1
|
Fu Y, Lu Y, Wang Y, Zhang B, Zhang Z, Yu G, Liu C, Clarke R, Herrington DM, Wang Y. DDN3.0: determining significant rewiring of biological network structure with differential dependency networks. Bioinformatics 2024; 40:btae376. [PMID: 38902940 PMCID: PMC11199198 DOI: 10.1093/bioinformatics/btae376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/09/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
MOTIVATION Complex diseases are often caused and characterized by misregulation of multiple biological pathways. Differential network analysis aims to detect significant rewiring of biological network structures under different conditions and has become an important tool for understanding the molecular etiology of disease progression and therapeutic response. With few exceptions, most existing differential network analysis tools perform differential tests on separately learned network structures that are computationally expensive and prone to collapse when grouped samples are limited or less consistent. RESULTS We previously developed an accurate differential network analysis method-differential dependency networks (DDN), that enables joint learning of common and rewired network structures under different conditions. We now introduce the DDN3.0 tool that improves this framework with three new and highly efficient algorithms, namely, unbiased model estimation with a weighted error measure applicable to imbalance sample groups, multiple acceleration strategies to improve learning efficiency, and data-driven determination of proper hyperparameters. The comparative experimental results obtained from both realistic simulations and case studies show that DDN3.0 can help biologists more accurately identify, in a study-specific and often unknown conserved regulatory circuitry, a network of significantly rewired molecular players potentially responsible for phenotypic transitions. AVAILABILITY AND IMPLEMENTATION The Python package of DDN3.0 is freely available at https://github.com/cbil-vt/DDN3. A user's guide and a vignette are provided at https://ddn-30.readthedocs.io/.
Collapse
Affiliation(s)
- Yi Fu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Yingzhou Lu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Yizhi Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Bai Zhang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Zhen Zhang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, P.R. China
| | - Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN 55912, United States
| | - David M Herrington
- Department of Internal Medicine, Wake Forest University, Winston-Salem, NC 27157, United States
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| |
Collapse
|
2
|
Ni Y, He J, Chalise P. Integration of differential expression and network structure for 'omics data analysis. Comput Biol Med 2022; 150:106133. [PMID: 36179515 DOI: 10.1016/j.compbiomed.2022.106133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/23/2022] [Accepted: 09/18/2022] [Indexed: 11/25/2022]
Abstract
Differential expression (DE) analysis has been routinely used to identify molecular features that are statistically significantly different between distinct biological groups. In recent years, differential network (DN) analysis has emerged as a powerful approach to uncover molecular network structure changes from one biological condition to the other where the molecular features with larger topological changes are selected as biomarkers. Although a large number of DE and a few DN-based methods are available, they have been usually implemented independently. DE analysis ignores the relationship among molecular features while DN analysis does not account for the expression changes at individual level. Therefore, an integrative analysis approach that accounts for both DE and DN is required to identify disease associated key features. Although, a handful of methods have been proposed, there is no method that optimizes the combination of DE and DN. We propose a novel integrative analysis method, DNrank, to identify disease-associated molecular features that leverages the strengths of both DE and DN by calculating a weight using resampling based cross validation scheme within the algorithm. First, differential expression analysis of individual molecular features is carried out. Second, a differential network structure is constructed using the differential partial correlation analysis. Third, the molecular features are ranked in the order of their significances by integrating their DE measures and DN structure using the modified Google's PageRank algorithm. In the algorithm, the optimum combination of DE and DN analyses is achieved by evaluating the prediction performance of top-ranked features utilizing support vector machine classifier with Monte Carlo cross validation. The proposed method is illustrated using both simulated data and three real data sets. The results show that the proposed method has a better performance in identifying important molecular features with respect to predictive discrimination. Also, as compared to existing feature selection methods, the top-ranked features selected by our method had a higher stability in selection. DNrank allows the researchers to identify the disease-associated features by utilizing both expression and network topology changes between two groups.
