51
|
Liu Z, Miller D, Li F, Liu X, Levy SF. A large accessory protein interactome is rewired across environments. eLife 2020; 9:e62365. [PMID: 32924934 PMCID: PMC7577743 DOI: 10.7554/elife.62365] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/30/2022] Open
Abstract
To characterize how protein-protein interaction (PPI) networks change, we quantified the relative PPI abundance of 1.6 million protein pairs in the yeast Saccharomyces cerevisiae across nine growth conditions, with replication, for a total of 44 million measurements. Our multi-condition screen identified 13,764 pairwise PPIs, a threefold increase over PPIs identified in one condition. A few 'immutable' PPIs are present across all conditions, while most 'mutable' PPIs are rarely observed. Immutable PPIs aggregate into highly connected 'core' network modules, with most network remodeling occurring within a loosely connected 'accessory' module. Mutable PPIs are less likely to co-express, co-localize, and be explained by simple mass action kinetics, and more likely to contain proteins with intrinsically disordered regions, implying that environment-dependent association and binding is critical to cellular adaptation. Our results show that protein interactomes are larger than previously thought and contain highly dynamic regions that reorganize to drive or respond to cellular changes.
Collapse
Affiliation(s)
- Zhimin Liu
- Department of Biochemistry, Stony Brook UniversityStony BrookUnited States
- Laufer Center for Physical and Quantitative Biology, Stony Brook UniversityStony BrookUnited States
| | - Darach Miller
- Joint Initiative for Metrology in BiologyStanfordUnited States
- Department of Genetics, Stanford UniversityStanfordUnited States
| | - Fangfei Li
- Laufer Center for Physical and Quantitative Biology, Stony Brook UniversityStony BrookUnited States
- Department of Applied Mathematics and Statistics, Stony Brook UniversityStony BrookUnited States
| | - Xianan Liu
- Department of Biochemistry, Stony Brook UniversityStony BrookUnited States
- Laufer Center for Physical and Quantitative Biology, Stony Brook UniversityStony BrookUnited States
| | - Sasha F Levy
- Department of Biochemistry, Stony Brook UniversityStony BrookUnited States
- Laufer Center for Physical and Quantitative Biology, Stony Brook UniversityStony BrookUnited States
- Joint Initiative for Metrology in BiologyStanfordUnited States
- Department of Genetics, Stanford UniversityStanfordUnited States
- Department of Applied Mathematics and Statistics, Stony Brook UniversityStony BrookUnited States
- SLAC National Accelerator LaboratoryMenlo ParkUnited States
| |
Collapse
|
52
|
Uddin N, Hussain M, Rauf I, Zaidi SF. Identification of key pathways and genes responsible for aggressive behavior. Comput Biol Chem 2020; 88:107349. [PMID: 32763796 DOI: 10.1016/j.compbiolchem.2020.107349] [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: 04/29/2019] [Revised: 03/03/2020] [Accepted: 07/26/2020] [Indexed: 11/15/2022]
Abstract
Aggression is a complex behavior, underpinned by cross talk between several biomolecules. To date a composite molecular network of the behavioral disorder has not been constructed. The present study aims to develop the same from the system network analyses recruiting genes with empirical evidence demonstrating their role in the incidence and progression of aggression. In short, 327 genes were recruited in the study after extensive literature survey and subsequent shortlisting by sieving out the comorbidities like cancer and other pathological and physiological ailments, other languages and repeated citations. Subsequent String network analysis coalesces 275 genes in a network with 2223 edges. The developed network was then subjected to delineate modules using MCODE which via gene clustering on the basis of gene ontology segregate all genes into 14 modules. Of these, as expected top 5 modules involved entailing of neuronal signaling pathways with redundant repetitions. Finally, 10 genes (known) were picked randomly, accounting average module size, and subjected to the network analysis with 100,000 bootstrap replicates. This results in the detection of certain novel genes that lacks empirical evidence for their association with the aggression. Amongst those, most notable are genes involved in protein turnover regulation like UBC, UBA, mitogenic proteins such as Rho and Myc, transcription factors like Tp53. The findings in turn fill caveats in the molecular resolution of cross talk that underscore the development of aggressive behavior and may then be exploited as screening biomarker and/or therapeutic intervention for aggression.
Collapse
Affiliation(s)
- Nasir Uddin
- Department of Computer Science, IBA, Karachi, Pakistan.
| | - Mushtaq Hussain
- Bioinformatics and Molecular Medicine Research Group, Dow Research Institute of Biotechnology and Biomedical Sciences, Dow College of Biotechnology, Dow University of Health Sciences, Karachi, Pakistan.
| | - Imran Rauf
- Department of Computer Science, IBA, Karachi, Pakistan.
| | | |
Collapse
|
53
|
Perscheid C. Integrative biomarker detection on high-dimensional gene expression data sets: a survey on prior knowledge approaches. Brief Bioinform 2020; 22:5881664. [PMID: 32761115 DOI: 10.1093/bib/bbaa151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Gene expression data provide the expression levels of tens of thousands of genes from several hundred samples. These data are analyzed to detect biomarkers that can be of prognostic or diagnostic use. Traditionally, biomarker detection for gene expression data is the task of gene selection. The vast number of genes is reduced to a few relevant ones that achieve the best performance for the respective use case. Traditional approaches select genes based on their statistical significance in the data set. This results in issues of robustness, redundancy and true biological relevance of the selected genes. Integrative analyses typically address these shortcomings by integrating multiple data artifacts from the same objects, e.g. gene expression and methylation data. When only gene expression data are available, integrative analyses instead use curated information on biological processes from public knowledge bases. With knowledge bases providing an ever-increasing amount of curated biological knowledge, such prior knowledge approaches become more powerful. This paper provides a thorough overview on the status quo of biomarker detection on gene expression data with prior biological knowledge. We discuss current shortcomings of traditional approaches, review recent external knowledge bases, provide a classification and qualitative comparison of existing prior knowledge approaches and discuss open challenges for this kind of gene selection.
Collapse
Affiliation(s)
- Cindy Perscheid
- Hasso Plattner Institute, University of Potsdam, Potsdam, 14482, Germany
| |
Collapse
|
54
|
Li Q, Dai W, Liu J, Sang Q, Li YX, Li YY. Gene dysregulation analysis builds a mechanistic signature for prognosis and therapeutic benefit in colorectal cancer. J Mol Cell Biol 2020; 12:881-893. [PMID: 32717065 PMCID: PMC7883816 DOI: 10.1093/jmcb/mjaa041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/09/2022] Open
Abstract
The implementation of cancer precision medicine requires biomarkers or signatures for predicting prognosis and therapeutic benefits. Most of current efforts in this field are paying much more attention to predictive accuracy than to molecular mechanistic interpretability. Mechanism-driven strategy has recently emerged, aiming to build signatures with both predictive power and explanatory power. Driven by this strategy, we developed a robust gene dysregulation analysis framework with machine learning algorithms, which is capable of exploring gene dysregulations underlying carcinogenesis from high-dimensional data with cooperativity and synergy between regulators and several other transcriptional regulation rules taken into consideration. We then applied the framework to a colorectal cancer (CRC) cohort from The Cancer Genome Atlas. The identified CRC-related dysregulations significantly covered known carcinogenic processes and exhibited good prognostic effect. By choosing dysregulations with greedy strategy, we built a four-dysregulation (4-DysReg) signature, which has the capability of predicting prognosis and adjuvant chemotherapy benefit. 4-DysReg has the potential to explain carcinogenesis in terms of dysfunctional transcriptional regulation. These results demonstrate that our gene dysregulation analysis framework could be used to develop predictive signature with mechanistic interpretability for cancer precision medicine, and furthermore, elucidate the mechanisms of carcinogenesis.
Collapse
Affiliation(s)
- Quanxue Li
- School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China.,Shanghai Center for Bioinformation Technology, Shanghai 201203, China
| | - Wentao Dai
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Engineering Research Center of Pharmaceutical Translation and Shanghai Industrial Technology Institute, Shanghai 201203, China
| | - Jixiang Liu
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China.,Shanghai Engineering Research Center of Pharmaceutical Translation and Shanghai Industrial Technology Institute, Shanghai 201203, China
| | - Qingqing Sang
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi-Xue Li
- School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China.,Shanghai Center for Bioinformation Technology, Shanghai 201203, China.,CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Shanghai Engineering Research Center of Pharmaceutical Translation and Shanghai Industrial Technology Institute, Shanghai 201203, China
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China.,Shanghai Engineering Research Center of Pharmaceutical Translation and Shanghai Industrial Technology Institute, Shanghai 201203, China
| |
Collapse
|
55
|
Chowdhury HA, Bhattacharyya DK, Kalita JK. (Differential) Co-Expression Analysis of Gene Expression: A Survey of Best Practices. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1154-1173. [PMID: 30668502 DOI: 10.1109/tcbb.2019.2893170] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Analysis of gene expression data is widely used in transcriptomic studies to understand functions of molecules inside a cell and interactions among molecules. Differential co-expression analysis studies diseases and phenotypic variations by finding modules of genes whose co-expression patterns vary across conditions. We review the best practices in gene expression data analysis in terms of analysis of (differential) co-expression, co-expression network, differential networking, and differential connectivity considering both microarray and RNA-seq data along with comparisons. We highlight hurdles in RNA-seq data analysis using methods developed for microarrays. We include discussion of necessary tools for gene expression analysis throughout the paper. In addition, we shed light on scRNA-seq data analysis by including preprocessing and scRNA-seq in co-expression analysis along with useful tools specific to scRNA-seq. To get insights, biological interpretation and functional profiling is included. Finally, we provide guidelines for the analyst, along with research issues and challenges which should be addressed.
Collapse
|
56
|
Ma X, Sun P, Gong M. An integrative framework of heterogeneous genomic data for cancer dynamic modules based on matrix decomposition. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 19:305-316. [PMID: 32750874 DOI: 10.1109/tcbb.2020.3004808] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer progression is dynamic, and tracking dynamic modules is promising for cancer diagnosis and therapy. Accumulated genomic data provide us an opportunity to investigate the underlying mechanisms of cancers. However, as far as we know, no algorithm has been designed for dynamic modules by integrating heterogeneous omics data. To address this issue, we propose an integrative framework for dynamic module detection based on regularized nonnegative matrix factorization method (DrNMF) by integrating the gene expression and protein interaction network. To remove the heterogeneity of genomic data, we divide the samples of expression profiles into groups to construct gene co-expression networks. To characterize the dynamics of modules, the temporal smoothness framework is adopted, in which the gene co-expression network at the previous stage and protein interaction network are incorporated into the objective function of DrNMF via regularization. The experimental results demonstrate that DrNMF is superior to state-of-the-art methods in terms of accuracy. For breast cancer data, the obtained dynamic modules are more enriched by the known pathways, and can be used to predict the stages of cancers and survival time of patients. The proposed model and algorithm provide an effective integrative analysis of heterogeneous genomic data for cancer progression.
