Lisa DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Camila Lopes-Ramos
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
5
|
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
|
6
|
Azim R, Wang S, Dipu SA, Islam N, Ala Muid MR, Elahe MF. A patient-specific functional module and path identification technique from RNA-seq data. Comput Biol Med 2023; 158:106871. [PMID: 37030265 DOI: 10.1016/j.compbiomed.2023.106871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/12/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
With the advancement of new technologies, a huge amount of high dimensional data is being generated which is opening new opportunities and challenges to the study of cancer and diseases. In particular, distinguishing the patient-specific key components and modules which drive tumorigenesis is necessary to analyze. A complex disease generally does not initiate from the dysregulation of a single component but it is the result of the dysfunction of a group of components and networks which differs from patient to patient. However, a patient-specific network is required to understand the disease and its molecular mechanism. We address this requirement by constructing a patient-specific network by sample-specific network theory with integrating cancer-specific differentially expressed genes and elite genes. By elucidating patient-specific networks, it can identify the regulatory modules, driver genes as well as personalized disease networks which can lead to personalized drug design. This method can provide insight into how genes are associating with each other and characterized the patient-specific disease subtypes. The results show that this method can be beneficial for the detection of patient-specific differential modules and interaction between genes. Extensive analysis using existing literature, gene enrichment and survival analysis for three cancer types STAD, PAAD and LUAD shows the effectiveness of this method over other existing methods. In addition, this method can be useful for personalized therapeutics and drug design. This methodology is implemented in the R language and is available at https://github.com/riasatazim/PatientSpecificRNANetwork.
Collapse
|
7
|
Jiang F, Yang L, Jiao X. Dynamic network biomarker to determine the critical point of breast cancer stage progression. Breast Cancer 2023; 30:453-465. [PMID: 36807044 DOI: 10.1007/s12282-023-01438-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 02/11/2023] [Indexed: 02/21/2023]
Abstract
BACKGROUND The discovery of early warning signs and biomarkers in patients with early breast cancer is crucial for the prevention and treatment of breast cancer. Dynamic Network Biomarker (DNB) is an approach based on nonlinear dynamics theory, which we exploited to identify a set of DNB members and their key genes as early warning signals during breast cancer staging progression. METHODS First, based on the gene expression profile of breast cancer in the TCGA database, the DNB algorithm was used to calculate the composite index (CI) of each gene cluster in the process of breast cancer anatomical staging. Then we calculated gene modules associated with the clinical phenotype stage based on weighted gene co-expression network analysis (WGCNA), combined with DNB membership to identify key genes in the network. RESULTS We identified a set of gene clusters with the highest CI in Stage II as DNBs, whose roles in related pathways indicate the emergence of a tipping point and impact on breast cancer development. In addition, analysis of the key gene GPRIN1 showed that high expression of GPRIN1 predicts poor prognosis, and related immune analysis showed that GPRIN1 is involved in the development of breast cancer through immune aspects. CONCLUSION The discovery of DNBs and the key gene GPRIN1 can provide potential biomarkers and therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Fa Jiang
- College of Biomedical Engineering, Taiyuan University of Technology, Jinzhong, 030600, China
| | - Lifeng Yang
- College of Information and Computer, Taiyuan University of Technology, Jinzhong, 030600, China
| | - Xiong Jiao
- College of Biomedical Engineering, Taiyuan University of Technology, Jinzhong, 030600, China.
| |
Collapse
|
8
|
Tang S, Yuan K, Chen L. Molecular biomarkers, network biomarkers, and dynamic network biomarkers for diagnosis and prediction of rare diseases. FUNDAMENTAL RESEARCH 2022; 2:894-902. [PMID: 38933388 PMCID: PMC11197705 DOI: 10.1016/j.fmre.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
The difficulty of converting scientific research findings into novel pharmacological treatments for rare and life-threatening diseases is enormous. Biomarkers related to multiple biological processes involved in cell growth, proliferation, and disease occurrence have been identified in recent years with the development of immunology, molecular biology, and genomics technologies. Biomarkers are capable of reflecting normal physiological processes, pathological processes, and the response to therapeutic intervention; as such, they play vital roles in disease diagnosis, prevention, drug response, and other aspects of biomedicine. The discovery of valuable biomarkers has become a focal point of current research. Numerous studies have identified molecular biomarkers based on the differential expression/concentration of molecules (e.g., genes/proteins) for disease state diagnosis, characterization, and treatment. Although technological breakthroughs in molecular analysis platforms have enabled the identification of a large number of candidate biomarkers for rare diseases, only a small number of these candidates have been properly validated for use in patient treatment. The traditional molecular biomarkers may lose vital information by ignoring molecular associations/interactions, and thus the concept of network biomarkers based on differential associations/correlations of molecule pairs has been established. This approach promises to be more stable and reliable in diagnosing disease states. Furthermore, the newly-emerged dynamic network biomarkers (DNBs) based on differential fluctuations/correlations of molecular groups are able to recognize pre-disease states or critical states instead of disease states, thereby achieving rare disease prediction or predictive/preventative medicine and providing deep insight into the dynamic characteristics of disease initiation and progression.
Collapse
Affiliation(s)
- Shijie Tang
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Yuan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
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
|
10
|
Zhang Y, Chang X, Xia J, Huang Y, Sun S, Chen L, Liu X. Identifying network biomarkers of cancer by sample-specific differential network. BMC Bioinformatics 2022; 23:230. [PMID: 35705908 PMCID: PMC9202129 DOI: 10.1186/s12859-022-04772-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 06/02/2022] [Indexed: 02/08/2023] Open
Abstract
Abundant datasets generated from various big science projects on diseases have presented great challenges and opportunities, which contributed to unfolding the complexity of diseases. The discovery of disease-associated molecular networks for each individual plays an important role in personalized therapy and precision treatment of cancer-based on the reference networks. However, there are no effective ways to distinguish the consistency of different reference networks. In this study, we developed a statistical method, i.e. a sample-specific differential network (SSDN), to construct and analyze such networks based on gene expression of a single sample against a reference dataset. We proved that the SSDN is structurally consistent even with different reference datasets if the reference dataset can follow certain conditions. The SSDN also can be used to identify patient-specific disease modules or network biomarkers as well as predict the potential driver genes of a tumor sample.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.,Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou, 310024, China.,School of Mathematics and Statistics, Shandong University, Weihai, 264209, Shandong, China
| | - Xiao Chang
- Institute of Statistics and Applied Mathematics, Anhui University of Finance & Economics, Bengbu, 233030, China.
| | - Jie Xia
- Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Science, Shanghai, 200031, China
| | - Yanhong Huang
- School of Mathematics and Statistics, Shandong University, Weihai, 264209, Shandong, China
| | - Shaoyan Sun
- School of Mathematics and Statistics, Ludong University, Yantai, 264025, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China. .,Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou, 310024, China. .,Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Science, Shanghai, 200031, China. .,West China Biomedical Big Data Center, Med-X center for informatics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiaoping Liu
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China. .,Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou, 310024, China. .,School of Mathematics and Statistics, Shandong University, Weihai, 264209, Shandong, China.
| |
Collapse
|
11
|
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
|
12
|
Sun P, Wu Y, Yin C, Jiang H, Xu Y, Sun H. Molecular Subtyping of Cancer Based on Distinguishing Co-Expression Modules and Machine Learning. Front Genet 2022; 13:866005. [PMID: 35586568 PMCID: PMC9108363 DOI: 10.3389/fgene.2022.866005] [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: 01/30/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Molecular subtyping of cancer is recognized as a critical and challenging step towards individualized therapy. Most existing computational methods solve this problem via multi-classification of gene-expressions of cancer samples. Although these methods, especially deep learning, perform well in data classification, they usually require large amounts of data for model training and have limitations in interpretability. Besides, as cancer is a complex systemic disease, the phenotypic difference between cancer samples can hardly be fully understood by only analyzing single molecules, and differential expression-based molecular subtyping methods are reportedly not conserved. To address the above issues, we present here a new framework for molecular subtyping of cancer through identifying a robust specific co-expression module for each subtype of cancer, generating network features for each sample by perturbing correlation levels of specific edges, and then training a deep neural network for multi-class classification. When applied to breast cancer (BRCA) and stomach adenocarcinoma (STAD) molecular subtyping, it has superior classification performance over existing methods. In addition to improving classification performance, we consider the specific co-expressed modules selected for subtyping to be biologically meaningful, which potentially offers new insight for diagnostic biomarker design, mechanistic studies of cancer, and individualized treatment plan selection.