Collapse
Affiliation(s)
- Yonghui Ni
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Jianghua He
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| |
Collapse
|
3
|
Degree of Freedom of Gene Expression in Saccharomyces cerevisiae. Microbiol Spectr 2022; 10:e0083821. [PMID: 35230153 PMCID: PMC9045123 DOI: 10.1128/spectrum.00838-21] [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] [Indexed: 11/20/2022] Open
Abstract
The complexity of genome-wide gene expression has not yet been adequately addressed due to a lack of comprehensive statistical analyses. In the present study, we introduce degree of freedom (DOF) as a summary statistic for evaluating gene expression complexity. Because DOF can be interpreted by a state-space representation, application of the DOF is highly useful for understanding gene activities. We used over 11,000 gene expression data sets to reveal that the DOF of gene expression in Saccharomyces cerevisiae is not greater than 450. We further demonstrated that various degrees of freedom of gene expression can be interpreted by different sequence motifs within promoter regions and Gene Ontology (GO) terms. The well-known TATA box is the most significant one among the identified motifs, while the GO term "ribosome genesis" is an associated biological process. On the basis of transcriptional freedom, our findings suggest that the regulation of gene expression can be modeled using only a few state variables. IMPORTANCE Yeast works like a well-organized factory. Each of its components works in its own way, while affecting the activities of others. The order of all activities is largely governed by the regulation of gene expression. In recent decades, biologists have recognized many regulations for yeast genes. However, it is not known how closely the regulation links each gene together to make all components of the cell work as a whole. In other words, biologists are very interested in how many independent control factors are needed to operate an artificial "cell" that works the same as a real one. In this work, we suggested that only 450 control factors were sufficient to represent the regulation of all 5800 yeast genes.
Collapse
|
4
|
Chen L, Wu CT, Lin CH, Dai R, Liu C, Clarke R, Yu G, Van Eyk JE, Herrington DM, Wang Y. swCAM: estimation of subtype-specific expressions in individual samples with unsupervised sample-wise deconvolution. Bioinformatics 2022; 38:1403-1410. [PMID: 34904628 PMCID: PMC8826012 DOI: 10.1093/bioinformatics/btab839] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/30/2021] [Accepted: 12/10/2021] [Indexed: 02/04/2023] Open
Abstract
MOTIVATION Complex biological tissues are often a heterogeneous mixture of several molecularly distinct cell subtypes. Both subtype compositions and subtype-specific (STS) expressions can vary across biological conditions. Computational deconvolution aims to dissect patterns of bulk tissue data into subtype compositions and STS expressions. Existing deconvolution methods can only estimate averaged STS expressions in a population, while many downstream analyses such as inferring co-expression networks in particular subtypes require subtype expression estimates in individual samples. However, individual-level deconvolution is a mathematically underdetermined problem because there are more variables than observations. RESULTS We report a sample-wise Convex Analysis of Mixtures (swCAM) method that can estimate subtype proportions and STS expressions in individual samples from bulk tissue transcriptomes. We extend our previous CAM framework to include a new term accounting for between-sample variations and formulate swCAM as a nuclear-norm and ℓ2,1-norm regularized matrix factorization problem. We determine hyperparameter values using cross-validation with random entry exclusion and obtain a swCAM solution using an efficient alternating direction method of multipliers. Experimental results on realistic simulation data show that swCAM can accurately estimate STS expressions in individual samples and successfully extract co-expression networks in particular subtypes that are otherwise unobtainable using bulk data. In two real-world applications, swCAM analysis of bulk RNASeq data from brain tissue of cases and controls with bipolar disorder or Alzheimer's disease identified significant changes in cell proportion, expression pattern and co-expression module in patient neurons. Comparative evaluation of swCAM versus peer methods is also provided. AVAILABILITY AND IMPLEMENTATION The R Scripts of swCAM are freely available at https://github.com/Lululuella/swCAM. A user's guide and a vignette are provided. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Lulu Chen
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Chiung-Ting Wu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Chia-Hsiang Lin
- Department of Electrical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Guoqiang Yu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David M Herrington
- Department of Internal Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| |
Collapse
|
5
|
Network Biology and Artificial Intelligence Drive the Understanding of the Multidrug Resistance Phenotype in Cancer. Drug Resist Updat 2022; 60:100811. [DOI: 10.1016/j.drup.2022.100811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
|
6
|
Chen L, Zhou J, Lin L. Hypothesis testing for populations of networks. COMMUN STAT-THEOR M 2021. [DOI: 10.1080/03610926.2021.1977961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Li Chen
- College of Mathematics, Southwest Minzu University, Chengdu, China
| | - Jie Zhou
- College of Mathematics, Sichuan University, Chengdu, China
| | - Lizhen Lin
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, South Bend, Indiana, USA
| |
Collapse
|
7
|
Ottenbros I, Govarts E, Lebret E, Vermeulen R, Schoeters G, Vlaanderen J. Network Analysis to Identify Communities Among Multiple Exposure Biomarkers Measured at Birth in Three Flemish General Population Samples. Front Public Health 2021; 9:590038. [PMID: 33643986 PMCID: PMC7902692 DOI: 10.3389/fpubh.2021.590038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/15/2021] [Indexed: 01/07/2023] Open
Abstract
Introduction: Humans are exposed to multiple environmental chemicals via different sources resulting in complex real-life exposure patterns. Insight into these patterns is important for applications such as linkage to health effects and (mixture) risk assessment. By providing internal exposure levels of (metabolites of) chemicals, biomonitoring studies can provide snapshots of exposure patterns and factors that drive them. Presentation of biomonitoring data in networks facilitates the detection of such exposure patterns and allows for the systematic comparison of observed exposure patterns between datasets and strata within datasets. Methods: We demonstrate the use of network techniques in human biomonitoring data from cord blood samples collected in three campaigns of the Flemish Environment and Health Studies (FLEHS) (sampling years resp. 2002-2004, 2008-2009, and 2013-2014). Measured biomarkers were multiple organochlorine compounds, PFAS and metals. Comparative network analysis (CNA) was conducted to systematically compare networks between sampling campaigns, smoking status during pregnancy, and maternal pre-pregnancy BMI. Results: Network techniques offered an intuitive approach to visualize complex correlation structures within human biomonitoring data. The identification of groups of highly connected biomarkers, "communities," within these networks highlighted which biomarkers should be considered collectively in the analysis and interpretation of epidemiological studies or in the design of toxicological mixture studies. Network analyses demonstrated in our example to which extent biomarker networks and its communities changed across the sampling campaigns, smoking status during pregnancy, and maternal pre-pregnancy BMI. Conclusion: Network analysis is a data-driven and intuitive screening method when dealing with multiple exposure biomarkers, which can easily be upscaled to high dimensional HBM datasets, and can inform mixture risk assessment approaches.