Collapse
|
57
|
Functional genomics identifies new synergistic therapies for retinoblastoma. Oncogene 2020; 39:5338-5357. [PMID: 32572160 PMCID: PMC7391301 DOI: 10.1038/s41388-020-1372-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/03/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022]
Abstract
Local intravitreal or intra-arterial chemotherapy has improved therapeutic success for the pediatric cancer retinoblastoma (RB), but toxicity remains a major caveat. RB initiates primarily with RB1 loss or, rarely, MYCN amplification, but the critical downstream networks are incompletely understood. We set out to uncover perturbed molecular hubs, identify synergistic drug combinations to target these vulnerabilities, and expose and overcome drug resistance. We applied dynamic transcriptomic analysis to identify network hubs perturbed in RB versus normal fetal retina, and performed in vivo RNAi screens in RB1null and RB1wt;MYCNamp orthotopic xenografts to pinpoint essential hubs. We employed in vitro and in vivo studies to validate hits, define mechanism, develop new therapeutic modalities, and understand drug resistance. We identified BRCA1 and RAD51 as essential for RB cell survival. Their oncogenic activity was independent of BRCA1 functions in centrosome, heterochromatin, or ROS regulation, and instead linked to DNA repair. RAD51 depletion or inhibition with the small molecule inhibitor, B02, killed RB cells in a Chk1/Chk2/p53-dependent manner. B02 further synergized with clinically relevant topotecan (TPT) to engage this pathway, activating p53-BAX mediated killing of RB but not human retinal progenitor cells. Paradoxically, a B02/TPT-resistant tumor exhibited more DNA damage than sensitive RB cells. Resistance reflected dominance of the p53-p21 axis, which mediated cell cycle arrest instead of death. Deleting p21 or applying the BCL2/BCL2L1 inhibitor Navitoclax re-engaged the p53-BAX axis, and synergized with B02, TPT or both to override resistance. These data expose new synergistic therapies to trigger p53-induced killing in diverse RB subtypes.
Collapse
|
58
|
Gysi DM, Nowick K. Construction, comparison and evolution of networks in life sciences and other disciplines. J R Soc Interface 2020; 17:20190610. [PMID: 32370689 PMCID: PMC7276545 DOI: 10.1098/rsif.2019.0610] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Network approaches have become pervasive in many research fields. They allow for a more comprehensive understanding of complex relationships between entities as well as their group-level properties and dynamics. Many networks change over time, be it within seconds or millions of years, depending on the nature of the network. Our focus will be on comparative network analyses in life sciences, where deciphering temporal network changes is a core interest of molecular, ecological, neuropsychological and evolutionary biologists. Further, we will take a journey through different disciplines, such as social sciences, finance and computational gastronomy, to present commonalities and differences in how networks change and can be analysed. Finally, we envision how borrowing ideas from these disciplines could enrich the future of life science research.
Collapse
Affiliation(s)
- Deisy Morselli Gysi
- Department of Computer Science, Interdisciplinary Center of Bioinformatics, University of Leipzig, 04109 Leipzig, Germany
- Swarm Intelligence and Complex Systems Group, Faculty of Mathematics and Computer Science, University of Leipzig, 04109 Leipzig, Germany
- Center for Complex Networks Research, Northeastern University, 177 Huntington Avenue, Boston, MA 02115, USA
| | - Katja Nowick
- Human Biology Group, Institute for Biology, Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Königin-Luise-Straβe 1-3, 14195 Berlin, Germany
| |
Collapse
|
59
|
Poverennaya EV, Kiseleva OI, Ivanov AS, Ponomarenko EA. Methods of Computational Interactomics for Investigating Interactions of Human Proteoforms. BIOCHEMISTRY (MOSCOW) 2020; 85:68-79. [PMID: 32079518 DOI: 10.1134/s000629792001006x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human genome contains ca. 20,000 protein-coding genes that could be translated into millions of unique protein species (proteoforms). Proteoforms coded by a single gene often have different functions, which implies different protein partners. By interacting with each other, proteoforms create a network reflecting the dynamics of cellular processes in an organism. Perturbations of protein-protein interactions change the network topology, which often triggers pathological processes. Studying proteoforms is a relatively new research area in proteomics, and this is why there are comparatively few experimental studies on the interaction of proteoforms. Bioinformatics tools can facilitate such studies by providing valuable complementary information to the experimental data and, in particular, expanding the possibilities of the studies of proteoform interactions.
Collapse
Affiliation(s)
| | - O I Kiseleva
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | - A S Ivanov
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | | |
Collapse
|
60
|
Adnan N, Liu Z, Huang THM, Ruan J. Comparative evaluation of network features for the prediction of breast cancer metastasis. BMC Med Genomics 2020; 13:40. [PMID: 32241278 PMCID: PMC7119280 DOI: 10.1186/s12920-020-0676-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Discovering a highly accurate and robust gene signature for the prediction of breast cancer metastasis from gene expression profiling of primary tumors is one of the most challenging tasks to reduce the number of deaths in women. Due to the limited success of gene-based features in achieving satisfactory prediction accuracy, many methodologies have been proposed in recent years to develop network-based features by integrating network information with gene expression. However, evaluation results are inconsistent to confirm the effectiveness of network-based features, because of many confounding factors involved in classification model learning process, such as data normalization, dimension reduction, and feature selection. An unbiased comparative evaluation is essential for uncovering the strength of network-based features. Methods In this study, we compared several types of network-based features obtained using different mathematical operators (Mean, Maximum, Minimum, Median, Variance) on geneset (i.e., a gene and its’ neighbors in the network) in protein-protein interaction network and gene co-expression network for their ability in predicting breast cancer metastasis using gene expression data from more than 10 patient cohorts. Results While network-based features are usually statistically more significant than gene-based feature, a consistent improvement of prediction performance using network-based features requires a substantial number of patients in the dataset. In contrary to many previous reports, no evidence was found to support the robustness of network-based features and we argue some of the robustness may be due to the inherent bias associated with node degree in the network. In addition, different types of network features seem to cover different pathways and are complementary to each other. Consequently, an ensemble classifier combining different network features was proposed and was found to significantly outperform classifiers based on gene-based feature or any single type of network-based features. Conclusions Network-based features and their combination show promise for improving the prediction of breast cancer metastasis but may require a large amount of training data. Robustness claim of network-based features needs to be re-examined with network node degree and other confounding factors in consideration.
Collapse
Affiliation(s)
- Nahim Adnan
- Department of Computer Science, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Zhijie Liu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA
| | - Tim H M Huang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA
| | - Jianhua Ruan
- Department of Computer Science, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA. .,Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA.
| |
Collapse
|
61
|
Safari-Alighiarloo N, Taghizadeh M, Mohammad Tabatabaei S, Namaki S, Rezaei-Tavirani M. Identification of common key genes and pathways between type 1 diabetes and multiple sclerosis using transcriptome and interactome analysis. Endocrine 2020; 68:81-92. [PMID: 31912409 DOI: 10.1007/s12020-019-02181-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/27/2019] [Indexed: 01/24/2023]
Abstract
PURPOSE Type 1 diabetes (T1D) and multiple sclerosis (MS) are classified as T cell-mediated autoimmune diseases. Although convergent evidence proposed common genetic architecture for autoimmune diseases, it remains a challenge to identify them. This study aimed to determine common gene signature and pathways in T1D and MS via systems biology approach. METHODS Gene expression profiles of peripheral blood mononuclear cells (PBMCs) and pancreatic-β cells in T1D as well as PBMCs and cerebrospinal fluid (CSF) in MS were analyzed in our previous published data, and differential expressed genes were integrated with protein-protein interactions data to construct Query-Query PPI (QQPPI) networks. In this study, QQPPI networks were further analyzed to investigate more central genes, functional modules and complexes shared in T1D and MS progression. Lastly, the interaction of common genes with drugs was also explored. RESULTS Several cytokines such as IL-23A, IL-32, IL-34, and IL-37 tend to be differentially expressed in both diseases. In addition, PSMA1, MYC, SRPK1, YBX1, HNRNPM, NF-κB2, IKBKE, RAC1, FN1, ARRB2, ESR1, HSP90AB1, and PPP1CA were common high central genes in QQPPI networks corresponding to each disease. Proteasome, spliceosome, immune responses, apoptosis, cellular communication/signaling transduction mechanism, interaction with environment, and activity of intercellular mediators were shared biological processes in T1D and MS. Finally, azathioprine, melatonin, resveratrol, and geldanamycin identified as prioritized drugs for the treatment of patients with T1D and MS. CONCLUSIONS This study represented novel key genes and pathways shared between T1D and MS, which may facilitate the identification of potential therapeutic targets in these diseases.
Collapse
Affiliation(s)
- Nahid Safari-Alighiarloo
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghizadeh
- Bioinformatics Department, Institute of Biochemistry and Biophysics, Tehran University, Tehran, Iran
| | - Seyyed Mohammad Tabatabaei
- Medical Informatics Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Namaki
- Immunology Department, Faculty of Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
62
|
Wang Y, Huang T, Li Y, Sha X. The self-organization model reveals systematic characteristics of aging. Theor Biol Med Model 2020; 17:4. [PMID: 32197622 PMCID: PMC7082995 DOI: 10.1186/s12976-020-00120-z] [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: 10/18/2019] [Accepted: 02/25/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aging is a fundamental biological process, where key bio-markers interact with each other and synergistically regulate the aging process. Thus aging dysfunction will induce many disorders. Finding aging markers and re-constructing networks based on multi-omics data (i.e. methylation, transcriptional and so on) are informative to study the aging process. However, optimizing the model to predict aging have not been performed systemically, although it is critical to identify potential molecular mechanism of aging related diseases. METHODS This paper aims to model the aging self-organization system using a series of supervised learning methods, and study complex molecular mechanisms of aging at system level: i.e. optimizing the aging network; summarizing interactions between aging markers; accumulating patterns of aging markers within module; finding order-parameters in the aging self-organization system. RESULTS In this work, the normal aging process is modeled based on multi-omics profiles across different tissues. In addition, the computational pipeline aims to model aging self-organizing systems and study the relationship between aging and related diseases (i.e. cancers), thus provide useful indicators of aging related diseases and could help to improve prediction abilities of diagnostics. CONCLUSIONS The aging process could be studied thoroughly by modelling the self-organization system, where key functions and the crosstalk between aging and cancers were identified.