Collapse
Affiliation(s)
- Peishuo Sun
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Ying Wu
- Phase I Clinical Trails Center, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chaoyi Yin
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Hongyang Jiang
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Ying Xu
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics University of Georgia, Athens, GA, United States
- *Correspondence: Huiyan Sun, ; Ying Xu,
| | - Huiyan Sun
- School of Artificial Intelligence, Jilin University, Changchun, China
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Huiyan Sun, ; Ying Xu,
| |
Collapse
|
13
|
Yuan K, Zeng T, Chen L. Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study. Front Cell Dev Biol 2022; 9:720321. [PMID: 35155440 PMCID: PMC8826544 DOI: 10.3389/fcell.2021.720321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022] Open
Abstract
An enormous challenge in the post-genome era is to annotate and resolve the consequences of genetic variation on diverse phenotypes. The genome-wide association study (GWAS) is a well-known method to identify potential genetic loci for complex traits from huge genetic variations, following which it is crucial to identify expression quantitative trait loci (eQTL). However, the conventional eQTL methods usually disregard the systematical role of single-nucleotide polymorphisms (SNPs) or genes, thereby overlooking many network-associated phenotypic determinates. Such a problem motivates us to recognize the network-based quantitative trait loci (QTL), i.e., network QTL (nQTL), which is to detect the cascade association as genotype → network → phenotype rather than conventional genotype → expression → phenotype in eQTL. Specifically, we develop the nQTL framework on the theory and approach of single-sample networks, which can identify not only network traits (e.g., the gene subnetwork associated with genotype) for analyzing complex biological processes but also network signatures (e.g., the interactive gene biomarker candidates screened from network traits) for characterizing targeted phenotype and corresponding subtypes. Our results show that the nQTL framework can efficiently capture associations between SNPs and network traits (i.e., edge traits) in various simulated data scenarios, compared with traditional eQTL methods. Furthermore, we have carried out nQTL analysis on diverse biological and biomedical datasets. Our analysis is effective in detecting network traits for various biological problems and can discover many network signatures for discriminating phenotypes, which can help interpret the influence of nQTL on disease subtyping, disease prognosis, drug response, and pathogen factor association. Particularly, in contrast to the conventional approaches, the nQTL framework could also identify many network traits from human bulk expression data, validated by matched single-cell RNA-seq data in an independent or unsupervised manner. All these results strongly support that nQTL and its detection framework can simultaneously explore the global genotype–network–phenotype associations and the underlying network traits or network signatures with functional impact and importance.
Collapse
Affiliation(s)
- Kai Yuan
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Guangzhou Laboratory, Guangzhou, China
- *Correspondence: Tao Zeng, ; Luonan Chen,
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- *Correspondence: Tao Zeng, ; Luonan Chen,
| |
Collapse
|
14
|
Nakazawa MA, Tamada Y, Tanaka Y, Ikeguchi M, Higashihara K, Okuno Y. Novel cancer subtyping method based on patient-specific gene regulatory network. Sci Rep 2021; 11:23653. [PMID: 34880275 PMCID: PMC8654869 DOI: 10.1038/s41598-021-02394-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
The identification of cancer subtypes is important for the understanding of tumor heterogeneity. In recent years, numerous computational methods have been proposed for this problem based on the multi-omics data of patients. It is widely accepted that different cancer subtypes are induced by different molecular regulatory networks. However, only a few incorporate the differences between their molecular systems into the identification processes. In this study, we present a novel method to identify cancer subtypes based on patient-specific molecular systems. Our method realizes this by quantifying patient-specific gene networks, which are estimated from their transcriptome data, and by clustering their quantified networks. Comprehensive analyses of The Cancer Genome Atlas (TCGA) datasets applied to our method confirmed that they were able to identify more clinically meaningful cancer subtypes than the existing subtypes and found that the identified subtypes comprised different molecular features. Our findings also show that the proposed method can identify the novel cancer subtypes even with single omics data, which cannot otherwise be captured by existing methods using multi-omics data.
Collapse
Affiliation(s)
| | - Yoshinori Tamada
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan.
- Innovation Center for Health Promotion, Hirosaki University, Hirosaki, 036-8562, Japan.
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8507, Japan
- Biomedical Computational Intelligence Unit, HPC- and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe, 650-0047, Japan
| | - Marie Ikeguchi
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Kako Higashihara
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan.
- Biomedical Computational Intelligence Unit, HPC- and AI-driven Drug Development Platform Division, RIKEN Center for Computational Science, Kobe, 650-0047, Japan.
| |
Collapse
|
15
|
Guo WF, Zhang SW, Zeng T, Akutsu T, Chen L. Network control principles for identifying personalized driver genes in cancer. Brief Bioinform 2021; 21:1641-1662. [PMID: 31711128 DOI: 10.1093/bib/bbz089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 02/02/2023] Open
Abstract
To understand tumor heterogeneity in cancer, personalized driver genes (PDGs) need to be identified for unraveling the genotype-phenotype associations corresponding to particular patients. However, most of the existing driver-focus methods mainly pay attention on the cohort information rather than on individual information. Recent developing computational approaches based on network control principles are opening a new way to discover driver genes in cancer, particularly at an individual level. To provide comprehensive perspectives of network control methods on this timely topic, we first considered the cancer progression as a network control problem, in which the expected PDGs are altered genes by oncogene activation signals that can change the individual molecular network from one health state to the other disease state. Then, we reviewed the network reconstruction methods on single samples and introduced novel network control methods on single-sample networks to identify PDGs in cancer. Particularly, we gave a performance assessment of the network structure control-based PDGs identification methods on multiple cancer datasets from TCGA, for which the data and evaluation package also are publicly available. Finally, we discussed future directions for the application of network control methods to identify PDGs in cancer and diverse biological processes.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, 611-0011, Japan
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
16
|
Facial Skin Microbiota-Mediated Host Response to Pollution Stress Revealed by Microbiome Networks of Individual. mSystems 2021; 6:e0031921. [PMID: 34313461 PMCID: PMC8407115 DOI: 10.1128/msystems.00319-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Urban living has been reported to cause various skin disorders. As an integral part of the skin barrier, the skin microbiome is among the key factors associated with urbanization-related skin alterations. The role of skin microbiome in mediating the effect of urban stressors (e.g., air pollutants) on skin physiology is not well understood. We generated 16S sequencing data and constructed a microbiome network of individual (MNI) to analyze the effect of pollution stressors on the microbiome network and its downstream mediation effect on skin physiology in a personalized manner. In particular, we found that the connectivity and fragility of MNIs significantly mediated the adverse effects of air pollution on skin health, and a smoking lifestyle deepened the negative effects of pollution stress on facial skin microbiota. This is the first study that describes the mediation effect of the microbiome network on the skin’s physiological response toward environmental factors as revealed by our newly developed MNI approach and conditional process analysis. IMPORTANCE The association between the skin microbiome and skin health has been widely reported. However, the role of the skin microbiome in mediating skin physiology remains a challenging and yet priority subject in the field. Through developing a novel MNI method followed by mediation analysis, we characterized the network signature of the skin microbiome at an individual level and revealed the role of the skin microbiome in mediating the skin’s responses toward environmental stressors. Our findings may shed new light on microbiome functions in skin health and lay the foundation for the design of a microbiome-based intervention strategy in the future.
Collapse
|
17
|
Zhang J, Zhang Y, Kang JY, Chen S, He Y, Han B, Liu MF, Lu L, Li L, Yi Z, Chen L. Potential transmission chains of variant B.1.1.7 and co-mutations of SARS-CoV-2. Cell Discov 2021; 7:44. [PMID: 34127650 PMCID: PMC8203788 DOI: 10.1038/s41421-021-00282-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/15/2021] [Indexed: 02/05/2023] Open
Abstract
The presence of SARS-CoV-2 mutants, including the emerging variant B.1.1.7, has raised great concerns in terms of pathogenesis, transmission, and immune escape. Characterizing SARS-CoV-2 mutations, evolution, and effects on infectivity and pathogenicity is crucial to the design of antibody therapies and surveillance strategies. Here, we analyzed 454,443 SARS-CoV-2 spike genes/proteins and 14,427 whole-genome sequences. We demonstrated that the early variant B.1.1.7 may not have evolved spontaneously in the United Kingdom or within human populations. Our extensive analyses suggested that Canidae, Mustelidae or Felidae, especially the Canidae family (for example, dog) could be a possible host of the direct progenitor of variant B.1.1.7. An alternative hypothesis is that the variant was simply yet to be sampled. Notably, the SARS-CoV-2 whole-genome represents a large number of potential co-mutations. In addition, we used an experimental SARS-CoV-2 reporter replicon system to introduce the dominant co-mutations NSP12_c14408t, 5'UTR_c241t, and NSP3_c3037t into the viral genome, and to monitor the effect of the mutations on viral replication. Our experimental results demonstrated that the co-mutations significantly attenuated the viral replication. The study provides valuable clues for discovering the transmission chains of variant B.1.1.7 and understanding the evolutionary process of SARS-CoV-2.