Collapse
Affiliation(s)
- Ilse Ottenbros
- Center for Sustainability, Environment and Health, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands.,Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Eva Govarts
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Erik Lebret
- Center for Sustainability, Environment and Health, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands.,Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Greet Schoeters
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jelle Vlaanderen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
8
|
Wang YXR, Li L, Li JJ, Huang H. Network Modeling in Biology: Statistical Methods for Gene and Brain Networks. Stat Sci 2021; 36:89-108. [PMID: 34305304 PMCID: PMC8296984 DOI: 10.1214/20-sts792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The rise of network data in many different domains has offered researchers new insight into the problem of modeling complex systems and propelled the development of numerous innovative statistical methodologies and computational tools. In this paper, we primarily focus on two types of biological networks, gene networks and brain networks, where statistical network modeling has found both fruitful and challenging applications. Unlike other network examples such as social networks where network edges can be directly observed, both gene and brain networks require careful estimation of edges using covariates as a first step. We provide a discussion on existing statistical and computational methods for edge esitimation and subsequent statistical inference problems in these two types of biological networks.
Collapse
Affiliation(s)
- Y X Rachel Wang
- School of Mathematics and Statistics, University of Sydney, Australia
| | - Lexin Li
- Department of Biostatistics and Epidemiology, School of Public Health, University of California, Berkeley
| | | | - Haiyan Huang
- Department of Statistics, University of California, Berkeley
| |
Collapse
|
9
|
Savino A, Provero P, Poli V. Differential Co-Expression Analyses Allow the Identification of Critical Signalling Pathways Altered during Tumour Transformation and Progression. Int J Mol Sci 2020; 21:E9461. [PMID: 33322692 PMCID: PMC7764314 DOI: 10.3390/ijms21249461] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 02/02/2023] Open
Abstract
Biological systems respond to perturbations through the rewiring of molecular interactions, organised in gene regulatory networks (GRNs). Among these, the increasingly high availability of transcriptomic data makes gene co-expression networks the most exploited ones. Differential co-expression networks are useful tools to identify changes in response to an external perturbation, such as mutations predisposing to cancer development, and leading to changes in the activity of gene expression regulators or signalling. They can help explain the robustness of cancer cells to perturbations and identify promising candidates for targeted therapy, moreover providing higher specificity with respect to standard co-expression methods. Here, we comprehensively review the literature about the methods developed to assess differential co-expression and their applications to cancer biology. Via the comparison of normal and diseased conditions and of different tumour stages, studies based on these methods led to the definition of pathways involved in gene network reorganisation upon oncogenes' mutations and tumour progression, often converging on immune system signalling. A relevant implementation still lagging behind is the integration of different data types, which would greatly improve network interpretability. Most importantly, performance and predictivity evaluation of the large variety of mathematical models proposed would urgently require experimental validations and systematic comparisons. We believe that future work on differential gene co-expression networks, complemented with additional omics data and experimentally tested, will considerably improve our insights into the biology of tumours.