Collapse
Affiliation(s)
- Yin Wang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110012, Liaoning Province, China.,Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Yixue Li
- 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, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, 200433, China. .,Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China.
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110012, Liaoning Province, China.
| |
Collapse
|
63
|
Chang JW, Ding Y, Tahir Ul Qamar M, Shen Y, Gao J, Chen LL. A deep learning model based on sparse auto-encoder for prioritizing cancer-related genes and drug target combinations. Carcinogenesis 2020; 40:624-632. [PMID: 30944926 DOI: 10.1093/carcin/bgz044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 01/06/2019] [Accepted: 03/10/2019] [Indexed: 12/21/2022] Open
Abstract
Prioritization of cancer-related genes from gene expression profiles and proteomic data is vital to improve the targeted therapies research. Although computational approaches have been complementing high-throughput biological experiments on the understanding of human diseases, it still remains a big challenge to accurately discover cancer-related proteins/genes via automatic learning from large-scale protein/gene expression data and protein-protein interaction data. Most of the existing methods are based on network construction combined with gene expression profiles, which ignore the diversity between normal samples and disease cell lines. In this study, we introduced a deep learning model based on a sparse auto-encoder to learn the specific characteristics of protein interactions in cancer cell lines integrated with protein expression data. The model showed learning ability to identify cancer-related proteins/genes from the input of different protein expression profiles by extracting the characteristics of protein interaction information, which could also predict cancer-related protein combinations. Comparing with other reported methods including differential expression and network-based methods, our model got the highest area under the curve value (>0.8) in predicting cancer-related genes. Our study prioritized ~500 high-confidence cancer-related genes; among these genes, 211 already known cancer drug targets were found, which supported the accuracy of our method. The above results indicated that the proposed auto-encoder model could computationally prioritize candidate proteins/genes involved in cancer and improve the targeted therapies research.
Collapse
Affiliation(s)
- Ji-Wei Chang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, P. R. China
| | - Yuduan Ding
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, P. R. China
| | - Muhammad Tahir Ul Qamar
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, P. R. China
| | - Yin Shen
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
| | - Junxiang Gao
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
| | - Ling-Ling Chen
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, P. R. China
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, P. R. China
| |
Collapse
|
64
|
Zhang J, Yan S, Jiang C, Ji Z, Wang C, Tian W. Network Properties of Cancer Prognostic Gene Signatures in the Human Protein Interactome. Genes (Basel) 2020; 11:genes11030247. [PMID: 32111006 PMCID: PMC7140842 DOI: 10.3390/genes11030247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 11/16/2022] Open
Abstract
Prognostic gene signatures are critical in cancer prognosis assessments and their pinpoint treatments. However, their network properties remain unclear. Here, we obtained nine prognostic gene sets including 1439 prognostic genes of different cancers from related publications. Four network centralities were used to examine the network properties of prognostic genes (PG) compared with other gene sets based on the Human Protein Reference Database (HPRD) and String networks. We also proposed three novel network measures for further investigating the network properties of prognostic gene sets (PGS) besides clustering coefficient. The results showed that PG did not occupy key positions in the human protein interaction network and were more similar to essential genes rather than cancer genes. However, PGS had significantly smaller intra-set distance (IAD) and inter-set distance (IED) in comparison with random sets (p-value < 0.001). Moreover, we also found that PGS tended to be distributed within network modules rather than between modules (p-value < 0.01), and the functional intersection of the modules enriched with PGS was closely related to cancer development and progression. Our research reveals the common network properties of cancer prognostic gene signatures in the human protein interactome. We argue that these are biologically meaningful and useful for understanding their molecular mechanism.
Collapse
Affiliation(s)
- Jifeng Zhang
- School of Biological Engineering, Huainan Normal University, Huainan 232001, China (C.J.); (C.W.)
- School of Life Science, Institute of Biostatistics, Fudan University, Shanghai 2004333, China
- Correspondence: (J.Z.); (W.T.); Tel.: +86-181-3013-7151 (J.Z.); +86-21-3124-6723 (W.T.)
| | - Shoubao Yan
- School of Biological Engineering, Huainan Normal University, Huainan 232001, China (C.J.); (C.W.)
| | - Cheng Jiang
- School of Biological Engineering, Huainan Normal University, Huainan 232001, China (C.J.); (C.W.)
| | - Zhicheng Ji
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Chenrun Wang
- School of Biological Engineering, Huainan Normal University, Huainan 232001, China (C.J.); (C.W.)
| | - Weidong Tian
- School of Life Science, Institute of Biostatistics, Fudan University, Shanghai 2004333, China
- Correspondence: (J.Z.); (W.T.); Tel.: +86-181-3013-7151 (J.Z.); +86-21-3124-6723 (W.T.)
| |
Collapse
|
65
|
Dey L, Mukhopadhyay A. Biclustering-based association rule mining approach for predicting cancer-associated protein interactions. IET Syst Biol 2020; 13:234-242. [PMID: 31538957 DOI: 10.1049/iet-syb.2019.0045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Protein-protein interactions (PPIs) have been widely used to understand different biological processes and cellular functions associated with several diseases like cancer. Although some cancer-related protein interaction databases are available, lack of experimental data and conflicting PPI data among different available databases have slowed down the cancer research. Therefore, in this study, the authors have focused on various proteins that are directly related to different types of cancer disease. They have prepared a PPI database between cancer-associated proteins with the rest of the human proteins. They have also incorporated the annotation type and direction of each interaction. Subsequently, a biclustering-based association rule mining algorithm is applied to predict new interactions with type and direction. This study shows the prediction power of association rule mining algorithm over the traditional classifier model without choosing a negative data set. The time complexity of the biclustering-based association rule mining is also analysed and compared to traditional association rule mining. The authors are able to discover 38 new PPIs which are not present in the cancer database. The biological relevance of these newly predicted interactions is analysed by published literature. Recognition of such interactions may accelerate a way of developing new drugs to prevent different cancer-related diseases.
Collapse
Affiliation(s)
- Lopamudra Dey
- Department of Computer Science and Engineering, Heritage Institute of Technology, 994 Madurdaha, Kolkata 700 107, West Bengal, India.
| | - Anirban Mukhopadhyay
- Department of Computer Science and Engineering, University of Kalyani, Nadia, Kalyani 741235, West Bengal, India
| |
Collapse
|
66
|
Wu D, Huo M, Chen X, Zhang Y, Qiao Y. Mechanism of tanshinones and phenolic acids from Danshen in the treatment of coronary heart disease based on co-expression network. BMC Complement Med Ther 2020; 20:28. [PMID: 32020855 PMCID: PMC7076864 DOI: 10.1186/s12906-019-2712-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023] Open
Abstract
Background The tanshinones and phenolic acids in Salvia miltiorrhiza (also named Danshen) have been confirmed for the treatment of coronary heart disease (CHD), but the action mechanisms remain elusive. Methods In the current study, the co-expression protein interaction network (Ce-PIN) was used to illustrate the differences between the tanshinones and phenolic acids of Danshen in the treatment of CHD. By integrating the gene expression profile data and protein-protein interactions (PPIs) data, the Ce-PINs of tanshinones and phenolic acids were constructed. Then, the Ce-PINs were analyzed by gene ontology enrichment analyzed based on the optimal algorithm. Results It turned out that Danshen is able to treat CHD by regulating the blood circulation, immune response and lipid metabolism. However, phenolic acids may regulate the blood circulation by Extracellular calcium-sensing receptor (CaSR), Endothelin-1 receptor (EDNRA), Endothelin-1 receptor (EDNRB), Kininogen-1 (KNG1), tanshinones may regulate the blood circulation by Guanylate cyclase soluble subunit alpha-1 (GUCY1A3) and Guanylate cyclase soluble subunit beta-1 (GUCY1B3). In addition, both the phenolic acids and tanshinones may regulate the immune response or inflammation by T-cell surface glycoprotein CD4 (CD4), Receptor-type tyrosine-protein phosphatase C (PTPRC). Conclusion Through the same targets of the same biological process and different targets of the same biological process, the tanshinones and phenolic acids synergistically treat coronary heart disease.
Collapse
Affiliation(s)
- Dongxue Wu
- Beijing University of Chinese Medicine, State Administration of Traditional Chinese Medicine, Research Center of TCM-Information Engineering, Beijing, 100102, China
| | - Mengqi Huo
- Beijing University of Chinese Medicine, State Administration of Traditional Chinese Medicine, Research Center of TCM-Information Engineering, Beijing, 100102, China
| | - Xi Chen
- Beijing University of Chinese Medicine, State Administration of Traditional Chinese Medicine, Research Center of TCM-Information Engineering, Beijing, 100102, China
| | - Yanling Zhang
- Beijing University of Chinese Medicine, State Administration of Traditional Chinese Medicine, Research Center of TCM-Information Engineering, Beijing, 100102, China.
| | - Yanjiang Qiao
- Beijing University of Chinese Medicine, State Administration of Traditional Chinese Medicine, Research Center of TCM-Information Engineering, Beijing, 100102, China.
| |
Collapse
|
67
|
Lin Q, Lin Y, Yu Q, Ma X. Clustering of Cancer Attributed Networks via Integration of Graph Embedding and Matrix Factorization. IEEE ACCESS 2020. [DOI: 10.1109/access.2020.3034623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
68
|
Lee KH, Xue L, Hunter DR. Model-based clustering of time-evolving networks through temporal exponential-family random graph models. J MULTIVARIATE ANAL 2020; 175:104540. [PMID: 32863458 PMCID: PMC7448400 DOI: 10.1016/j.jmva.2019.104540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dynamic networks are a general language for describing time-evolving complex systems, and discrete time network models provide an emerging statistical technique for various applications. It is a fundamental research question to detect a set of nodes sharing similar connectivity patterns in time-evolving networks. Our work is primarily motivated by detecting groups based on interesting features of the time-evolving networks (e.g., stability). In this work, we propose a model-based clustering framework for time-evolving networks based on discrete time exponential-family random graph models, which simultaneously allows both modeling and detecting group structure. To choose the number of groups, we use the conditional likelihood to construct an effective model selection criterion. Furthermore, we propose an efficient variational expectation-maximization (EM) algorithm to find approximate maximum likelihood estimates of network parameters and mixing proportions. The power of our method is demonstrated in simulation studies and empirical applications to international trade networks and the collaboration networks of a large research university.