Collapse
Affiliation(s)
- Jingsong Zhang
- grid.9227.e0000000119573309State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yang Zhang
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun-Yan Kang
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Shanghai, China
| | - Shuiye Chen
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongqun He
- grid.214458.e0000000086837370Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI USA
| | - Benhao Han
- grid.9227.e0000000119573309State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Mo-Fang Liu
- grid.9227.e0000000119573309State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Shanghai, China
| | - Lina Lu
- grid.9227.e0000000119573309State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Li Li
- grid.38142.3c000000041936754XDepartment of Genetics, Harvard Medical School, Boston, MA USA
| | - Zhigang Yi
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Luonan Chen
- grid.9227.e0000000119573309State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China ,grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, China ,grid.410726.60000 0004 1797 8419Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China ,Pazhou Lab, Guangzhou, China
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
c-CSN: Single-cell RNA Sequencing Data Analysis by Conditional Cell-specific Network. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:319-329. [PMID: 33684532 PMCID: PMC8602759 DOI: 10.1016/j.gpb.2020.05.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/13/2020] [Accepted: 07/08/2020] [Indexed: 12/28/2022]
Abstract
The rapid advancement of single-cell technologies has shed new light on the complex mechanisms of cellular heterogeneity. However, compared to bulk RNA sequencing (RNA-seq), single-cell RNA-seq (scRNA-seq) suffers from higher noise and lower coverage, which brings new computational difficulties. Based on statistical independence, cell-specific network (CSN) is able to quantify the overall associations between genes for each cell, yet suffering from a problem of overestimation related to indirect effects. To overcome this problem, we propose the c-CSN method, which can construct the conditional cell-specific network (CCSN) for each cell. c-CSN method can measure the direct associations between genes by eliminating the indirect associations. c-CSN can be used for cell clustering and dimension reduction on a network basis of single cells. Intuitively, each CCSN can be viewed as the transformation from less “reliable” gene expression to more “reliable” gene–gene associations in a cell. Based on CCSN, we further design network flow entropy (NFE) to estimate the differentiation potency of a single cell. A number of scRNA-seq datasets were used to demonstrate the advantages of our approach. 1) One direct association network is generated for one cell. 2) Most existing scRNA-seq methods designed for gene expression matrices are also applicable to c-CSN-transformed degree matrices. 3) CCSN-based NFE helps resolving the direction of differentiation trajectories by quantifying the potency of each cell. c-CSN is publicly available at https://github.com/LinLi-0909/c-CSN.
Collapse
|
21
|
Jahagirdar S, Saccenti E. Evaluation of Single Sample Network Inference Methods for Metabolomics-Based Systems Medicine. J Proteome Res 2020; 20:932-949. [PMID: 33267585 PMCID: PMC7786380 DOI: 10.1021/acs.jproteome.0c00696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Networks
and network analyses are fundamental tools of systems
biology. Networks are built by inferring pair-wise relationships among
biological entities from a large number of samples such that subject-specific
information is lost. The possibility of constructing these sample
(individual)-specific networks from single molecular profiles might
offer new insights in systems and personalized medicine and as a consequence
is attracting more and more research interest. In this study, we evaluated
and compared LIONESS (Linear Interpolation to Obtain Network Estimates
for Single Samples) and ssPCC (single sample network based on Pearson
correlation) in the metabolomics context of metabolite–metabolite
association networks. We illustrated and explored the characteristics
of these two methods on (i) simulated data, (ii) data generated from
a dynamic metabolic model to simulate real-life observed metabolite
concentration profiles, and (iii) 22 metabolomic data sets and (iv)
we applied single sample network inference to a study case pertaining
to the investigation of necrotizing soft tissue infections to show
how these methods can be applied in metabolomics. We also proposed
some adaptations of the methods that can be used for data exploration.
Overall, despite some limitations, we found single sample networks
to be a promising tool for the analysis of metabolomics data.
Collapse
Affiliation(s)
- Sanjeevan Jahagirdar
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
22
|
Identification of Long Noncoding RNA Biomarkers for Hepatocellular Carcinoma Using Single-Sample Networks. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8579651. [PMID: 33299877 PMCID: PMC7700720 DOI: 10.1155/2020/8579651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023]
Abstract
Objective Many studies have found that long noncoding RNAs (lncRNAs) are differentially expressed in hepatocellular carcinoma (HCC) and closely associated with the occurrence and prognosis of HCC. Since patients with HCC are usually diagnosed in late stages, more effective biomarkers for early diagnosis and prognostic prediction are in urgent need. Methods The RNA-seq data of liver hepatocellular carcinoma (LIHC) were downloaded from The Cancer Genome Atlas (TCGA). Differentially expressed lncRNAs and mRNAs were obtained using the edgeR package. The single-sample networks of the 371 tumor samples were constructed to identify the candidate lncRNA biomarkers. Univariate Cox regression analysis was performed to further select the potential lncRNA biomarkers. By multivariate Cox regression analysis, a 3-lncRNA-based risk score model was established on the training set. Then, the survival prediction ability of the 3-lncRNA-based risk score model was evaluated on the testing set and the entire set. Function enrichment analyses were performed using Metascape. Results Three lncRNAs (RP11-150O12.3, RP11-187E13.1, and RP13-143G15.4) were identified as the potential lncRNA biomarkers for LIHC. The 3-lncRNA-based risk model had a good survival prediction ability for the patients with LIHC. Multivariate Cox regression analysis proved that the 3-lncRNA-based risk score was an independent predictor for the survival prediction of patients with LIHC. Function enrichment analysis indicated that the three lncRNAs may be associated with LIHC via their involvement in many known cancer-associated biological functions. Conclusion This study could provide novel insights to identify lncRNA biomarkers for LIHC at a molecular network level.
Collapse
|
23
|
Li C, Sun YD, Yu GY, Cui JR, Lou Z, Zhang H, Huang Y, Bai CG, Deng LL, Liu P, Zheng K, Wang YH, Wang QQ, Li QR, Wu QQ, Liu Q, Shyr Y, Li YX, Chen LN, Wu JR, Zhang W, Zeng R. Integrated Omics of Metastatic Colorectal Cancer. Cancer Cell 2020; 38:734-747.e9. [PMID: 32888432 DOI: 10.1016/j.ccell.2020.08.002] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/22/2020] [Accepted: 08/06/2020] [Indexed: 12/23/2022]
Abstract
We integrate the genomics, proteomics, and phosphoproteomics of 480 clinical tissues from 146 patients in a Chinese colorectal cancer (CRC) cohort, among which 70 had metastatic CRC (mCRC). Proteomic profiling differentiates three CRC subtypes characterized by distinct clinical prognosis and molecular signatures. Proteomic and phosphoproteomic profiling of primary tumors alone successfully distinguishes cases with metastasis. Metastatic tissues exhibit high similarities with primary tumors at the genetic but not the proteomic level, and kinase network analysis reveals significant heterogeneity between primary colorectal tumors and their liver metastases. In vivo xenograft-based drug tests using 31 primary and metastatic tumors show personalized responses, which could also be predicted by kinase-substrate network analysis no matter whether tumors carry mutations in the drug-targeted genes. Our study provides a valuable resource for better understanding of mCRC and has potential for clinical application.
Collapse
Affiliation(s)
- Chen Li
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi-Di Sun
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guan-Yu Yu
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jing-Ru Cui
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zheng Lou
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Hang Zhang
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Ya Huang
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chen-Guang Bai
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lu-Lu Deng
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Peng Liu
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Kuo Zheng
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan-Hua Wang
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qin-Qin Wang
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qing-Run Li
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing-Qing Wu
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qi Liu
- Department of Biostatistics and Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yu Shyr
- Department of Biostatistics and Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yi-Xue Li
- CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luo-Nan Chen
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jia-Rui Wu
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Wei Zhang
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Rong Zeng
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
24
|
Fang J, Pian C, Xu M, Kong L, Li Z, Ji J, Zhang L, Chen Y. Revealing Prognosis-Related Pathways at the Individual Level by a Comprehensive Analysis of Different Cancer Transcription Data. Genes (Basel) 2020; 11:genes11111281. [PMID: 33138076 PMCID: PMC7692404 DOI: 10.3390/genes11111281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Identifying perturbed pathways at an individual level is important to discover the causes of cancer and develop individualized custom therapeutic strategies. Though prognostic gene lists have had success in prognosis prediction, using single genes that are related to the relevant system or specific network cannot fully reveal the process of tumorigenesis. We hypothesize that in individual samples, the disruption of transcription homeostasis can influence the occurrence, development, and metastasis of tumors and has implications for patient survival outcomes. Here, we introduced the individual-level pathway score, which can measure the correlation perturbation of the pathways in a single sample well. We applied this method to the expression data of 16 different cancer types from The Cancer Genome Atlas (TCGA) database. Our results indicate that different cancer types as well as their tumor-adjacent tissues can be clearly distinguished by the individual-level pathway score. Additionally, we found that there was strong heterogeneity among different cancer types and the percentage of perturbed pathways as well as the perturbation proportions of tumor samples in each pathway were significantly different. Finally, the prognosis-related pathways of different cancer types were obtained by survival analysis. We demonstrated that the individual-level pathway score (iPS) is capable of classifying cancer types and identifying some key prognosis-related pathways.