Collapse
Affiliation(s)
- Aurora Savino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Corso Massimo D’Ázeglio 52, 10126 Turin, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Valeria Poli
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
10
|
Xie J, Yang F, Wang J, Karikomi M, Yin Y, Sun J, Wen T, Nie Q. DNF: A differential network flow method to identify rewiring drivers for gene regulatory networks. Neurocomputing 2020; 410:202-210. [PMID: 34025035 PMCID: PMC8139126 DOI: 10.1016/j.neucom.2020.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Differential network analysis has become an important approach in identifying driver genes in development and disease. However, most studies capture only local features of the underlying gene-regulatory network topology. These approaches are vulnerable to noise and other changes which mask driver-gene activity. Therefore, methods are urgently needed which can separate the impact of true regulatory elements from stochastic changes and downstream effects. We propose the differential network flow (DNF) method to identify key regulators of progression in development or disease. Given the network representation of consecutive biological states, DNF quantifies the essentiality of each node by differences in the distribution of network flow, which are capable of capturing comprehensive topological differences from local to global feature domains. DNF achieves more accurate driver-gene identification than other state-of-the-art methods when applied to four human datasets from The Cancer Genome Atlas and three single-cell RNA-seq datasets of murine neural and hematopoietic differentiation. Furthermore, we predict key regulators of crosstalk between separate networks underlying both neuronal differentiation and the progression of neurodegenerative disease, among which APP is predicted as a driver gene of neural stem cell differentiation. Our method is a new approach for quantifying the essentiality of genes across networks of different biological states.
Collapse
Affiliation(s)
- Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China
| | - Fuzhang Yang
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Mathew Karikomi
- Department of Mathematics, Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-3875, USA
| | - Yiting Yin
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China
| | - Jiamin Sun
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Qing Nie
- Department of Mathematics, Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-3875, USA
| |
Collapse
|
11
|
Clarke R, Kraikivski P, Jones BC, Sevigny CM, Sengupta S, Wang Y. A systems biology approach to discovering pathway signaling dysregulation in metastasis. Cancer Metastasis Rev 2020; 39:903-918. [PMID: 32776157 PMCID: PMC7487029 DOI: 10.1007/s10555-020-09921-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
Total metastatic burden is the primary cause of death for many cancer patients. While the process of metastasis has been studied widely, much remains to be understood. Moreover, few agents have been developed that specifically target the major steps of the metastatic cascade. Many individual genes and pathways have been implicated in metastasis but a holistic view of how these interact and cooperate to regulate and execute the process remains somewhat rudimentary. It is unclear whether all of the signaling features that regulate and execute metastasis are yet fully understood. Novel features of a complex system such as metastasis can often be discovered by taking a systems-based approach. We introduce the concepts of systems modeling and define some of the central challenges facing the application of a multidisciplinary systems-based approach to understanding metastasis and finding actionable targets therein. These challenges include appreciating the unique properties of the high-dimensional omics data often used for modeling, limitations in knowledge of the system (metastasis), tumor heterogeneity and sampling bias, and some of the issues key to understanding critical features of molecular signaling in the context of metastasis. We also provide a brief introduction to integrative modeling that focuses on both the nodes and edges of molecular signaling networks. Finally, we offer some observations on future directions as they relate to developing a systems-based model of the metastatic cascade.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA.
- Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN, 55912, USA.
| | - Pavel Kraikivski
- Academy of Integrated Science, Division of Systems Biology, Virginia Polytechnic and State University, Blacksburg, VA, 24061, USA
| | - Brandon C Jones
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Catherine M Sevigny
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Surojeet Sengupta
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Yue Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| |
Collapse
|
12
|
Kernel Differential Subgraph Analysis to Reveal the Key Period Affecting Glioblastoma. Biomolecules 2020; 10:biom10020318. [PMID: 32079293 PMCID: PMC7072688 DOI: 10.3390/biom10020318] [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: 10/23/2019] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma (GBM) is a fast-growing type of malignant primary brain tumor. To explore the mechanisms in GBM, complex biological networks are used to reveal crucial changes among different biological states, which reflect on the development of living organisms. It is critical to discover the kernel differential subgraph (KDS) that leads to drastic changes. However, identifying the KDS is similar to the Steiner Tree problem that is an NP-hard problem. In this paper, we developed a criterion to explore the KDS (CKDS), which considered the connectivity and scale of KDS, the topological difference of nodes and function relevance between genes in the KDS. The CKDS algorithm was applied to simulated datasets and three single-cell RNA sequencing (scRNA-seq) datasets including GBM, fetal human cortical neurons (FHCN) and neural differentiation. Then we performed the network topology and functional enrichment analyses on the extracted KDSs. Compared with the state-of-art methods, the CKDS algorithm outperformed on simulated datasets to discover the KDSs. In the GBM and FHCN, seventeen genes (one biomarker, nine regulatory genes, one driver genes, six therapeutic targets) and KEGG pathways in KDSs were strongly supported by literature mining that they were highly interrelated with GBM. Moreover, focused on GBM, there were fifteen genes (including ten regulatory genes, three driver genes, one biomarkers, one therapeutic target) and KEGG pathways found in the KDS of neural differentiation process from activated neural stem cells (aNSC) to neural progenitor cells (NPC), while few genes and no pathway were found in the period from NPC to astrocytes (Ast). These experiments indicated that the process from aNSC to NPC is a key differentiation period affecting the development of GBM. Therefore, the CKDS algorithm provides a unique perspective in identifying cell-type-specific genes and KDSs.