Collapse
Affiliation(s)
- Kevin H. Lee
- Department of Statistics, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Lingzhou Xue
- Department of Statistics, The Pennsylvania State University, University Park, PA 16802, USA
| | - David R. Hunter
- Department of Statistics, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
69
|
Ghazanfar S, Strbenac D, Ormerod JT, Yang JYH, Patrick E. DCARS: differential correlation across ranked samples. Bioinformatics 2019; 35:823-829. [PMID: 30102408 DOI: 10.1093/bioinformatics/bty698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/19/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
MOTIVATION Genes act as a system and not in isolation. Thus, it is important to consider coordinated changes of gene expression rather than single genes when investigating biological phenomena such as the aetiology of cancer. We have developed an approach for quantifying how changes in the association between pairs of genes may inform the outcome of interest called Differential Correlation across Ranked Samples (DCARS). Modelling gene correlation across a continuous sample ranking does not require the dichotomisation of samples into two distinct classes and can identify differences in gene correlation across early, mid or late stages of the outcome of interest. RESULTS When we evaluated DCARS against the typical Fisher Z-transformation test for differential correlation, as well as a typical approach testing for interaction within a linear model, on real TCGA data, DCARS significantly ranked gene pairs containing known cancer genes more highly across several cancers. Similar results are found with our simulation study. DCARS was applied to 13 cancers datasets in TCGA, revealing several distinct relationships for which survival ranking was found to be associated with a change in correlation between genes. Furthermore, we demonstrated that DCARS can be used in conjunction with network analysis techniques to extract biological meaning from multi-layered and complex data. AVAILABILITY AND IMPLEMENTATION DCARS R package and sample data are available at https://github.com/shazanfar/DCARS. Publicly available data from The Cancer Genome Atlas (TCGA) was used using the TCGABiolinks R package. Supplementary Files and DCARS R package is available at https://github.com/shazanfar/DCARS. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Shila Ghazanfar
- The Judith and David Coffey Life Lab, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Dario Strbenac
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - John T Ormerod
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia.,ARC Centre of Excellence for Mathematical and Statistical Frontiers (ACEMS), Richard Berry Building, The University of Melbourne, Melbourne, Parkville, VIC, Australia
| | - Jean Y H Yang
- The Judith and David Coffey Life Lab, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Ellis Patrick
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia.,Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
70
|
Lu J, Lu Y, Ding Y, Xiao Q, Liu L, Cai Q, Kong Y, Bai Y, Yu T. DNLC: differential network local consistency analysis. BMC Bioinformatics 2019; 20:489. [PMID: 31874600 PMCID: PMC6929334 DOI: 10.1186/s12859-019-3046-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 08/21/2019] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The biological network is highly dynamic. Functional relations between genes can be activated or deactivated depending on the biological conditions. On the genome-scale network, subnetworks that gain or lose local expression consistency may shed light on the regulatory mechanisms related to the changing biological conditions, such as disease status or tissue developmental stages. RESULTS In this study, we develop a new method to select genes and modules on the existing biological network, in which local expression consistency changes significantly between clinical conditions. The method is called DNLC: Differential Network Local Consistency. In simulations, our algorithm detected artificially created local consistency changes effectively. We applied the method on two publicly available datasets, and the method detected novel genes and network modules that were biologically plausible. CONCLUSIONS The new method is effective in finding modules in which the gene expression consistency change between clinical conditions. It is a useful tool that complements traditional differential expression analyses to make discoveries from gene expression data. The R package is available at https://cran.r-project.org/web/packages/DNLC.
Collapse
Affiliation(s)
- Jianwei Lu
- School of Software Engineering, Tongji University, Shanghai, China
- Institute of Advanced Translational Medicine, Tongji University, Shanghai, China
| | - Yao Lu
- School of Software Engineering, Tongji University, Shanghai, China
| | - Yusheng Ding
- School of Software Engineering, Tongji University, Shanghai, China
| | - Qingyang Xiao
- Department of Environmental Health, Emory University, Atlanta, GA USA
| | - Linqing Liu
- School of Software Engineering, Tongji University, Shanghai, China
| | - Qingpo Cai
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA USA
| | - Yunchuan Kong
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA USA
| | - Yun Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine, Georgia Campus, Suwanee, GA USA
| | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA USA
| |
Collapse
|
71
|
M B, P C. Comparative analysis of differential proteome-wide protein-protein interaction network of Methanobrevibacter ruminantium M1. Biochem Biophys Rep 2019; 20:100698. [PMID: 31763465 PMCID: PMC6859225 DOI: 10.1016/j.bbrep.2019.100698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 11/22/2022] Open
Abstract
A proteome-wide protein-protein interaction (PPI) network of Methanobrevibacter ruminantium M1 (MRU), a predominant rumen methanogen, was constructed from its metabolic genes using a gene neighborhood algorithm and then compared with closely related rumen methanogens Using proteome-wide PPI approach, we constructed network encompassed 2194 edges and 637 nodes interacting with 634 genes. Network quality and robustness of functional modules were assessed with gene ontology terms. A structure-function-metabolism mapping for each protein has been carried out with efforts to extract experimental PPI concomitant information from the literature. The results of our study revealed that some topological properties of its network were robust for sharing homologous protein interactions across heterotrophic and hydrogenotrophic methanogens. MRU proteome has shown to establish many PPI sub-networks for associated metabolic subsystems required to survive in the rumen environment. MRU genome found to share interacting proteins from its PPI network involved in specific metabolic subsystems distinct to heterotrophic and hydrogenotrophic methanogens. Across these proteomes, the interacting proteins from differential PPI networks were shared in common for the biosynthesis of amino acids, nucleosides, and nucleotides and energy metabolism in which more fractions of protein pairs shared with Methanosarcina acetivorans. Our comparative study expedites our knowledge to understand a complex proteome network associated with typical metabolic subsystems of MRU and to improve its genome-scale reconstruction in the future.
Collapse
Affiliation(s)
| | - Chellapandi P
- Molecular Systems Engineering Lab, Department of Bioinformatics, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620 024, Tamil Nadu, India
| |
Collapse
|
72
|
Gao X, Cai Y, Wang Z, He W, Cao S, Xu R, Chen H. Estrogen receptors promote NSCLC progression by modulating the membrane receptor signaling network: a systems biology perspective. J Transl Med 2019; 17:308. [PMID: 31511014 PMCID: PMC6737693 DOI: 10.1186/s12967-019-2056-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background Estrogen receptors (ERs) are thought to play an important role in non-small cell lung cancer (NSCLC). However, the effect of ERs in NSCLC is still controversial and needs further investigation. A new consideration is that ERs may affect NSCLC progression through complicated molecular signaling networks rather than individual targets. Therefore, this study aims to explore the effect of ERs in NSCLC from the perspective of cancer systems biology. Methods The gene expression profile of NSCLC samples in TCGA dataset was analyzed by bioinformatics method. Variations of cell behaviors and protein expression were detected in vitro. The kinetic process of molecular signaling network was illustrated by a systemic computational model. At last, immunohistochemical (IHC) and survival analysis was applied to evaluate the clinical relevance and prognostic effect of key receptors in NSCLC. Results Bioinformatics analysis revealed that ERs might affect many cancer-related molecular events and pathways in NSCLC, particularly membrane receptor activation and signal transduction, which might ultimately lead to changes in cell behaviors. Experimental results confirmed that ERs could regulate cell behaviors including cell proliferation, apoptosis, invasion and migration; ERs also regulated the expression or activation of key members in membrane receptor signaling pathways such as epidermal growth factor receptor (EGFR), Notch1 and Glycogen synthase kinase-3β/β-Catenin (GSK3β/β-Catenin) pathways. Modeling results illustrated that the promotive effect of ERs in NSCLC was implemented by modulating the signaling network composed of EGFR, Notch1 and GSK3β/β-Catenin pathways; ERs maintained and enhanced the output of oncogenic signals by adding redundant and positive-feedback paths into the network. IHC results echoed that high expression of ERs, EGFR and Notch1 had a synergistic effect on poor prognosis of advanced NSCLC. Conclusions This study indicated that ERs were likely to promote NSCLC progression by modulating the integrated membrane receptor signaling network composed of EGFR, Notch1 and GSK3β/β-Catenin pathways and then affecting tumor cell behaviors. It also complemented the molecular mechanisms underlying the progression of NSCLC and provided new opportunities for optimizing therapeutic scheme of NSCLC.
Collapse
Affiliation(s)
- Xiujuan Gao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China
| | - Yue Cai
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China
| | - Zhuo Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China
| | - Wenjuan He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China
| | - Sisi Cao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China
| | - Rong Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China
| | - Hui Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13, HangKong Road, Wuhan, 430030, Hubei, China. .,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
73
|
Prathiviraj R, Berchmans S, Chellapandi P. Analysis of modularity in proteome-wide protein interaction networks of Methanothermobacter thermautotrophicus strain ΔH and metal-loving bacteria. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s42485-019-00019-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
74
|
Tan A, Huang H, Zhang P, Li S. Network-based cancer precision medicine: A new emerging paradigm. Cancer Lett 2019; 458:39-45. [DOI: 10.1016/j.canlet.2019.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
75
|
Couch D, Yu Z, Nam JH, Allen C, Ramos PS, da Silveira WA, Hunt KJ, Hazard ES, Hardiman G, Lawson A, Chung D. GAIL: An interactive webserver for inference and dynamic visualization of gene-gene associations based on gene ontology guided mining of biomedical literature. PLoS One 2019; 14:e0219195. [PMID: 31260503 PMCID: PMC6602258 DOI: 10.1371/journal.pone.0219195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/18/2019] [Indexed: 01/08/2023] Open
Abstract
In systems biology, inference of functional associations among genes is compelling because the construction of functional association networks facilitates biomarker discovery. Specifically, such gene associations in human can help identify putative biomarkers that can be used as diagnostic tools in treating patients. Although biomedical literature is considered a valuable data source for this task, currently only a limited number of webservers are available for mining gene-gene associations from the vast amount of biomedical literature using text mining techniques. Moreover, these webservers often have limited coverage of biomedical literature and also lack efficient and user-friendly tools to interpret and visualize mined relationships among genes. To address these limitations, we developed GAIL (Gene-gene Association Inference based on biomedical Literature), an interactive webserver that infers human gene-gene associations from Gene Ontology (GO) guided biomedical literature mining and provides dynamic visualization of the resulting association networks and various gene set enrichment analysis tools. We evaluate the utility and performance of GAIL with applications to gene signatures associated with systemic lupus erythematosus and breast cancer. Results show that GAIL allows effective interrogation and visualization of gene-gene networks and their subnetworks, which facilitates biological understanding of gene-gene associations. GAIL is available at http://chunglab.io/GAIL/.