Collapse
Affiliation(s)
- Jingya Fang
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Cong Pian
- Department of Mathematics, College of Science, Nanjing Agricultural University, Nanjing 210095, China;
| | - Mingmin Xu
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Lingpeng Kong
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Zutan Li
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Jinwen Ji
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Liangyun Zhang
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
- Correspondence: (L.Z.); (Y.C.)
| | - Yuanyuan Chen
- Department of Mathematics, College of Science, Nanjing Agricultural University, Nanjing 210095, China;
- Correspondence: (L.Z.); (Y.C.)
| |
Collapse
|
25
|
Sun Y, Li C, Pang S, Yao Q, Chen L, Li Y, Zeng R. Kinase-substrate Edge Biomarkers Provide a More Accurate Prognostic Prediction in ER-negative Breast Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2020; 18:525-538. [PMID: 33450402 PMCID: PMC8377385 DOI: 10.1016/j.gpb.2019.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 08/27/2019] [Accepted: 11/11/2019] [Indexed: 11/19/2022]
Abstract
The estrogen receptor (ER)-negative breast cancer subtype is aggressive with few treatment options available. To identify specific prognostic factors for ER-negative breast cancer, this study included 705,729 and 1034 breast invasive cancer patients from the Surveillance, Epidemiology, and End Results (SEER) and The Cancer Genome Atlas (TCGA) databases, respectively. To identify key differential kinase-substrate node and edge biomarkers between ER-negative and ER-positive breast cancer patients, we adopted a network-based method using correlation coefficients between molecular pairs in the kinase regulatory network. Integrated analysis of the clinical and molecular data revealed the significant prognostic power of kinase-substrate node and edge features for both subtypes of breast cancer. Two promising kinase-substrate edge features, CSNK1A1-NFATC3 and SRC-OCLN, were identified for more accurate prognostic prediction in ER-negative breast cancer patients.
Collapse
Affiliation(s)
- Yidi Sun
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen Li
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shichao Pang
- Deptartment of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qianlan Yao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Luonan Chen
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, Shanghai 201210, China; CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.
| | - Yixue Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Life Sciences, ShanghaiTech University, Shanghai 201210, China; Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai 200032, China; Shanghai Center for Bioinformation Technology, Shanghai Academy of Science & Technology, Shanghai 201203, China.
| | - Rong Zeng
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
26
|
Su Y, Su X, Wang Q, Zhang L. A multi-objective optimization method for identification of module biomarkers for disease diagnosis. Methods 2020; 192:35-45. [PMID: 32949693 DOI: 10.1016/j.ymeth.2020.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/03/2020] [Accepted: 09/07/2020] [Indexed: 01/14/2023] Open
Abstract
Biomarker identification aims at finding a set of biological indicators that best discriminate biological samples of different phenotypes. In this paper, we take the module containing the significant disease-related genes and their interactions from biological networks as a module biomarker, and propose an evolutionary multi-objective optimization method to identify module biomarkers for disease diagnosis. To be specific, we take the classification accuracy on control and disease samples, the association with disease and the intra-link density in the module as the optimization objectives. To achieve the best performance, a novel population initiation strategy is tailored to generate dense-connected initial solutions, and a specific population update strategy is employed to direct the evolution towards the global optimums with abundant diversity. Experimental results show that our method outperforms the previous state-of-the-art disease diagnosis methods. Meantime, the detected biomarker module can reflect the basic and significant biological functions and has a great correlation with a disease phenotype.
Collapse
Affiliation(s)
- Yansen Su
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei 230601, China
| | - Xiaochun Su
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei 230601, China
| | - Qijun Wang
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei 230601, China.
| | - Lejun Zhang
- Yangzhou Univeristy, Yangzhou 225009, China.
| |
Collapse
|
27
|
Fang Z, Chen L. Personalized prediction of human diseases with single-sample dynamic network biomarkers. Biomark Med 2020; 14:615-620. [PMID: 32530294 DOI: 10.2217/bmm-2020-0066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Zhaoyuan Fang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry & Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry & Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,CAS Center for Excellence in Animal Evolution & Genetics, Chinese Academy of Sciences, Kunming 650223, China.,School of Life Science & Technology, Shanghai Tech University, Shanghai 201210, China.,Shanghai Research Center for Brain Science & Brain-Inspired Intelligence, Shanghai 201210, China
| |
Collapse
|
28
|
Turki T, Taguchi YH. SCGRNs: Novel supervised inference of single-cell gene regulatory networks of complex diseases. Comput Biol Med 2020; 118:103656. [PMID: 32174324 DOI: 10.1016/j.compbiomed.2020.103656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/19/2022]
Abstract
Single-cell gene regulatory network (SCGRN) inference refers to the process of inferring gene regulatory networks from single-cell data, which are generated via single-cell RNA-sequencing (scRNA-seq) technologies. Although scRNA-seq leads to the generation of data pertaining to cells of particular interest, the single-cell data are noisy and highly sparse, which makes the analysis of such data a challenging task. In this study, we model an SCGRN as a directed graph where an edge from a source node (also called transcription factor (TF)) to a target node (also called target gene) indicates that a TF regulates a target gene. Inferring the SCGRN via predicting TF-target gene regulations would help biologists better understand various diseases in terms of networks. Following the modeling step, we propose three machine learning approaches. The first approach considers feature vectors encoding regulatory relationships of expressed TFs-target genes as input. The resulting model is then used to predict unseen TF-target gene regulations. The second machine learning approach constructs new feature vectors via incorporating features obtained from stacked autoencoders, which are provided to a machine learning algorithm to induce a model and predict unseen regulations of TFs-target genes. The third approach extends the second approach via including topological features extracted from an SCGRN. We perform an experimental study comparing our approaches against adapted unsupervised approaches. Experimental results on SCGRNs pertaining to healthy and type 2 pancreatic diabetes demonstrate the clinical importance and the accurate prediction performance of the proposed approaches.
Collapse
Affiliation(s)
- Turki Turki
- King Abdulaziz University, Department of Computer Science, Jeddah, 21589, Saudi Arabia.
| | - Y-H Taguchi
- Chuo University, Department of Physics, Tokyo, 112-8551, Japan.
| |
Collapse
|
29
|
Lan X, Lin W, Xu Y, Xu Y, Lv Z, Chen W. The detection and analysis of differential regulatory communities in lung cancer. Genomics 2020; 112:2535-2540. [PMID: 32045668 DOI: 10.1016/j.ygeno.2020.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/06/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
The tumorgenesis process of lung cancer involves the regulatory dysfunctions of multiple pathways. Although many signaling pathways have been identified to be associated with lung cancer, there are little quantitative models of how inactions between genes change during the process from normal to cancer. These changes belong to different dynamic co-expressions patterns. We quantitatively analyzed differential co-expression of gene pairs in four datasets. Each dataset included a large number of lung cancer and normal samples. By overlapping their results, we got 14 highly confident gene pairs with consistent co-expression change patterns. Some of they, such as ARHGAP30 and GIMAP4, had been recorded in STRING network database while some of them were novel discoveries, such as C9orf135 and MORN5, TEKT1 and TSPAN1 were positively correlated in both normal and cancer but more correlated in normal than cancer. These gene pairs revealed the underlying mechanisms of lung cancer occurrence.
Collapse
Affiliation(s)
- Xiu Lan
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Weilong Lin
- Department of Orthopedics, Lishui Traditional Chinese Medicine Hospital, Lishui, China
| | - Yufen Xu
- Department of Oncology, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing, China; Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Yanyan Xu
- Department of Pharmacy, Lishui Central Hospital, Lishui, China
| | - Zhuqing Lv
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Wenyu Chen
- Department of Respiration, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing, China.
| |
Collapse
|
30
|
Song D, Tang L, Huang J, Wang L, Zeng T, Wang X. Roles of transforming growth factor-β and phosphatidylinositol 3-kinase isoforms in integrin β1-mediated bio-behaviors of mouse lung telocytes. J Transl Med 2019; 17:431. [PMID: 31888636 PMCID: PMC6936066 DOI: 10.1186/s12967-019-02181-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/17/2019] [Indexed: 12/28/2022] Open
Abstract
Background Telocytes (TCs) have the capacity of cell–cell communication with adjacent cells within the tissue, contributing to tissue repair and recovery from injury. The present study aims at investigating the molecular mechanisms by which the TGFβ1-ITGB1-PI3K signal pathways regulate TC cycle and proliferation. Methods Gene expression of integrin (ITG) family were measured in mouse primary TCs to compare with other cells. TC proliferation, movement, cell cycle, and PI3K isoform protein genes were assayed in ITGB1-negative or positive mouse lung TCs treated with the inhibition of PI3Kp110α, PI3Kα/δ, PKCβ, or GSK3, followed by TGFβ1 treatment. Results We found the characters and interactions of ITG or PKC family member networks in primary mouse lung TCs, different from other cells in the lung tissue. The deletion of ITGB1 changed TCs sensitivity to treatment with multifunctional cytokines or signal pathway inhibitors. The compensatory mechanisms occur among TGFβ1-induced PI3Kp110α, PI3Kα/δ, PKCβ, or GSK3 when ITGB1 gene was deleted, leading to alterations of TC cell cycle and proliferation. Of those PI3K isoform protein genes, mRNA expression of PIK3CG altered with ITGB1-negative TC cycle and proliferation. Conclusion TCs have strong capacity of proliferation through the compensatory signaling mechanisms and contribute to the development of drug resistance due to alterations of TC sensitivity.