Collapse
|
13
|
X-Module: A novel fusion measure to associate co-expressed gene modules from condition-specific expression profiles. J Biosci 2020. [DOI: 10.1007/s12038-020-0007-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Kakati T, Bhattacharyya DK, Kalita JK. X-Module: A novel fusion measure to associate co-expressed gene modules from condition-specific expression profiles. J Biosci 2020; 45:33. [PMID: 32098912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A gene co-expression network (CEN) is of biological interest, since co-expressed genes share common functions and biological processes or pathways. Finding relationships among modules can reveal inter-modular preservation, and similarity in transcriptome, functional, and biological behaviors among modules of the same or two different datasets. There is no method which explores the one-to-one relationships and one-to-many relationships among modules extracted from control and disease samples based on both topological and semantic similarity using both microarray and RNA seq data. In this work, we propose a novel fusion measure to detect mapping between modules from two sets of co-expressed modules extracted from control and disease stages of Alzheimer's disease (AD) and Parkinson's disease (PD) datasets. Our measure considers both topological and biological information of a module and is an estimation of four parameters, namely, semantic similarity, eigengene correlation, degree difference, and the number of common genes. We analyze the consensus modules shared between both control and disease stages in terms of their association with diseases. We also validate the close associations between human and chimpanzee modules and compare with the state-ofthe- art method. Additionally, we propose two novel observations on the relationships between modules for further analysis.
Collapse
Affiliation(s)
- Tulika Kakati
- Department of Computer Science and Engineering, Tezpur University, Tezpur, Assam, India
| | | | | |
Collapse
|
15
|
Tolios A, De Las Rivas J, Hovig E, Trouillas P, Scorilas A, Mohr T. Computational approaches in cancer multidrug resistance research: Identification of potential biomarkers, drug targets and drug-target interactions. Drug Resist Updat 2019; 48:100662. [PMID: 31927437 DOI: 10.1016/j.drup.2019.100662] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Like physics in the 19th century, biology and molecular biology in particular, has been fertilized and enhanced like few other scientific fields, by the incorporation of mathematical methods. In the last decades, a whole new scientific field, bioinformatics, has developed with an output of over 30,000 papers a year (Pubmed search using the keyword "bioinformatics"). Huge databases of mass throughput data have been established, with ArrayExpress alone containing more than 2.7 million assays (October 2019). Computational methods have become indispensable tools in molecular biology, particularly in one of the most challenging areas of cancer research, multidrug resistance (MDR). However, confronted with a plethora of different algorithms, approaches, and methods, the average researcher faces key questions: Which methods do exist? Which methods can be used to tackle the aims of a given study? Or, more generally, how do I use computational biology/bioinformatics to bolster my research? The current review is aimed at providing guidance to existing methods with relevance to MDR research. In particular, we provide an overview on: a) the identification of potential biomarkers using expression data; b) the prediction of treatment response by machine learning methods; c) the employment of network approaches to identify gene/protein regulatory networks and potential key players; d) the identification of drug-target interactions; e) the use of bipartite networks to identify multidrug targets; f) the identification of cellular subpopulations with the MDR phenotype; and, finally, g) the use of molecular modeling methods to guide and enhance drug discovery. This review shall serve as a guide through some of the basic concepts useful in MDR research. It shall give the reader some ideas about the possibilities in MDR research by using computational tools, and, finally, it shall provide a short overview of relevant literature.
Collapse
Affiliation(s)
- A Tolios
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Institute of Clinical Chemistry and Laboratory Medicine, Heinrich Heine University, Duesseldorf, Germany.
| | - J De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Campus Miguel de Unamuno s/n, Salamanca, Spain.
| | - E Hovig
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital and Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway.
| | - P Trouillas
- UMR 1248 INSERM, Univ. Limoges, 2 rue du Dr Marland, 87052, Limoges, France; RCPTM, University Palacký of Olomouc, tr. 17. listopadu 12, 771 46, Olomouc, Czech Republic.
| | - A Scorilas
- Department of Biochemistry & Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| | - T Mohr
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria; ScienceConsult - DI Thomas Mohr KG, Guntramsdorf, Austria.