Collapse
Affiliation(s)
- Daniel Couch
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Zhenning Yu
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Jin Hyun Nam
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Carter Allen
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Paula S. Ramos
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Willian A. da Silveira
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States of America
- Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Kelly J. Hunt
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Edward S. Hazard
- Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Gary Hardiman
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States of America
- Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Andrew Lawson
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| | - Dongjun Chung
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States of America
| |
Collapse
|
76
|
Zhao X, Xu M, Cai Z, Yuan W, Cui W, Li MD. Identification of LIFR, PIK3R1, and MMP12 as Novel Prognostic Signatures in Gallbladder Cancer Using Network-Based Module Analysis. Front Oncol 2019; 9:325. [PMID: 31119098 PMCID: PMC6504688 DOI: 10.3389/fonc.2019.00325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/10/2019] [Indexed: 01/17/2023] Open
Abstract
Background: Gallbladder cancer (GBC) is a rare and aggressive malignancy of the biliary tract with a dismal survival rate. Effective biomarkers and therapeutic targets are urgently needed. Methods: We analyzed gene expression profiles of GBC to identify differentially expressed genes (DEGs) and then used these DEGs to identify functional module biomarkers based on protein functional interaction (FI) networks. We further evaluated the module-gene protein expression and clinical significance with immunohistochemistry staining (IHC) in a tissue microarray (TMA) from 80 GBC samples. Results: Five functional modules were identified. Module 0 included classical cancer signaling pathways, such as Ras and PI3K-Akt; and modules 1–4 included genes associated with muscle cells, fibrinogen, extracellular matrix, and integrins, respectively. We validated the expression of LIFR, PIK3R1, and MMP12, which were hubs or functional nodes in modules. Compared with paired peritumoural tissues, we found that the expression of LIFR (P = 0.002) and PIK3R1 (P = 0.046) proteins were significantly downregulated, and MMP12 (P = 0.006) was significantly upregulated. Further prognostic analysis showed that patients with low expression of LIFR had shorter overall survival than those with high expression (log-rank test P = 0.028), the same trend as for PIK3R1 (P = 0.053) and MMP12 (P = 0.006). Multivariate analysis indicated that expression of MMP12 protein (hazard ratio [HR] = 0.429; 95% confidence interval [CI] 0.198, 0.930; P = 0.032) was one of the significant independent prognostic factors for overall survival. Conclusions: We found a highly reliable FI network, which revealed LIFR, PIK3R1, and MMP12 as novel prognostic biomarker candidates for GBC. These findings could accelerate biomarker discovery and therapeutic development in this cancer.
Collapse
Affiliation(s)
- Xinyi Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengxiang Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen Cai
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenji Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenyan Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.,Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, United States
| |
Collapse
|
77
|
George G, Valiya Parambath S, Lokappa SB, Varkey J. Construction of Parkinson's disease marker-based weighted protein-protein interaction network for prioritization of co-expressed genes. Gene 2019; 697:67-77. [DOI: 10.1016/j.gene.2019.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/12/2019] [Accepted: 02/01/2019] [Indexed: 12/31/2022]
|
78
|
Sonawane AR, Weiss ST, Glass K, Sharma A. Network Medicine in the Age of Biomedical Big Data. Front Genet 2019; 10:294. [PMID: 31031797 PMCID: PMC6470635 DOI: 10.3389/fgene.2019.00294] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Network medicine is an emerging area of research dealing with molecular and genetic interactions, network biomarkers of disease, and therapeutic target discovery. Large-scale biomedical data generation offers a unique opportunity to assess the effect and impact of cellular heterogeneity and environmental perturbations on the observed phenotype. Marrying the two, network medicine with biomedical data provides a framework to build meaningful models and extract impactful results at a network level. In this review, we survey existing network types and biomedical data sources. More importantly, we delve into ways in which the network medicine approach, aided by phenotype-specific biomedical data, can be gainfully applied. We provide three paradigms, mainly dealing with three major biological network archetypes: protein-protein interaction, expression-based, and gene regulatory networks. For each of these paradigms, we discuss a broad overview of philosophies under which various network methods work. We also provide a few examples in each paradigm as a test case of its successful application. Finally, we delineate several opportunities and challenges in the field of network medicine. We hope this review provides a lexicon for researchers from biological sciences and network theory to come on the same page to work on research areas that require interdisciplinary expertise. Taken together, the understanding gained from combining biomedical data with networks can be useful for characterizing disease etiologies and identifying therapeutic targets, which, in turn, will lead to better preventive medicine with translational impact on personalized healthcare.
Collapse
Affiliation(s)
- Abhijeet R. Sonawane
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Amitabh Sharma
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
79
|
Abstract
Reliable identification of molecular biomarkers is essential for accurate patient stratification. While state-of-the-art machine learning approaches for sample classification continue to push boundaries in terms of performance, most of these methods are not able to integrate different data types and lack generalization power, limiting their application in a clinical setting. Furthermore, many methods behave as black boxes, and we have very little understanding about the mechanisms that lead to the prediction. While opaqueness concerning machine behavior might not be a problem in deterministic domains, in health care, providing explanations about the molecular factors and phenotypes that are driving the classification is crucial to build trust in the performance of the predictive system. We propose Pathway-Induced Multiple Kernel Learning (PIMKL), a methodology to reliably classify samples that can also help gain insights into the molecular mechanisms that underlie the classification. PIMKL exploits prior knowledge in the form of a molecular interaction network and annotated gene sets, by optimizing a mixture of pathway-induced kernels using a Multiple Kernel Learning (MKL) algorithm, an approach that has demonstrated excellent performance in different machine learning applications. After optimizing the combination of kernels to predict a specific phenotype, the model provides a stable molecular signature that can be interpreted in the light of the ingested prior knowledge and that can be used in transfer learning tasks. The reliable classification of biomedical samples to predict phenotypic differences requires not only robust methods, but also interpretable approaches that can explain the reasons behind a prediction. A team led by María Rodríguez Martínez at IBM Research - Zürich has developed PIMKL, a methodology that exploits prior knowledge and enables the integration of multiple types of data with varying predictive power. Even when noisy datasets are simultaneously analyzed, PIMKL is able to discard uninformative data and achieve strong prediction power. Importantly, PIMKL produces a molecular signature that enables the interpretation of the results in terms of known biological functions. This signature can be transferred to other cohorts without loss of performance, demonstrating surprising robustness across cohorts. Interpretable algorithms can effectively help gain insights about disease mechanisms and build trust in a model.
Collapse
|
80
|
Zhou XH, Chu XY, Xue G, Xiong JH, Zhang HY. Identifying cancer prognostic modules by module network analysis. BMC Bioinformatics 2019; 20:85. [PMID: 30777030 PMCID: PMC6380061 DOI: 10.1186/s12859-019-2674-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/08/2019] [Indexed: 02/08/2023] Open
Abstract
Background The identification of prognostic genes that can distinguish the prognostic risks of cancer patients remains a significant challenge. Previous works have proven that functional gene sets were more reliable for this task than the gene signature. However, few works have considered the cross-talk among functional gene sets, which may result in neglecting important prognostic gene sets for cancer. Results Here, we proposed a new method that considers both the interactions among modules and the prognostic correlation of the modules to identify prognostic modules in cancers. First, dense sub-networks in the gene co-expression network of cancer patients were detected. Second, cross-talk between every two modules was identified by a permutation test, thus generating the module network. Third, the prognostic correlation of each module was evaluated by the resampling method. Then, the GeneRank algorithm, which takes the module network and the prognostic correlations of all the modules as input, was applied to prioritize the prognostic modules. Finally, the selected modules were validated by survival analysis in various data sets. Our method was applied in three kinds of cancers, and the results show that our method succeeded in identifying prognostic modules in all the three cancers. In addition, our method outperformed state-of-the-art methods. Furthermore, the selected modules were significantly enriched with known cancer-related genes and drug targets of cancer, which may indicate that the genes involved in the modules may be drug targets for therapy. Conclusions We proposed a useful method to identify key modules in cancer prognosis and our prognostic genes may be good candidates for drug targets. Electronic supplementary material The online version of this article (10.1186/s12859-019-2674-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xin-Yi Chu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Gang Xue
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiang-Hui Xiong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, People's Republic of China.,Lab of Epigenetics and Health Tracking Technology, Space Institute of Southern China, Shenzhen, People's Republic of China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
81
|
Abstract
In recent years, open-source software (OSS) development has grown, with many developers around the world working on different OSS projects. A variety of open-source software ecosystems have emerged, for instance, GitHub, StackOverflow, and SourceForge. One of the most typical social-programming and code-hosting sites, GitHub, has amassed numerous open-source-software projects and developers in the same virtual collaboration platform. Since GitHub itself is a large open-source community, it hosts a collection of software projects that are developed together and coevolve. The great challenge here is how to identify the relationship between these projects, i.e., project relevance. Software-ecosystem identification is the basis of other studies in the ecosystem. Therefore, how to extract useful information in GitHub and identify software ecosystems is particularly important, and it is also a research area in symmetry. In this paper, a Topic-based Project Knowledge Metrics Framework (TPKMF) is proposed. By collecting the multisource dataset of an open-source ecosystem, project-relevance analysis of the open-source software is carried out on the basis of software-ecosystem identification. Then, we used our Spectral Clustering algorithm based on Core Project (CP-SC) to identify software-ecosystem projects and further identify software ecosystems. We verified that most software ecosystems usually contain a core software project, and most other projects are associated with it. Furthermore, we analyzed the characteristics of the ecosystem, and we also found that interactive information has greater impact on project relevance. Finally, we summarize the Topic-based Project Knowledge Metrics Framework.
Collapse
|
82
|
Allahyar A, Ubels J, de Ridder J. A data-driven interactome of synergistic genes improves network-based cancer outcome prediction. PLoS Comput Biol 2019; 15:e1006657. [PMID: 30726216 PMCID: PMC6380593 DOI: 10.1371/journal.pcbi.1006657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 02/19/2019] [Accepted: 11/20/2018] [Indexed: 12/13/2022] Open
Abstract
Robustly predicting outcome for cancer patients from gene expression is an important challenge on the road to better personalized treatment. Network-based outcome predictors (NOPs), which considers the cellular wiring diagram in the classification, hold much promise to improve performance, stability and interpretability of identified marker genes. Problematically, reports on the efficacy of NOPs are conflicting and for instance suggest that utilizing random networks performs on par to networks that describe biologically relevant interactions. In this paper we turn the prediction problem around: instead of using a given biological network in the NOP, we aim to identify the network of genes that truly improves outcome prediction. To this end, we propose SyNet, a gene network constructed ab initio from synergistic gene pairs derived from survival-labelled gene expression data. To obtain SyNet, we evaluate synergy for all 69 million pairwise combinations of genes resulting in a network that is specific to the dataset and phenotype under study and can be used to in a NOP model. We evaluated SyNet and 11 other networks on a compendium dataset of >4000 survival-labelled breast cancer samples. For this purpose, we used cross-study validation which more closely emulates real world application of these outcome predictors. We find that SyNet is the only network that truly improves performance, stability and interpretability in several existing NOPs. We show that SyNet overlaps significantly with existing gene networks, and can be confidently predicted (~85% AUC) from graph-topological descriptions of these networks, in particular the breast tissue-specific network. Due to its data-driven nature, SyNet is not biased to well-studied genes and thus facilitates post-hoc interpretation. We find that SyNet is highly enriched for known breast cancer genes and genes related to e.g. histological grade and tamoxifen resistance, suggestive of a role in determining breast cancer outcome.