Collapse
Affiliation(s)
- Dongli Song
- Zhongshan Hospital Institute for Clinical Science, Shanghai Institute of Clinical Bioinformatics, Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Tang
- Zhongshan Hospital Institute for Clinical Science, Shanghai Institute of Clinical Bioinformatics, Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianan Huang
- Zhongshan Hospital Institute for Clinical Science, Shanghai Institute of Clinical Bioinformatics, Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu Wang
- Zhongshan Hospital Institute for Clinical Science, Shanghai Institute of Clinical Bioinformatics, Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiangdong Wang
- Zhongshan Hospital Institute for Clinical Science, Shanghai Institute of Clinical Bioinformatics, Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Roles of TGFβ1 in the expression of phosphoinositide 3-kinase isoform genes and sensitivity and response of lung telocytes to PI3K inhibitors. Cell Biol Toxicol 2019; 36:51-64. [PMID: 31522336 DOI: 10.1007/s10565-019-09487-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The mouse lung telocyte cell line (TCSV40) recently established provides further opportunities to learn TC biology and functions. The present study aims at investigating regulatory roles of phosphoinositide 3-kinase (PI3K) isoforms in TC proliferation and movement and in TGFβ1-induced sensitivity and response of lung TCs to PI3K inhibitors. MATERIALS AND METHODS Network and molecular interactions of genes coding PI3K family or TGFβ family proteins in mouse primary TCs were defined. Mouse lung TCSV40 proliferation, apoptosis, cell cycle, and dynamical bio-behaviors were measured with or without TGFβ1 stimulation or PI3K catalytic isoform protein (PI3K/mTOR, PI3Kα/δ/β, PI3K p110δ, or pan-PI3K) inhibitions. RESULTS The present study showed the difference of network characteristics and interactions of genes coding PI3K isoform proteins or TGFβ family proteins in primary lung telocytes from mouse lungs compared to those of other cells residing in the lung. TGFβ1 had diverse effects on TC proliferation with altered TC number in G2 or S phase, independent upon the administered dose of TGFβ1. PI3Kα/δ/β, PI3K/mTOR, and PI3K p110δ were involved in TC proliferation, of which PI3Kα/δ/β was more sensitive. The effects of pan-PI3K inhibitor indicate that more PI3K isoforms were stimulated by the administering of external TGFβ1 and contributed to TGFβ1-induced TC proliferation. PI3K p110δ upregulated TC proliferation and movement dynamically without TGFβ1, and downregulated TC proliferation with TGFβ1 stimulation, but not TC movement. PI3Kα/δ/β and PI3K/mTOR were more active in TGFβ1-induced S phase accumulation and had similar dynamic effects to PI3K p110δ. Gene expression of PI3K isoforms in TCs was upregulated after TGFβ1 stimulation. The expression of PIK3CA coding p110-α or PIK3CG coding p110-γ were up- or downregulated in TCs without TGFβ1, respectively, when PI3K/mTOR, PI3Kα/δ/β, PI3K p110δ, or pan-PI3K were inhibited. TGFβ1 upregulated the expression of PIK3CA and PIK3CB, while downregulated the expression of PIK3CD and PIK3CG. CONCLUSION Our data imply that TGFβ1 plays divergent roles in the expression of PI3K isoform genes in lung TCs and can alter the sensitivity and response of lung TCs to PI3K inhibitors.
Collapse
|
32
|
Wang J, Li W, Zhou F, Feng R, Wang F, Zhang S, Li J, Li Q, Wang Y, Xie J, Wen T. ATP11B deficiency leads to impairment of hippocampal synaptic plasticity. J Mol Cell Biol 2019; 11:688-702. [PMID: 31152587 PMCID: PMC7261485 DOI: 10.1093/jmcb/mjz042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/28/2019] [Accepted: 03/15/2019] [Indexed: 12/13/2022] Open
Abstract
Synaptic plasticity is known to regulate and support signal transduction between neurons, while synaptic dysfunction contributes to multiple neurological and other brain disorders; however, the specific mechanism underlying this process remains unclear. In the present study, abnormal neural and dendritic morphology was observed in the hippocampus following knockout of Atp11b both in vitro and in vivo. Moreover, ATP11B modified synaptic ultrastructure and promoted spine remodeling via the asymmetrical distribution of phosphatidylserine and enhancement of glutamate release, glutamate receptor expression, and intracellular Ca2+ concentration. Furthermore, experimental results also indicate that ATP11B regulated synaptic plasticity in hippocampal neurons through the MAPK14 signaling pathway. In conclusion, our data shed light on the possible mechanisms underlying the regulation of synaptic plasticity and lay the foundation for the exploration of proteins involved in signal transduction during this process.
Collapse
Affiliation(s)
- Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Weihao Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fangfang Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Ruili Feng
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fushuai Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Shibo Zhang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jie Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yajiang Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
33
|
Chen L. Computational systems biology for omics data analysis. J Mol Cell Biol 2019; 11:631-632. [PMID: 31509200 PMCID: PMC6788723 DOI: 10.1093/jmcb/mjz095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/17/2022] Open
Affiliation(s)
- Luonan Chen
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
34
|
|
35
|
Li M, Li C, Liu WX, Liu C, Cui J, Li Q, Ni H, Yang Y, Wu C, Chen C, Zhen X, Zeng T, Zhao M, Chen L, Wu J, Zeng R, Chen L. Dysfunction of PLA2G6 and CYP2C44-associated network signals imminent carcinogenesis from chronic inflammation to hepatocellular carcinoma. J Mol Cell Biol 2019; 9:489-503. [PMID: 28655161 PMCID: PMC5907842 DOI: 10.1093/jmcb/mjx021] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 06/16/2017] [Indexed: 12/14/2022] Open
Abstract
Little is known about how chronic inflammation contributes to the progression of hepatocellular carcinoma (HCC), especially the initiation of cancer. To uncover the critical transition from chronic inflammation to HCC and the molecular mechanisms at a network level, we analyzed the time-series proteomic data of woodchuck hepatitis virus/c-myc mice and age-matched wt-C57BL/6 mice using our dynamical network biomarker (DNB) model. DNB analysis indicated that the 5th month after birth of transgenic mice was the critical period of cancer initiation, just before the critical transition, which is consistent with clinical symptoms. Meanwhile, the DNB-associated network showed a drastic inversion of protein expression and coexpression levels before and after the critical transition. Two members of DNB, PLA2G6 and CYP2C44, along with their associated differentially expressed proteins, were found to induce dysfunction of arachidonic acid metabolism, further activate inflammatory responses through inflammatory mediator regulation of transient receptor potential channels, and finally lead to impairments of liver detoxification and malignant transition to cancer. As a c-Myc target, PLA2G6 positively correlated with c-Myc in expression, showing a trend from decreasing to increasing during carcinogenesis, with the minimal point at the critical transition or tipping point. Such trend of homologous PLA2G6 and c-Myc was also observed during human hepatocarcinogenesis, with the minimal point at high-grade dysplastic nodules (a stage just before the carcinogenesis). Our study implies that PLA2G6 might function as an oncogene like famous c-Myc during hepatocarcinogenesis, while downregulation of PLA2G6 and c-Myc could be a warning signal indicating imminent carcinogenesis.