| |
Collapse
|
16
|
Clarke R, Tyson JJ, Tan M, Baumann WT, Jin L, Xuan J, Wang Y. Systems biology: perspectives on multiscale modeling in research on endocrine-related cancers. Endocr Relat Cancer 2019; 26:R345-R368. [PMID: 30965282 PMCID: PMC7045974 DOI: 10.1530/erc-18-0309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
Drawing on concepts from experimental biology, computer science, informatics, mathematics and statistics, systems biologists integrate data across diverse platforms and scales of time and space to create computational and mathematical models of the integrative, holistic functions of living systems. Endocrine-related cancers are well suited to study from a systems perspective because of the signaling complexities arising from the roles of growth factors, hormones and their receptors as critical regulators of cancer cell biology and from the interactions among cancer cells, normal cells and signaling molecules in the tumor microenvironment. Moreover, growth factors, hormones and their receptors are often effective targets for therapeutic intervention, such as estrogen biosynthesis, estrogen receptors or HER2 in breast cancer and androgen receptors in prostate cancer. Given the complexity underlying the molecular control networks in these cancers, a simple, intuitive understanding of how endocrine-related cancers respond to therapeutic protocols has proved incomplete and unsatisfactory. Systems biology offers an alternative paradigm for understanding these cancers and their treatment. To correctly interpret the results of systems-based studies requires some knowledge of how in silico models are built, and how they are used to describe a system and to predict the effects of perturbations on system function. In this review, we provide a general perspective on the field of cancer systems biology, and we explore some of the advantages, limitations and pitfalls associated with using predictive multiscale modeling to study endocrine-related cancers.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Ming Tan
- Department of Biostatistics, Bioinformatics & Biomathematics, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - William T Baumann
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Lu Jin
- Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Jianhua Xuan
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, Virginia, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, Virginia, USA
| |
Collapse
|
17
|
Jung S. KEDDY: a knowledge-based statistical gene set test method to detect differential functional protein-protein interactions. Bioinformatics 2019; 35:619-627. [PMID: 30101275 DOI: 10.1093/bioinformatics/bty686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 07/18/2018] [Accepted: 08/06/2018] [Indexed: 12/30/2022] Open
Abstract
MOTIVATION Identifying differential patterns between conditions is a popular approach to understanding the discrepancy between different biological contexts. Although many statistical tests were proposed for identifying gene sets with differential patterns based on different definitions of differentiality, few methods were suggested to identify gene sets with differential functional protein networks due to computational complexity. RESULTS We propose a method of Knowledge-based Evaluation of Dependency DifferentialitY (KEDDY), which is a statistical test for differential functional protein networks of a set of genes between two conditions with utilizing known functional protein-protein interaction information. Unlike other approaches focused on differential expressions of individual genes or differentiality of individual interactions, KEDDY compares two conditions by evaluating the probability distributions of functional protein networks based on known functional protein-protein interactions. The method has been evaluated and compared with previous methods through simulation studies, where KEDDY achieves significantly improved performance in accuracy and speed than the previous method that does not use prior knowledge and better performance in identifying gene sets with differential interactions than other methods evaluating changes in gene expressions. Applications to cancer data sets show that KEDDY identifies alternative cancer subtype-related differential gene sets compared to other differential expression-based methods, and the results also provide detailed gene regulatory information that drives the differentiality of the gene sets. AVAILABILITY AND IMPLEMENTATION The Java implementation of KEDDY is freely available to non-commercial users at https://sites.google.com/site/sjunggsm/keddy. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sungwon Jung
- Department of Genome Medicine and Science, Gachon University College of Medicine, Incheon, Republic of Korea.,Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
18
|
Gomez-Varela D, Barry AM, Schmidt M. Proteome-based systems biology in chronic pain. J Proteomics 2019; 190:1-11. [DOI: 10.1016/j.jprot.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/15/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
|
19
|
Harvey LD, Chan SY. Evolving systems biology approaches to understanding non-coding RNAs in pulmonary hypertension. J Physiol 2018; 597:1199-1208. [PMID: 30113078 DOI: 10.1113/jp275855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/04/2018] [Indexed: 01/17/2023] Open
Abstract
Our appreciation of the roles of non-coding RNAs, in particular microRNAs, in the manifestation of pulmonary hypertension (PH) has advanced considerably over the past decade. Comprised of small nucleotide sequences, microRNAs have demonstrated critical and broad regulatory roles in the pathogenesis of PH via the direct binding to messenger RNA transcripts for degradation or inhibition of translation, thereby exerting a profound influence on cellular activity. Yet, as inherently pleiotropic molecules, microRNAs have been difficult to study using traditional, reductionist approaches alone. With the advent of high-throughput -omics technologies and more advanced computational modelling, the study of microRNAs and their multi-faceted and complex functions in human disease serves as a fertile platform for the application of systems biology methodologies in combination with traditional experimental techniques. Here, we offer our viewpoint of past successes of systems biology in elucidating the otherwise hidden actions of microRNAs in PH, as well as areas for future development to integrate these strategies into the discovery of RNA pathobiology in this disease. We contend that such successful applications of systems biology in elucidating the functional architecture of microRNA regulation will further reveal the molecular mechanisms of disease, while simultaneously revealing potential diagnostic and therapeutic strategies in disease amelioration.