Collapse
Affiliation(s)
- Amin Allahyar
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Delft Bioinformatics Lab, Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, The Netherlands
| | - Joske Ubels
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Skyline DX, Rotterdam
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam
| | - Jeroen de Ridder
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
83
|
Dutta P, Saha S, Gulati S. Graph-Based Hub Gene Selection Technique Using Protein Interaction Information: Application to Sample Classification. IEEE J Biomed Health Inform 2019; 23:2670-2676. [PMID: 30676987 DOI: 10.1109/jbhi.2019.2894374] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Classification of samples of gene expression profile plays a significant role in prediction and diagnosis of diseases. In the task of sample classification, a robust feature selection algorithm is very much essential to identify the important genes from the high dimensional gene expression data. This paper explores the information of protein-protein interaction with a graph mining technique for finding a proper subset of features (genes), which further takes part in sample classification. Here, our contribution for feature selection is three-fold: first, all the genes are grouped into different clusters based on the integrated information of the gene expression values and their protein interactions using a multi-objective optimization based clustering approach. Second, the confidence scores of the protein interactions are incorporated in a popular graph mining algorithm namely Goldberg algorithm to find out the relevant features. These features are the topologically and functionally significant genes, named as hub genes. Finally, these hub genes are identified varying the degrees of the nodes, and those are utilized for the sample classification task. Different machine learning classifiers are exploited for this purpose, and the classification performance is measured with respect to various performance metrics namely accuracy, sensitivity, specificity, precision, F-measure, and Mathews coefficient correlation. Comparative analysis with respect to two baselines and several existing approaches proves the efficiency of the proposed approach. Furthermore, the robustness of the identified hub-gene modules is endorsed using some strong biological significance analysis.
Collapse
|
84
|
Li M, He Q, Ma J, He F, Zhu Y, Chang C, Chen T. PPICurator: A Tool for Extracting Comprehensive Protein-Protein Interaction Information. Proteomics 2019; 19:e1800291. [PMID: 30521143 DOI: 10.1002/pmic.201800291] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/12/2018] [Indexed: 11/07/2022]
Abstract
Protein-protein interaction extraction through biological literature curation is widely employed for proteome analysis. There is a strong need for a tool that can assist researchers in extracting comprehensive PPI information through literature curation, which is critical in research on protein, for example, construction of protein interaction network, identification of protein signaling pathway, and discovery of meaningful protein interaction. However, most of current tools can only extract PPI relations. None of them are capable of extracting other important PPI information, such as interaction directions, effects, and functional annotations. To address these issues, this paper proposes PPICurator, a novel tool for extracting comprehensive PPI information with a variety of logic and syntax features based on a new support vector machine classifier. PPICurator provides a friendly web-based user interface. It is a platform that automates the extraction of comprehensive PPI information through literature, including PPI relations, as well as their confidential scores, interaction directions, effects, and functional annotations. Thus, PPICurator is more comprehensive than state-of-the-art tools. Moreover, it outperforms state-of-the-art tools in the accuracy of PPI relation extraction measured by F-score and recall on the widely used open datasets. PPICurator is available at https://ppicurator.hupo.org.cn.
Collapse
Affiliation(s)
- Mansheng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, 102206, P. R. China
| | - Qiang He
- School of Software and Electrical Engineering, Swinburne University of Technology, Melbourne, Victoria, 3122, Australia
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, 102206, P. R. China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, 102206, P. R. China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, 102206, P. R. China
| | - Cheng Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, 102206, P. R. China
| | - Tao Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, 102206, P. R. China
| |
Collapse
|
85
|
Capriotti E, Ozturk K, Carter H. Integrating molecular networks with genetic variant interpretation for precision medicine. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2018; 11:e1443. [PMID: 30548534 PMCID: PMC6450710 DOI: 10.1002/wsbm.1443] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/23/2018] [Accepted: 10/30/2018] [Indexed: 02/01/2023]
Abstract
More reliable and cheaper sequencing technologies have revealed the vast mutational landscapes characteristic of many phenotypes. The analysis of such genetic variants has led to successful identification of altered proteins underlying many Mendelian disorders. Nevertheless the simple one‐variant one‐phenotype model valid for many monogenic diseases does not capture the complexity of polygenic traits and disorders. Although experimental and computational approaches have improved detection of functionally deleterious variants and important interactions between gene products, the development of comprehensive models relating genotype and phenotypes remains a challenge in the field of genomic medicine. In this context, a new view of the pathologic state as significant perturbation of the network of interactions between biomolecules is crucial for the identification of biochemical pathways associated with complex phenotypes. Seminal studies in systems biology combined the analysis of genetic variation with protein–protein interaction networks to demonstrate that even as biological systems evolve to be robust to genetic variation, their topologies create disease vulnerabilities. More recent analyses model the impact of genetic variants as changes to the “wiring” of the interactome to better capture heterogeneity in genotype–phenotype relationships. These studies lay the foundation for using networks to predict variant effects at scale using machine‐learning or algorithmic approaches. A wealth of databases and resources for the annotation of genotype–phenotype relationships have been developed to support developments in this area. This overview describes how study of the molecular interactome has generated insights linking the organization of biological systems to disease mechanism, and how this information can enable precision medicine. This article is categorized under:
Translational, Genomic, and Systems Medicine > Translational Medicine Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models Analytical and Computational Methods > Computational Methods
Collapse
Affiliation(s)
- Emidio Capriotti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Kivilcim Ozturk
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, California
| | - Hannah Carter
- Department of Medicine and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
86
|
Interaction paths promote module integration and network-level robustness of spliceosome to cascading effects. Sci Rep 2018; 8:17441. [PMID: 30487551 PMCID: PMC6261937 DOI: 10.1038/s41598-018-35160-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 10/26/2018] [Indexed: 11/22/2022] Open
Abstract
The functionality of distinct types of protein networks depends on the patterns of protein-protein interactions. A problem to solve is understanding the fragility of protein networks to predict system malfunctioning due to mutations and other errors. Spectral graph theory provides tools to understand the structural and dynamical properties of a system based on the mathematical properties of matrices associated with the networks. We combined two of such tools to explore the fragility to cascading effects of the network describing protein interactions within a key macromolecular complex, the spliceosome. Using S. cerevisiae as a model system we show that the spliceosome network has more indirect paths connecting proteins than random networks. Such multiplicity of paths may promote routes to cascading effects to propagate across the network. However, the modular network structure concentrates paths within modules, thus constraining the propagation of such cascading effects, as indicated by analytical results from the spectral graph theory and by numerical simulations of a minimal mathematical model parameterized with the spliceosome network. We hypothesize that the concentration of paths within modules favors robustness of the spliceosome against failure, but may lead to a higher vulnerability of functional subunits, which may affect the temporal assembly of the spliceosome. Our results illustrate the utility of spectral graph theory for identifying fragile spots in biological systems and predicting their implications.
Collapse
|
87
|
Dasgupta N, Kumar Thakur B, Chakraborty A, Das S. Butyrate-Induced In Vitro Colonocyte Differentiation Network Model Identifies ITGB1, SYK, CDKN2A, CHAF1A, and LRP1 as the Prognostic Markers for Colorectal Cancer Recurrence. Nutr Cancer 2018; 71:257-271. [PMID: 30475060 DOI: 10.1080/01635581.2018.1540715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Numerous mechanisms are believed to contribute to the role of dietary fiber-derived butyrate in the protection against the development of colorectal cancers (CRCs). To identify the most crucial butyrate-regulated genes, we exploited whole genome microarray of HT-29 cells differentiated in vitro by butyrate treatment. Butyrate differentiates HT-29 cells by relaxing the perturbation, caused by mutations of Adenomatous polyposis coli (APC) and TP53 genes, the most frequent mutations observed in CRC. We constructed protein-protein interaction network (PPIN) with the differentially expressed genes after butyrate treatment and extracted the hub genes from the PPIN, which also participated in the APC-TP53 network. The idea behind this approach was that the expression of these hub genes also regulated cell differentiation, and subsequently CRC prognosis by evading the APC-TP53 mutational effect. We used mRNA expression profile of these critical hub genes from seven large CRC cohorts. Logistic Regression showed strong evidence for association of these common hubs with CRC recurrence. In this study, we exploited PPIN to reduce the dimension of microarray biologically and identified five prognostic markers for the CRC recurrence, which were validated across different datasets. Moreover, these five biomarkers we identified increase the predictive value of the TNM staging for CRC recurrence.
Collapse
Affiliation(s)
- Nirmalya Dasgupta
- a Tumor Initiation and Maintenance Program , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , California, USA.,b Department of Clinical Medicine , National Institute of Cholera and Enteric Diseases , Beliaghata , Kolkata, India
| | - Bhupesh Kumar Thakur
- b Department of Clinical Medicine , National Institute of Cholera and Enteric Diseases , Beliaghata , Kolkata, India.,c Department of Immunology , University of Toronto , Toronto , Ontario, CANADA
| | - Abhijit Chakraborty
- d Division of Vaccine Discovery , La Jolla Institute for Allergy and Immunology , La Jolla , California, USA
| | - Santasabuj Das
- b Department of Clinical Medicine , National Institute of Cholera and Enteric Diseases , Beliaghata , Kolkata, India.,e Biomedical Informatics Centre, National Institute of Cholera and Enteric Diseases , Beliaghata , Kolkata, India
| |
Collapse
|
88
|
Haider S, Yao CQ, Sabine VS, Grzadkowski M, Stimper V, Starmans MHW, Wang J, Nguyen F, Moon NC, Lin X, Drake C, Crozier CA, Brookes CL, van de Velde CJH, Hasenburg A, Kieback DG, Markopoulos CJ, Dirix LY, Seynaeve C, Rea DW, Kasprzyk A, Lambin P, Lio' P, Bartlett JMS, Boutros PC. Pathway-based subnetworks enable cross-disease biomarker discovery. Nat Commun 2018; 9:4746. [PMID: 30420699 PMCID: PMC6232113 DOI: 10.1038/s41467-018-07021-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/29/2018] [Indexed: 11/29/2022] Open
Abstract
Biomarkers lie at the heart of precision medicine. Surprisingly, while rapid genomic profiling is becoming ubiquitous, the development of biomarkers usually involves the application of bespoke techniques that cannot be directly applied to other datasets. There is an urgent need for a systematic methodology to create biologically-interpretable molecular models that robustly predict key phenotypes. Here we present SIMMS (Subnetwork Integration for Multi-Modal Signatures): an algorithm that fragments pathways into functional modules and uses these to predict phenotypes. We apply SIMMS to multiple data types across five diseases, and in each it reproducibly identifies known and novel subtypes, and makes superior predictions to the best bespoke approaches. To demonstrate its ability on a new dataset, we profile 33 genes/nodes of the PI3K pathway in 1734 FFPE breast tumors and create a four-subnetwork prediction model. This model out-performs a clinically-validated molecular test in an independent cohort of 1742 patients. SIMMS is generic and enables systematic data integration for robust biomarker discovery.