Collapse
Affiliation(s)
- Meiyi Li
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China.,Minhang Hospital, Fudan University, Shanghai, China
| | - Chen Li
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Xin Liu
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,University of Chinese Academy of sciences, Beijing, China
| | - Conghui Liu
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of sciences, Beijing, China
| | - Jingru Cui
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Qingrun Li
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Hong Ni
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Yingcheng Yang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
| | - Chaochao Wu
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Chunlei Chen
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Xing Zhen
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Mujun Zhao
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Lei Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China.,National Center for Liver Cancer, Shanghai, China
| | - Jiarui Wu
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Rong Zeng
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, China.,Minhang Hospital, Fudan University, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
36
|
The onset mechanism of Parkinson's beta oscillations: A theoretical analysis. J Theor Biol 2019; 470:1-16. [PMID: 30858065 DOI: 10.1016/j.jtbi.2019.03.008] [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: 08/24/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 11/20/2022]
Abstract
In this paper, we build a basal ganglia-cortex-thalamus model to study the oscillatory mechanisms and boundary conditions of the beta frequency band (13-30 Hz) that appears in the subthalamic nucleus. First, a theoretical oscillatory boundary formula is obtained in a simplified model by using the Laplace transform and linearization process of the system at fixed points. Second, we simulate the oscillatory boundary conditions through numerical calculations, which fit with our theoretical results very well, at least in the changing trend. We find that several critical coupling strengths in the model exert great effects on the oscillations, the mechanisms of which differ but can be explained in detail by our model and the oscillatory boundary formula. Specifically, we note that the relatively small or large sizes of the coupling strength from the fast-spiking interneurons to the medium spiny neurons and from the cortex to the fast-spiking interneurons both have obvious maintenance roles on the states. Similar phenomena have been reported in other neurological diseases, such as absence epilepsy. However, some of those interesting mutual regulation mechanisms in the model have rarely been considered in previous studies. In addition to the coupling weight in the pathway, in this work, we show that the delay is a key parameter that affects oscillations. On the one hand, the system needs a minimum delay to generate oscillations; on the other hand, in the appropriate range, a longer delay leads to a higher activation level of the subthalamic nucleus. In this paper, we study the oscillation activities that appear on the subthalamic nucleus. Moreover, all populations in the model show the dynamic behaviour of a synchronous resonance. Therefore, we infer that the mechanisms obtained can be expanded to explore the state of other populations, and that the model provides a unified framework for studying similar problems in the future. Moreover, the oscillatory boundary curves obtained are all critical conditions between the stable state and beta frequency oscillation. The method is also suitable for depicting other common frequency bands during brain oscillations, such as the alpha band (8-12 Hz), theta band (4-7 Hz) and delta band (1-3 Hz). Thus, the results of this work are expected to help us better understand the onset mechanism of parkinson's oscillations and can inspire related experimental research in this field.
Collapse
|
37
|
Ding T, Gao J, Zhu S, Xu J, Wu M. Predicting microRNA-disease association based on microRNA structural and functional similarity network. QUANTITATIVE BIOLOGY 2019. [DOI: 10.1007/s40484-019-0170-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
38
|
Tang H, Zeng T, Chen L. High-Order Correlation Integration for Single-Cell or Bulk RNA-seq Data Analysis. Front Genet 2019; 10:371. [PMID: 31080457 PMCID: PMC6497731 DOI: 10.3389/fgene.2019.00371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/09/2019] [Indexed: 12/19/2022] Open
Abstract
Quantifying or labeling the sample type with high quality is a challenging task, which is a key step for understanding complex diseases. Reducing noise pollution to data and ensuring the extracted intrinsic patterns in concordance with the primary data structure are important in sample clustering and classification. Here we propose an effective data integration framework named as HCI (High-order Correlation Integration), which takes an advantage of high-order correlation matrix incorporated with pattern fusion analysis (PFA), to realize high-dimensional data feature extraction. On the one hand, the high-order Pearson's correlation coefficient can highlight the latent patterns underlying noisy input datasets and thus improve the accuracy and robustness of the algorithms currently available for sample clustering. On the other hand, the PFA can identify intrinsic sample patterns efficiently from different input matrices by optimally adjusting the signal effects. To validate the effectiveness of our new method, we firstly applied HCI on four single-cell RNA-seq datasets to distinguish the cell types, and we found that HCI is capable of identifying the prior-known cell types of single-cell samples from scRNA-seq data with higher accuracy and robustness than other methods under different conditions. Secondly, we also integrated heterogonous omics data from TCGA datasets and GEO datasets including bulk RNA-seq data, which outperformed the other methods at identifying distinct cancer subtypes. Within an additional case study, we also constructed the mRNA-miRNA regulatory network of colorectal cancer based on the feature weight estimated from HCI, where the differentially expressed mRNAs and miRNAs were significantly enriched in well-known functional sets of colorectal cancer, such as KEGG pathways and IPA disease annotations. All these results supported that HCI has extensive flexibility and applicability on sample clustering with different types and organizations of RNA-seq data.
Collapse
Affiliation(s)
- Hui Tang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| |
Collapse
|
39
|
Kabir MH, Patrick R, Ho JWK, O'Connor MD. Identification of active signaling pathways by integrating gene expression and protein interaction data. BMC SYSTEMS BIOLOGY 2018; 12:120. [PMID: 30598083 PMCID: PMC6311899 DOI: 10.1186/s12918-018-0655-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Signaling pathways are the key biological mechanisms that transduce extracellular signals to affect transcription factor mediated gene regulation within cells. A number of computational methods have been developed to identify the topological structure of a specific signaling pathway using protein-protein interaction data, but they are not designed for identifying active signaling pathways in an unbiased manner. On the other hand, there are statistical methods based on gene sets or pathway data that can prioritize likely active signaling pathways, but they do not make full use of active pathway structure that link receptor, kinases and downstream transcription factors. Results Here, we present a method to simultaneously predict the set of active signaling pathways, together with their pathway structure, by integrating protein-protein interaction network and gene expression data. We evaluated the capacity for our method to predict active signaling pathways for dental epithelial cells, ocular lens epithelial cells, human pluripotent stem cell-derived lens epithelial cells, and lens fiber cells. This analysis showed our approach could identify all the known active pathways that are associated with tooth formation and lens development. Conclusions The results suggest that SPAGI can be a useful approach to identify the potential active signaling pathways given a gene expression profile. Our method is implemented as an open source R package, available via https://github.com/VCCRI/SPAGI/. Electronic supplementary material The online version of this article (10.1186/s12918-018-0655-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Md Humayun Kabir
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Department of Computer Science and Engineering, University of Rajshahi, Rajshahi, Bangladesh
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Joshua W K Ho
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. .,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia. .,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR, China.
| | - Michael D O'Connor
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia. .,Molecular Medicine Research Group, Western Sydney University, Campbelltown, NSW, Australia.
| |
Collapse
|
40
|
Liu X, Chang X, Leng S, Tang H, Aihara K, Chen L. Detection for disease tipping points by landscape dynamic network biomarkers. Natl Sci Rev 2018; 6:775-785. [PMID: 34691933 PMCID: PMC8291500 DOI: 10.1093/nsr/nwy162] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 12/19/2018] [Accepted: 12/25/2018] [Indexed: 12/15/2022] Open
Abstract
A new model-free method has been developed and termed the landscape dynamic network biomarker (l-DNB) methodology. The method is based on bifurcation theory, which can identify tipping points prior to serious disease deterioration using only single-sample omics data. Here, we show that l-DNB provides early-warning signals of disease deterioration on a single-sample basis and also detects critical genes or network biomarkers (i.e. DNB members) that promote the transition from normal to disease states. As a case study, l-DNB was used to predict severe influenza symptoms prior to the actual symptomatic appearance in influenza virus infections. The l-DNB approach was then also applied to three tumor disease datasets from the TCGA and was used to detect critical stages prior to tumor deterioration using an individual DNB for each patient. The individual DNBs were further used as individual biomarkers in the analysis of physiological data, which led to the identification of two biomarker types that were surprisingly effective in predicting the prognosis of tumors. The biomarkers can be considered as common biomarkers for cancer, wherein one indicates a poor prognosis and the other indicates a good prognosis.