Collapse
Affiliation(s)
- Lloyd D Harvey
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Barry AM, Sondermann JR, Sondermann JH, Gomez-Varela D, Schmidt M. Region-Resolved Quantitative Proteome Profiling Reveals Molecular Dynamics Associated With Chronic Pain in the PNS and Spinal Cord. Front Mol Neurosci 2018; 11:259. [PMID: 30154697 PMCID: PMC6103001 DOI: 10.3389/fnmol.2018.00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022] Open
Abstract
To obtain a thorough understanding of chronic pain, large-scale molecular mapping of the pain axis at the protein level is necessary, but has not yet been achieved. We applied quantitative proteome profiling to build a comprehensive protein compendium of three regions of the pain neuraxis in mice: the sciatic nerve (SN), the dorsal root ganglia (DRG), and the spinal cord (SC). Furthermore, extensive bioinformatics analysis enabled us to reveal unique protein subsets which are specifically enriched in the peripheral nervous system (PNS) and SC. The immense value of these datasets for the scientific community is highlighted by validation experiments, where we monitored protein network dynamics during neuropathic pain. Here, we resolved profound region-specific differences and distinct changes of PNS-enriched proteins under pathological conditions. Overall, we provide a unique and validated systems biology proteome resource (summarized in our online database painproteome.em.mpg.de), which facilitates mechanistic insights into somatosensory biology and chronic pain—a prerequisite for the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Allison M Barry
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Julia R Sondermann
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Jan-Hendrik Sondermann
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - David Gomez-Varela
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Manuela Schmidt
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| |
Collapse
|
21
|
Ji J, He D, Feng Y, He Y, Xue F, Xie L. JDINAC: joint density-based non-parametric differential interaction network analysis and classification using high-dimensional sparse omics data. Bioinformatics 2018; 33:3080-3087. [PMID: 28582486 DOI: 10.1093/bioinformatics/btx360] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 06/01/2017] [Indexed: 12/26/2022] Open
Abstract
Motivation A complex disease is usually driven by a number of genes interwoven into networks, rather than a single gene product. Network comparison or differential network analysis has become an important means of revealing the underlying mechanism of pathogenesis and identifying clinical biomarkers for disease classification. Most studies, however, are limited to network correlations that mainly capture the linear relationship among genes, or rely on the assumption of a parametric probability distribution of gene measurements. They are restrictive in real application. Results We propose a new Joint density based non-parametric Differential Interaction Network Analysis and Classification (JDINAC) method to identify differential interaction patterns of network activation between two groups. At the same time, JDINAC uses the network biomarkers to build a classification model. The novelty of JDINAC lies in its potential to capture non-linear relations between molecular interactions using high-dimensional sparse data as well as to adjust confounding factors, without the need of the assumption of a parametric probability distribution of gene measurements. Simulation studies demonstrate that JDINAC provides more accurate differential network estimation and lower classification error than that achieved by other state-of-the-art methods. We apply JDINAC to a Breast Invasive Carcinoma dataset, which includes 114 patients who have both tumor and matched normal samples. The hub genes and differential interaction patterns identified were consistent with existing experimental studies. Furthermore, JDINAC discriminated the tumor and normal sample with high accuracy by virtue of the identified biomarkers. JDINAC provides a general framework for feature selection and classification using high-dimensional sparse omics data. Availability and implementation R scripts available at https://github.com/jijiadong/JDINAC. Contact lxie@iscb.org. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jiadong Ji
- Department of Mathematical Statistics, School of Statistics, Shandong University of Finance and Economics, Jinan 250014, China
| | - Di He
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Yang Feng
- Department of Statistics, Columbia University, New York, NY 10027, USA
| | - Yong He
- Department of Mathematical Statistics, School of Statistics, Shandong University of Finance and Economics, Jinan 250014, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, Jinan 250012, China
| | - Lei Xie
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY 10016, USA.,Department of Computer Science, Hunter College, The City University of New York, NY 10065, USA
| |
Collapse
|
22
|
Li C, Liu L, Dinu V. Pathways of topological rank analysis (PoTRA): a novel method to detect pathways involved in hepatocellular carcinoma. PeerJ 2018; 6:e4571. [PMID: 29666752 PMCID: PMC5896492 DOI: 10.7717/peerj.4571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