Collapse
Affiliation(s)
- Syed Haider
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada.
- Computer Laboratory, University of Cambridge, Cambridge, CB3 0FD, United Kingdom.
| | - Cindy Q Yao
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, Canada
| | - Vicky S Sabine
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Michal Grzadkowski
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Vincent Stimper
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Maud H W Starmans
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jianxin Wang
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Francis Nguyen
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, Canada
| | - Nathalie C Moon
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Xihui Lin
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Camilla Drake
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Cheryl A Crozier
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Cassandra L Brookes
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | | | | | | | | | | | | | - Daniel W Rea
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Arek Kasprzyk
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada
| | - Philippe Lambin
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pietro Lio'
- Computer Laboratory, University of Cambridge, Cambridge, CB3 0FD, United Kingdom
| | - John M S Bartlett
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada.
| | - Paul C Boutros
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, M5G 0A3, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada.
| |
Collapse
|
89
|
Chiu YC, Hsiao TH, Wang LJ, Chen Y, Chuang EY. Analyzing Differential Regulatory Networks Modulated by Continuous-State Genomic Features in Glioblastoma Multiforme. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1754-1764. [PMID: 28114032 DOI: 10.1109/tcbb.2016.2635646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Gene regulatory networks are a global representation of complex interactions between molecules that dictate cellular behavior. Study of a regulatory network modulated by single or multiple modulators' expression levels, including microRNAs (miRNAs) and transcription factors (TFs), in different conditions can further reveal the modulators' roles in diseases such as cancers. Existing computational methods for identifying such modulated regulatory networks are typically carried out by comparing groups of samples dichotomized with respect to the modulator status, ignoring the fact that most biological features are intrinsically continuous variables. Here, we devised a sliding window-based regression scheme and proposed the Regression-based Inference of Modulation (RIM) algorithm to infer the dynamic gene regulation modulated by continuous-state modulators. We demonstrated the improvement in performance as well as computation efficiency achieved by RIM. Applying RIM to genome-wide expression profiles of 520 glioblastoma multiforme (GBM) tumors, we investigated miRNA- and TF-modulated gene regulatory networks and showed their association with dynamic cellular processes and brain-related functions in GBM. Overall, the proposed algorithm provides an efficient and robust scheme for comprehensively studying modulated gene regulatory networks.
Collapse
|
90
|
Gysi DM, Voigt A, Fragoso TDM, Almaas E, Nowick K. wTO: an R package for computing weighted topological overlap and a consensus network with integrated visualization tool. BMC Bioinformatics 2018; 19:392. [PMID: 30355288 PMCID: PMC6201546 DOI: 10.1186/s12859-018-2351-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 08/30/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Network analyses, such as of gene co-expression networks, metabolic networks and ecological networks have become a central approach for the systems-level study of biological data. Several software packages exist for generating and analyzing such networks, either from correlation scores or the absolute value of a transformed score called weighted topological overlap (wTO). However, since gene regulatory processes can up- or down-regulate genes, it is of great interest to explicitly consider both positive and negative correlations when constructing a gene co-expression network. RESULTS Here, we present an R package for calculating the weighted topological overlap (wTO), that, in contrast to existing packages, explicitly addresses the sign of the wTO values, and is thus especially valuable for the analysis of gene regulatory networks. The package includes the calculation of p-values (raw and adjusted) for each pairwise gene score. Our package also allows the calculation of networks from time series (without replicates). Since networks from independent datasets (biological repeats or related studies) are not the same due to technical and biological noise in the data, we additionally, incorporated a novel method for calculating a consensus network (CN) from two or more networks into our R package. To graphically inspect the resulting networks, the R package contains a visualization tool, which allows for the direct network manipulation and access of node and link information. When testing the package on a standard laptop computer, we can conduct all calculations for systems of more than 20,000 genes in under two hours. We compare our new wTO package to state of art packages and demonstrate the application of the wTO and CN functions using 3 independently derived datasets from healthy human pre-frontal cortex samples. To showcase an example for the time series application we utilized a metagenomics data set. CONCLUSION In this work, we developed a software package that allows the computation of wTO networks, CNs and a visualization tool in the R statistical environment. It is publicly available on CRAN repositories under the GPL -2 Open Source License ( https://cran.r-project.org/web/packages/wTO/ ).
Collapse
Affiliation(s)
- Deisy Morselli Gysi
- Department of Computer Science, Interdisciplinary Center of Bioinformatics, University of Leipzig, Haertelstrasse 16-18, Leipzig, 04109 Germany
- Swarm Intelligence and Complex Systems Group, Faculty of Mathematics and Computer Science, University of Leipzig, Augustusplatz 10, Leipzig, 04109 Germany
| | - Andre Voigt
- Department of Biotechnology, NTNU - Norwegian University of Science and Technology, Trondheim, N-7049 Norway
| | | | - Eivind Almaas
- Department of Biotechnology, NTNU - Norwegian University of Science and Technology, Trondheim, N-7049 Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, NTNU - Norwegian University of Science and Technology, Trondheim, N-7049 Norway
| | - Katja Nowick
- Freie Universität Berlin, Human Biology Group, Institute for Zoology, Department of Biology, Chemistry and Pharmacy, Königin-Luise-Straße 1-3, Berlin, D-14195 Germany
| |
Collapse
|
91
|
Ibáñez M, Carbonell-Caballero J, Such E, García-Alonso L, Liquori A, López-Pavía M, Llop M, Alonso C, Barragán E, Gómez-Seguí I, Neef A, Hervás D, Montesinos P, Sanz G, Sanz MA, Dopazo J, Cervera J. The modular network structure of the mutational landscape of Acute Myeloid Leukemia. PLoS One 2018; 13:e0202926. [PMID: 30303964 PMCID: PMC6179200 DOI: 10.1371/journal.pone.0202926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is associated with the sequential accumulation of acquired genetic alterations. Although at diagnosis cytogenetic alterations are frequent in AML, roughly 50% of patients present an apparently normal karyotype (NK), leading to a highly heterogeneous prognosis. Due to this significant heterogeneity, it has been suggested that different molecular mechanisms may trigger the disease with diverse prognostic implications. We performed whole-exome sequencing (WES) of tumor-normal matched samples of de novo AML-NK patients lacking mutations in NPM1, CEBPA or FLT3-ITD to identify new gene mutations with potential prognostic and therapeutic relevance to patients with AML. Novel candidate-genes, together with others previously described, were targeted resequenced in an independent cohort of 100 de novo AML patients classified in the cytogenetic intermediate-risk (IR) category. A mean of 4.89 mutations per sample were detected in 73 genes, 35 of which were mutated in more than one patient. After a network enrichment analysis, we defined a single in silico model and established a set of seed-genes that may trigger leukemogenesis in patients with normal karyotype. The high heterogeneity of gene mutations observed in AML patients suggested that a specific alteration could not be as essential as the interaction of deregulated pathways.
Collapse
Affiliation(s)
- Mariam Ibáñez
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, Valencia, Spain
| | - José Carbonell-Caballero
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona, Spain
| | - Esperanza Such
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Luz García-Alonso
- European Molecular Biology Laboratory—European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Alessandro Liquori
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - María López-Pavía
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Marta Llop
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - Carmen Alonso
- Hematology Service, Hospital Arnau de Villanoba, Valencia, Spain
| | - Eva Barragán
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - Inés Gómez-Seguí
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Alexander Neef
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Pau Montesinos
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Guillermo Sanz
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Miguel Angel Sanz
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Joaquín Dopazo
- Functional Genomics Node, Spanish National Institute of Bioinformatics at CIPF, Valencia, Spain
- Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, Sevilla, Spain
- * E-mail: (JC); (JD)
| | - José Cervera
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- * E-mail: (JC); (JD)
| |
Collapse
|
92
|
Typing tumors using pathways selected by somatic evolution. Nat Commun 2018; 9:4159. [PMID: 30297789 PMCID: PMC6175900 DOI: 10.1038/s41467-018-06464-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 09/03/2018] [Indexed: 01/01/2023] Open
Abstract
Many recent efforts to analyze cancer genomes involve aggregation of mutations within reference maps of molecular pathways and protein networks. Here, we find these pathway studies are impeded by molecular interactions that are functionally irrelevant to cancer or the patient’s tumor type, as these interactions diminish the contrast of driver pathways relative to individual frequently mutated genes. This problem can be addressed by creating stringent tumor-specific networks of biophysical protein interactions, identified by signatures of epistatic selection during tumor evolution. Using such an evolutionarily selected pathway (ESP) map, we analyze the major cancer genome atlases to derive a hierarchical classification of tumor subtypes linked to characteristic mutated pathways. These pathways are clinically prognostic and predictive, including the TP53-AXIN-ARHGEF17 combination in liver and CYLC2-STK11-STK11IP in lung cancer, which we validate in independent cohorts. This ESP framework substantially improves the definition of cancer pathways and subtypes from tumor genome data. Informative pathways driving cancer pathogenesis and subtypes can be difficult to identify in the presence of many gene interactions irrelevant to cancer. Here, the authors describe an approach for cancer gene pathway analysis based on key molecular interactions that drive cancer in relevant tissue types, and they assemble a focused map of Evolutionarily Selected Pathways (ESP) with interactions supported by both protein–protein binding and genetic epistasis during somatic tumor evolution.