Collapse
Affiliation(s)
- Xiaoping Liu
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- School of Mathematics and Statistics, Shandong University at Weihai, Weihai 264209, China
- Institute of Industrial Science, the University of Tokyo, Tokyo 153–8505, Japan
| | - Xiao Chang
- Institute of Industrial Science, the University of Tokyo, Tokyo 153–8505, Japan
- Institute of Statistics and Applied Mathematics, Anhui University of Finance & Economics, Bengbu 233030, China
| | - Siyang Leng
- Institute of Industrial Science, the University of Tokyo, Tokyo 153–8505, Japan
| | - Hui Tang
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kazuyuki Aihara
- Institute of Industrial Science, the University of Tokyo, Tokyo 153–8505, Japan
| | - Luonan Chen
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Industrial Science, the University of Tokyo, Tokyo 153–8505, Japan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Center for Excellence in Animal Evolution and Genetics, Kunming 650223, China
- Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| |
Collapse
|
41
|
PWCDA: Path Weighted Method for Predicting circRNA-Disease Associations. Int J Mol Sci 2018; 19:ijms19113410. [PMID: 30384427 PMCID: PMC6274797 DOI: 10.3390/ijms19113410] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
CircRNAs have particular biological structure and have proven to play important roles in diseases. It is time-consuming and costly to identify circRNA-disease associations by biological experiments. Therefore, it is appealing to develop computational methods for predicting circRNA-disease associations. In this study, we propose a new computational path weighted method for predicting circRNA-disease associations. Firstly, we calculate the functional similarity scores of diseases based on disease-related gene annotations and the semantic similarity scores of circRNAs based on circRNA-related gene ontology, respectively. To address missing similarity scores of diseases and circRNAs, we calculate the Gaussian Interaction Profile (GIP) kernel similarity scores for diseases and circRNAs, respectively, based on the circRNA-disease associations downloaded from circR2Disease database (http://bioinfo.snnu.edu.cn/CircR2Disease/). Then, we integrate disease functional similarity scores and circRNA semantic similarity scores with their related GIP kernel similarity scores to construct a heterogeneous network made up of three sub-networks: disease similarity network, circRNA similarity network and circRNA-disease association network. Finally, we compute an association score for each circRNA-disease pair based on paths connecting them in the heterogeneous network to determine whether this circRNA-disease pair is associated. We adopt leave one out cross validation (LOOCV) and five-fold cross validations to evaluate the performance of our proposed method. In addition, three common diseases, Breast Cancer, Gastric Cancer and Colorectal Cancer, are used for case studies. Experimental results illustrate the reliability and usefulness of our computational method in terms of different validation measures, which indicates PWCDA can effectively predict potential circRNA-disease associations.
Collapse
|
42
|
Hu B, Shi Q, Guo Y, Diao X, Guo H, Zhang J, Yu L, Dai H, Chen L. The oscillatory boundary conditions of different frequency bands in Parkinson's disease. J Theor Biol 2018; 451:67-79. [PMID: 29727632 DOI: 10.1016/j.jtbi.2018.04.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/10/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease that is common in the elderly population. The most important pathological change in PD is the degeneration and death of dopaminergic neurons in the substantia nigra of the midbrain, which results in a decrease in the dopamine (DA) content of the striatum. The exact cause of this pathological change is still unknown. Numerous studies have shown that the evolution of PD is associated with abnormal oscillatory activities in the basal ganglia, with different oscillation frequency ranges, such as the typical beta band (13-30 Hz), the alpha band (8-12 Hz), the theta band (4-7 Hz) and the delta band (1-3 Hz). Although some studies have implied that abnormal interactions between the subthalamic nucleus (STN) and globus pallidus (GP) neurons may be a key factor required to induce these oscillations, the relative mechanism is still unclear. The effects of other nerve nuclei in the basal ganglia, such as the striatum, on these oscillations are still unknown. The thalamus and cortex both have close input and output relationships with the basal ganglia, and many previous studies have indicated that they may also exert effects on Parkinson's disease oscillation, but the mechanisms involved are unclear. In this paper, we built a corticothalamic-basal ganglia (CTBG) mean firing-rate model to explore the onset mechanisms of these different oscillation phenomena. We found that, in addition to the STN-GP network, Parkinson's disease oscillations may also be induced by changing the coupling strength and delays in other pathways. Different frequency bands appear in the oscillating region, and various boundary conditions are depicted in parameter diagrams. The onset mechanism is well explained both by the model and by the numerical simulation results. Therefore, this model provides a unifying framework for studying the mechanism of Parkinson's disease oscillations, and we hope that the results obtained in this work can inspire future experimental studies.
Collapse
Affiliation(s)
- Bing Hu
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Qianqian Shi
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Yu Guo
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiyezi Diao
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Heng Guo
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinsong Zhang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Yu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hao Dai
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
43
|
Zhang J, Guo J, Zhang M, Yu X, Yu X, Guo W, Zeng T, Chen L. Efficient Mining Multi-mers in a Variety of Biological Sequences. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 17:949-958. [PMID: 29993642 DOI: 10.1109/tcbb.2018.2828313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Counting the occurrence frequency of each -mer in a biological sequence is a preliminary yet important step in many bioinformatics applications. However, most -mer counting algorithms rely on a given k to produce single-length -mers, which is inefficient for sequence analysis for different k. Moreover, existing -mer counters focus more on DNA and RNA sequences and less on protein ones. In practice, the analysis of -mers in protein sequences can provide substantial biological insights in structure, function and evolution. To this end, an efficient algorithm, called MulMer (Multiple-Mer mining), is proposed to mine -mers of various lengths termed multi-mers via inverted-index technique, which is orders of magnitude faster than the conventional forward-index methods. Moreover, to the best of our knowledge, MulMer is the first able to mine multi-mers in a variety of sequences, including DNARNA and protein sequences.
Collapse
|
44
|
Hu B, Guo Y, Shi F, Zou X, Dong J, Pan L, Yu M, Zhou C, Cheng Z, Tang W, Sun H, Chen L. The generation mechanism of spike-and-slow wave discharges appearing on thalamic relay nuclei. Sci Rep 2018; 8:4953. [PMID: 29563579 PMCID: PMC5862852 DOI: 10.1038/s41598-018-23280-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 03/08/2018] [Indexed: 12/19/2022] Open
Abstract
In this paper, we use a model modified from classic corticothalamic network(CT) to explore the mechanism of absence seizures appearing on specific relay nuclei (SRN) of the thalamus. It is found that typical seizure states appear on SRN through tuning several critical connection strengths in the model. In view of previous experimental and theoretical works which were mainly on epilepsy seizure phenomena appearing on excitatory pyramidal neurons (EPN) of the cortex, this is a novel model to consider the seizure observed on thalamus. In particular, the onset mechanism is different from previous theoretical studies. Inspired by some previous clinical and experimental studies, we employ the external stimuli voltage on EPN and SRN in the network, and observe that the seizure can be well inhibited by tuning the stimulus intensity appropriately. We further explore the effect of the signal transmission delays on seizures, and found that the polyspike phenomenon appears only when the delay is sufficiently large. The experimental data also confirmed our model. Since there is a complex network in the brain and all organizations are interacting closely with each other, the results obtained in this paper provide not only biological insights into the regulatory mechanisms but also a reference for the prevention and treatment of epilepsy in future.
Collapse
Affiliation(s)
- Bing Hu
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China.
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yu Guo
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Feng Shi
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaoqiang Zou
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jing Dong
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Long Pan
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Min Yu
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chaowei Zhou
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhang Cheng
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wanyue Tang
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Haochen Sun
- Institute of Applied Mathematics, Department of Mathematics and Statistics, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
45
|
Li QR, Wang ZM, Wewer Albrechtsen NJ, Wang DD, Su ZD, Gao XF, Wu QQ, Zhang HP, Zhu L, Li RX, Jacobsen S, Jørgensen NB, Dirksen C, Bojsen-Møller KN, Petersen JS, Madsbad S, Clausen TR, Diderichsen B, Chen LN, Holst JJ, Zeng R, Wu JR. Systems Signatures Reveal Unique Remission-path of Type 2 Diabetes Following Roux-en-Y Gastric Bypass Surgery. EBioMedicine 2018; 28:234-240. [PMID: 29422288 PMCID: PMC5835566 DOI: 10.1016/j.ebiom.2018.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/14/2022] Open
Abstract
Roux-en-Y Gastric bypass surgery (RYGB) is emerging as a powerful tool for treatment of obesity and may also cause remission of type 2 diabetes. However, the molecular mechanism of RYGB leading to diabetes remission independent of weight loss remains elusive. In this study, we profiled plasma metabolites and proteins of 10 normal glucose-tolerant obese (NO) and 9 diabetic obese (DO) patients before and 1-week, 3-months, 1-year after RYGB. 146 proteins and 128 metabolites from both NO and DO groups at all four stages were selected for further analysis. By analyzing a set of bi-molecular associations among the corresponding network of the subjects with our newly developed computational method, we defined the represented physiological states (called the edge-states that reflect the interactions among the bio-molecules), and the related molecular networks of NO and DO patients, respectively. The principal component analyses (PCA) revealed that the edge states of the post-RYGB NO subjects were significantly different from those of the post-RYGB DO patients. Particularly, the time-dependent changes of the molecular hub-networks differed between DO and NO groups after RYGB. In conclusion, by developing molecular network-based systems signatures, we for the first time reveal that RYGB generates a unique path for diabetes remission independent of weight loss.