Collapse
|
93
|
Hao T, Wang Q, Zhao L, Wu D, Wang E, Sun J. Analyzing of Molecular Networks for Human Diseases and Drug Discovery. Curr Top Med Chem 2018; 18:1007-1014. [PMID: 30101711 PMCID: PMC6174636 DOI: 10.2174/1568026618666180813143408] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 06/22/2018] [Accepted: 07/03/2018] [Indexed: 01/11/2023]
Abstract
Molecular networks represent the interactions and relations of genes/proteins, and also encode molecular mechanisms of biological processes, development and diseases. Among the molecular networks, protein-protein Interaction Networks (PINs) have become effective platforms for uncovering the molecular mechanisms of diseases and drug discovery. PINs have been constructed for various organisms and utilized to solve many biological problems. In human, most proteins present their complex functions by interactions with other proteins, and the sum of these interactions represents the human protein interactome. Especially in the research on human disease and drugs, as an emerging tool, the PIN provides a platform to systematically explore the molecular complexities of specific diseases and the references for drug design. In this review, we summarized the commonly used approaches to aid disease research and drug discovery with PINs, including the network topological analysis, identification of novel pathways, drug targets and sub-network biomarkers for diseases. With the development of bioinformatic techniques and biological networks, PINs will play an increasingly important role in human disease research and drug discovery.
Collapse
Affiliation(s)
- Tong Hao
- Tianjin Key Laboratory of Animal and Plant Resistance/College of Life Sciences, Tianjin Normal University, Tianjin 300387, China
| | - Qian Wang
- Tianjin Key Laboratory of Animal and Plant Resistance/College of Life Sciences, Tianjin Normal University, Tianjin 300387, China
| | - Lingxuan Zhao
- Tianjin Key Laboratory of Animal and Plant Resistance/College of Life Sciences, Tianjin Normal University, Tianjin 300387, China
| | - Dan Wu
- Tianjin Key Laboratory of Animal and Plant Resistance/College of Life Sciences, Tianjin Normal University, Tianjin 300387, China
| | - Edwin Wang
- Tianjin Key Laboratory of Animal and Plant Resistance/College of Life Sciences, Tianjin Normal University, Tianjin 300387, China.,University of Calgary Cumming School of Medicine, Calgary, Alberta T2N 4Z6, Canada
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance/College of Life Sciences, Tianjin Normal University, Tianjin 300387, China.,Tianjin Bohai Fisheries Research Institute, Tianjin 300221, China
| |
Collapse
|
94
|
Banerjee SL, Dionne U, Lambert JP, Bisson N. Targeted proteomics analyses of phosphorylation-dependent signalling networks. J Proteomics 2018; 189:39-47. [DOI: 10.1016/j.jprot.2018.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 01/18/2023]
|
95
|
Pires HR, Boxem M. Mapping the Polarity Interactome. J Mol Biol 2018; 430:3521-3544. [DOI: 10.1016/j.jmb.2017.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
|
96
|
Wang L, Cheng F, Hu J, Wang H, Tan N, Li S, Wang X. Pathway-based gene-gene interaction network modelling to predict potential biomarkers of essential hypertension. Biosystems 2018; 172:18-25. [PMID: 30110599 DOI: 10.1016/j.biosystems.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/05/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022]
Abstract
Essential hypertension (EH) is a major risk factor for cardiovascular disease. Despite considerable efforts to elucidate the pathogenesis of EH, there is an imperious need for novel indicators of EH. This study aimed to develop a method to predict potential biomarkers of EH from the point of view of network. A pathway-based gene-gene interaction (GGI) network model was constructed and analyzed, containing 116 nodes and 1272 connections. The nodes represented EH-related genes, and that connections represented their interactions. The network showed a small-world property and uneven degree distribution, suggesting that a few highly interconnected hubs played a vital role in EH. An inherent hierarchy and assortative mixing pattern were also observed in the network. GNAS, GNB3, PF4 and PPBP showed the highest values of degrees and centrality indices, and were chosen as potential biomarkers of EH. A two-mode network model based on the potential biomarkers demonstrated that hemostasis and GPCR ligand binding pathway were key pathways contributing to EH. Results of this study improve our current understanding of the molecular mechanisms driving EH. The selected genes and pathways have the potential to be used in the diagnosis and treatment of EH. Moreover, the combination of pathway analysis and complex network methodology provides a novel strategy for searching new genetic indicators of complex diseases.
Collapse
Affiliation(s)
- Le Wang
- Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China
| | - Fuhong Cheng
- Department of Orthopedics, Weinan Central Hospital, Shaanxi, 714000, China
| | - Jingbo Hu
- College of Electronic and Electrical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China; Center for Nonlinear Complex Systems, Department of Physics, School of Physics and Astronomy, Yunnan University, Kunming, 650091, China
| | - Huan Wang
- College of Computer Science and Technology, Baoji University of Arts and Sciences, Baoji, 721013, China
| | - Nana Tan
- Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China
| | - Shaokang Li
- Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China
| | - Xiaoling Wang
- Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji, 721013, China.
| |
Collapse
|
97
|
Amiri Dash Atan N, Koushki M, Rezaei Tavirani M, Ahmadi NA. Protein-Protein Interaction Network Analysis of Salivary Proteomic Data in Oral Cancer Cases. Asian Pac J Cancer Prev 2018; 19:1639-1645. [PMID: 29937423 PMCID: PMC6103602 DOI: 10.22034/apjcp.2018.19.6.1639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background: Oral cancer is a frequently encountered neoplasm of the head and neck region, being the eight most common type of human malignancy worldwide. Despite improvement in its control, morbidity and mortality rates have improved little in the past decades. Therefore, prevention and/or early detection are a high priority. Proteomics with network analysis have emerged as a powerful tool to identify important proteins associated with cancer development and progression that can be potential targets for early diagnosis. In the present study, network- based protein- protein interactions (PPI) for oral cancer were identified and then analyzed for use as key proteins/potential biomarkers. Material and Methods: Gene expression data in articles which focused on saliva proteomics of oral cancer were collected and 74 candidate genes or proteins were extracted. Related protein networks of differentially expressed proteins were explored and visualized using cytoscape software. Further PPI analysis was performed by Molecular Complex Detection (MCODE) and BiNGO methods. Results: Network analysis of genes/proteins related to oral cancer identified kininogen-1, angiotensinogen, annexin A1, IL-8, IgG heavy variable and constant chains, CRP, collagen alpha-1 and fibronectin as 9 hub-bottleneck proteins. In addition, based on clustering with the MCODE tool, vitronectin, collagen alpha-2, IL-8 and integrin alpha-v were established as 5 distinct seed proteins. Conclusion: A hub-bottleneck protein panel may offer a potential /candidate biomarker pattern for diagnosis and treatment of oral cancer disease. Further investigation and validation of these proteins are warranted.
Collapse
Affiliation(s)
- Nasrin Amiri Dash Atan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | | | | |
Collapse
|
98
|
Ahmed H, Howton TC, Sun Y, Weinberger N, Belkhadir Y, Mukhtar MS. Network biology discovers pathogen contact points in host protein-protein interactomes. Nat Commun 2018; 9:2312. [PMID: 29899369 PMCID: PMC5998135 DOI: 10.1038/s41467-018-04632-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
In all organisms, major biological processes are controlled by complex protein-protein interactions networks (interactomes), yet their structural complexity presents major analytical challenges. Here, we integrate a compendium of over 4300 phenotypes with Arabidopsis interactome (AI-1MAIN). We show that nodes with high connectivity and betweenness are enriched and depleted in conditional and essential phenotypes, respectively. Such nodes are located in the innermost layers of AI-1MAIN and are preferential targets of pathogen effectors. We extend these network-centric analyses to Cell Surface Interactome (CSILRR) and predict its 35 most influential nodes. To determine their biological relevance, we show that these proteins physically interact with pathogen effectors and modulate plant immunity. Overall, our findings contrast with centrality-lethality rule, discover fast information spreading nodes, and highlight the structural properties of pathogen targets in two different interactomes. Finally, this theoretical framework could possibly be applicable to other inter-species interactomes to reveal pathogen contact points.
Collapse
Affiliation(s)
- Hadia Ahmed
- Department of Computer Science, University of Alabama at Birmingham, 115A Campbell Hall, 1300 University Boulevard, Birmingham, AL, 35294, USA
| | - T C Howton
- Department of Biology, University of Alabama at Birmingham, 464 Campbell Hall, 1300 University Boulevard, Birmingham, AL, 35294, USA
| | - Yali Sun
- Department of Biology, University of Alabama at Birmingham, 464 Campbell Hall, 1300 University Boulevard, Birmingham, AL, 35294, USA
| | - Natascha Weinberger
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna Biocenter (VBC), Dr Bohr Gasse 3, 1030, Vienna, Austria
| | - Youssef Belkhadir
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna Biocenter (VBC), Dr Bohr Gasse 3, 1030, Vienna, Austria
| | - M Shahid Mukhtar
- Department of Biology, University of Alabama at Birmingham, 464 Campbell Hall, 1300 University Boulevard, Birmingham, AL, 35294, USA.
- Nutrition Obesity Research Center, University of Alabama at Birmingham, 1675 University Blvd, WEBB 568, Birmingham, AL, 35294, USA.
| |
Collapse
|
99
|
Wu M, Zhu L, Feng X. Network-based feature screening with applications to genome data. Ann Appl Stat 2018. [DOI: 10.1214/17-aoas1097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
100
|
Liluashvili V, Kalayci S, Fluder E, Wilson M, Gabow A, Gümüs ZH. iCAVE: an open source tool for visualizing biomolecular networks in 3D, stereoscopic 3D and immersive 3D. Gigascience 2018; 6:1-13. [PMID: 28814063 PMCID: PMC5554349 DOI: 10.1093/gigascience/gix054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/05/2017] [Indexed: 02/02/2023] Open
Abstract
Visualizations of biomolecular networks assist in systems-level data exploration in many cellular processes. Data generated from high-throughput experiments increasingly inform these networks, yet current tools do not adequately scale with concomitant increase in their size and complexity. We present an open source software platform, interactome-CAVE (iCAVE), for visualizing large and complex biomolecular interaction networks in 3D. Users can explore networks (i) in 3D using a desktop, (ii) in stereoscopic 3D using 3D-vision glasses and a desktop, or (iii) in immersive 3D within a CAVE environment. iCAVE introduces 3D extensions of known 2D network layout, clustering, and edge-bundling algorithms, as well as new 3D network layout algorithms. Furthermore, users can simultaneously query several built-in databases within iCAVE for network generation or visualize their own networks (e.g., disease, drug, protein, metabolite). iCAVE has modular structure that allows rapid development by addition of algorithms, datasets, or features without affecting other parts of the code. Overall, iCAVE is the first freely available open source tool that enables 3D (optionally stereoscopic or immersive) visualizations of complex, dense, or multi-layered biomolecular networks. While primarily designed for researchers utilizing biomolecular networks, iCAVE can assist researchers in any field.
Collapse
Affiliation(s)
- Vaja Liluashvili
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Selim Kalayci
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eugene Fluder
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Manda Wilson
- Computational Biology Center, Memorial-Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aaron Gabow
- Computational Biology Center, Memorial-Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zeynep H Gümüs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|