Collapse
Affiliation(s)
- Qing-Run Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zi-Ming Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, China
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dan-Dan Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, China
| | - Zhi-Duan Su
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xian-Fu Gao
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qing-Qing Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hui-Ping Zhang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Li Zhu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Rong-Xia Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - SivHesse Jacobsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nils Bruun Jørgensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Dirksen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine N Bojsen-Møller
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Sten Madsbad
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Luo-Nan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Rong Zeng
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.
| | - Jia-Rui Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.
| |
Collapse
|
46
|
Zhao XM, Li S. HISP: a hybrid intelligent approach for identifying directed signaling pathways. J Mol Cell Biol 2018; 9:453-462. [DOI: 10.1093/jmcb/mjx054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/20/2017] [Indexed: 01/15/2023] Open
Affiliation(s)
- Xing-Ming Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
| | - Shan Li
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| |
Collapse
|
47
|
Guo WF, Zhang SW, Shi QQ, Zhang CM, Zeng T, Chen L. A novel algorithm for finding optimal driver nodes to target control complex networks and its applications for drug targets identification. BMC Genomics 2018; 19:924. [PMID: 29363426 PMCID: PMC5780855 DOI: 10.1186/s12864-017-4332-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background The advances in target control of complex networks not only can offer new insights into the general control dynamics of complex systems, but also be useful for the practical application in systems biology, such as discovering new therapeutic targets for disease intervention. In many cases, e.g. drug target identification in biological networks, we usually require a target control on a subset of nodes (i.e., disease-associated genes) with minimum cost, and we further expect that more driver nodes consistent with a certain well-selected network nodes (i.e., prior-known drug-target genes). Results Therefore, motivated by this fact, we pose and address a new and practical problem called as target control problem with objectives-guided optimization (TCO): how could we control the interested variables (or targets) of a system with the optional driver nodes by minimizing the total quantity of drivers and meantime maximizing the quantity of constrained nodes among those drivers. Here, we design an efficient algorithm (TCOA) to find the optional driver nodes for controlling targets in complex networks. We apply our TCOA to several real-world networks, and the results support that our TCOA can identify more precise driver nodes than the existing control-fucus approaches. Furthermore, we have applied TCOA to two bimolecular expert-curate networks. Source code for our TCOA is freely available from http://sysbio.sibcb.ac.cn/cb/chenlab/software.htm or https://github.com/WilfongGuo/guoweifeng. Conclusions In the previous theoretical research for the full control, there exists an observation and conclusion that the driver nodes tend to be low-degree nodes. However, for target control the biological networks, we find interestingly that the driver nodes tend to be high-degree nodes, which is more consistent with the biological experimental observations. Furthermore, our results supply the novel insights into how we can efficiently target control a complex system, and especially many evidences on the practical strategic utility of TCOA to incorporate prior drug information into potential drug-target forecasts. Thus applicably, our method paves a novel and efficient way to identify the drug targets for leading the phenotype transitions of underlying biological networks. Electronic supplementary material The online version of this article (doi: 10.1186/s12864-017-4332-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Qian-Qian Shi
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China
| | - Cheng-Ming Zhang
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China.
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China. .,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 200000, China.
| |
Collapse
|
48
|
Yu X, Zhang J, Sun S, Zhou X, Zeng T, Chen L. Individual-specific edge-network analysis for disease prediction. Nucleic Acids Res 2017; 45:e170. [PMID: 28981699 PMCID: PMC5714249 DOI: 10.1093/nar/gkx787] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/10/2017] [Indexed: 12/19/2022] Open
Abstract
Predicting pre-disease state or tipping point just before irreversible deterioration of health is a difficult task. Edge-network analysis (ENA) with dynamic network biomarker (DNB) theory opens a new way to study this problem by exploring rich dynamical and high-dimensional information of omics data. Although theoretically ENA has the ability to identify the pre-disease state during the disease progression, it requires multiple samples for such prediction on each individual, which are generally not available in clinical practice, thus limiting its applications in personalized medicine. In this work to overcome this problem, we propose the individual-specific ENA (iENA) with DNB to identify the pre-disease state of each individual in a single-sample manner. In particular, iENA can identify individual-specific biomarkers for the disease prediction, in addition to the traditional disease diagnosis. To demonstrate the effectiveness, iENA was applied to the analysis on omics data of H3N2 cohorts and successfully detected early-warning signals of the influenza infection for each individual both on the occurred time and event in an accurate manner, which actually achieves the AUC larger than 0.9. iENA not only found the new individual-specific biomarkers but also recovered the common biomarkers of influenza infection reported from previous works. In addition, iENA also detected the critical stages of multiple cancers with significant edge-biomarkers, which were further validated by survival analysis on both TCGA data and other independent data.
Collapse
Affiliation(s)
- Xiangtian Yu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Chinese Academy Science, Shanghai 200031, China
| | - Jingsong Zhang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Chinese Academy Science, Shanghai 200031, China
| | - Shaoyan Sun
- School of Mathematics and Information, Ludong University, Yantai 264025, China
| | - Xin Zhou
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Chinese Academy Science, Shanghai 200031, China.,University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Chinese Academy Science, Shanghai 200031, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Chinese Academy Science, Shanghai 200031, China.,University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
| |
Collapse
|
49
|
Zhang L, Liu Y, Wang M, Wu Z, Li N, Zhang J, Yang C. EZH2-, CHD4-, and IDH-linked epigenetic perturbation and its association with survival in glioma patients. J Mol Cell Biol 2017; 9:477-488. [PMID: 29272522 PMCID: PMC5907834 DOI: 10.1093/jmcb/mjx056] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/12/2017] [Accepted: 12/18/2017] [Indexed: 12/13/2022] Open
Abstract
Glioma is a complex disease with limited treatment options. Recent advances have identified isocitrate dehydrogenase (IDH) mutations in up to 80% lower grade gliomas (LGG) and in 76% secondary glioblastomas (GBM). IDH mutations are also seen in 10%-20% of acute myeloid leukemia (AML). In AML, it was determined that mutations of IDH and other genes involving epigenetic regulations are early events, emerging in the pre-leukemic stem cells (pre-LSCs) stage, whereas mutations in genes propagating oncogenic signal are late events in leukemia. IDH mutations are also early events in glioma, occurring before TP53 mutation, 1p/19q deletion, etc. Despite these advances in glioma research, studies into other molecular alterations have lagged considerably. In this study, we analyzed currently available databases. We identified EZH2, KMT2C, and CHD4 as important genes in glioma in addition to the known gene IDH1/2. We also showed that genomic alterations of PIK3CA, CDKN2A, CDK4, FIP1L1, or FUBP1 collaborate with IDH mutations to negatively affect patients' survival in LGG. In LGG patients with TP53 mutations or IDH1/2 mutations, additional genomic alterations of EZH2, KMC2C, and CHD4 individually or in combination were associated with a markedly decreased disease-free survival than patients without such alterations. Alterations of EZH2, KMT2C, and CHD4 at genetic level or protein level could perturb epigenetic program, leading to malignant transformation in glioma. By reviewing current literature on both AML and glioma and performing bioinformatics analysis on available datasets, we developed a hypothetical model on the tumorigenesis from premalignant stem cells to glioma.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer Science, Sichuan University, Chengdu, China
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Ying Liu
- The Vivian Smith Department of Neurosurgery, Center for Stem Cell and Regenerative Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mengning Wang
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Zhenhai Wu
- Department of neurosurgery, ShouGuang People’s Hospital, Shandong, China
| | - Na Li
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Jinsong Zhang
- Pharmacological & Physiological Science, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Chuanwei Yang
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
50
|
Liu X, Wang Y, Ji H, Aihara K, Chen L. Personalized characterization of diseases using sample-specific networks. Nucleic Acids Res 2016; 44:e164. [PMID: 27596597 PMCID: PMC5159538 DOI: 10.1093/nar/gkw772] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 01/20/2023] Open
Abstract
A complex disease generally results not from malfunction of individual molecules but from dysfunction of the relevant system or network, which dynamically changes with time and conditions. Thus, estimating a condition-specific network from a single sample is crucial to elucidating the molecular mechanisms of complex diseases at the system level. However, there is currently no effective way to construct such an individual-specific network by expression profiling of a single sample because of the requirement of multiple samples for computing correlations. We developed here with a statistical method, i.e. a sample-specific network (SSN) method, which allows us to construct individual-specific networks based on molecular expressions of a single sample. Using this method, we can characterize various human diseases at a network level. In particular, such SSNs can lead to the identification of individual-specific disease modules as well as driver genes, even without gene sequencing information. Extensive analysis by using the Cancer Genome Atlas data not only demonstrated the effectiveness of the method, but also found new individual-specific driver genes and network patterns for various types of cancer. Biological experiments on drug resistance further validated one important advantage of our method over the traditional methods, i.e. we can even identify such drug resistance genes that actually have no clear differential expression between samples with and without the resistance, due to the additional network information.
Collapse
Affiliation(s)
- Xiaoping Liu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Industrial Science, University of Tokyo, Tokyo 153-8505, Japan
| | - Yuetong Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongbin Ji
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Kazuyuki Aihara
- Institute of Industrial Science, University of Tokyo, Tokyo 153-8505, Japan
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Industrial Science, University of Tokyo, Tokyo 153-8505, Japan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|