Complex diseases such as cancer are usually the result of a combination of environmental factors and one or several biological pathways consisting of sets of genes. Each biological pathway exerts its function by delivering signaling through the gene network. Theoretically, a pathway is supposed to have a robust topological structure under normal physiological conditions. However, the pathway's topological structure could be altered under some pathological condition. It is well known that a normal biological network includes a small number of well-connected hub nodes and a large number of nodes that are non-hubs. In addition, it is reported that the loss of connectivity is a common topological trait of cancer networks, which is an assumption of our method. Hence, from normal to cancer, the process of the network losing connectivity might be the process of disrupting the structure of the network, namely, the number of hub genes might be altered in cancer compared to that in normal or the distribution of topological ranks of genes might be altered. Based on this, we propose a new PageRank-based method called Pathways of Topological Rank Analysis (PoTRA) to detect pathways involved in cancer. We use PageRank to measure the relative topological ranks of genes in each biological pathway, then select hub genes for each pathway, and use Fisher's exact test to test if the number of hub genes in each pathway is altered from normal to cancer. Alternatively, if the distribution of topological ranks of gene in a pathway is altered between normal and cancer, this pathway might also be involved in cancer. Hence, we use the Kolmogorov-Smirnov test to detect pathways that have an altered distribution of topological ranks of genes between two phenotypes. We apply PoTRA to study hepatocellular carcinoma (HCC) and several subtypes of HCC. Very interestingly, we discover that all significant pathways in HCC are cancer-associated generally, while several significant pathways in subtypes of HCC are HCC subtype-associated specifically. In conclusion, PoTRA is a new approach to explore and discover pathways involved in cancer. PoTRA can be used as a complement to other existing methods to broaden our understanding of the biological mechanisms behind cancer at the system-level.
Collapse
Affiliation(s)
- Chaoxing Li
- School of Life Sciences, Arizona State University, Tempe, AZ, United States of America
| | - Li Liu
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ, United States of America
| | - Valentin Dinu
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ, United States of America
| |
Collapse
|
23
|
Xie J, Lu D, Li J, Wang J, Zhang Y, Li Y, Nie Q. Kernel differential subgraph reveals dynamic changes in biomolecular networks. J Bioinform Comput Biol 2017; 16:1750027. [PMID: 29281952 DOI: 10.1142/s0219720017500275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Many major diseases, including various types of cancer, are increasingly threatening human health. However, the mechanisms of the dynamic processes underlying these diseases remain ambiguous. From the holistic perspective of systems science, complex biological networks can reveal biological phenomena. Changes among networks in different states influence the direction of living organisms. The identification of the kernel differential subgraph (KDS) that leads to drastic changes is critical. The existing studies contribute to the identification of a KDS in networks with the same nodes; however, networks in different states involve the disappearance of some nodes or the appearance of some new nodes. In this paper, we propose a new topology-based KDS (TKDS) method to explore the core module from gene regulatory networks with different nodes in this process. For the common nodes, the TKDS method considers the differential value (D-value) of the topological change. For the different nodes, TKDS identifies the most similar gene pairs and computes the D-value. Hence, TKDS discovers the essential KDS, which considers the relationships between the same nodes as well as different nodes. After applying this method to non-small cell lung cancer (NSCLC), we identified 30 genes that are most likely related to NSCLC and extracted the KDSs in both the cancer and normal states. Two significance functional modules were revealed, and gene ontology (GO) analyses and literature mining indicated that the KDSs are essential to the processes in NSCLC. In addition, compared with existing methods, TKDS provides a unique perspective in identifying particular genes and KDSs related to NSCLC. Moreover, TKDS has the potential to predict other critical disease-related genes and modules.
Collapse
Affiliation(s)
- Jiang Xie
- * School of Computer Engineering and Science, Shanghai University, 99 Shang Da Road, Shanghai 200444, P. R. China
| | - Dongfang Lu
- * School of Computer Engineering and Science, Shanghai University, 99 Shang Da Road, Shanghai 200444, P. R. China
| | - Jiaxin Li
- * School of Computer Engineering and Science, Shanghai University, 99 Shang Da Road, Shanghai 200444, P. R. China
| | - Jiao Wang
- † Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, 99 Shang Da Road, Shanghai 200444, P. R. China
| | - Yong Zhang
- ‡ Pulmonary Department, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, P. R. China
| | - Yanhui Li
- ‡ Pulmonary Department, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, P. R. China
| | - Qing Nie
- § Department of Mathematics, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
24
|
Thomas SN, Chen L, Liu Y, Höti N, Zhang H. Targeted Proteomic Analyses of Histone H4 Acetylation Changes Associated with Homologous-Recombination-Deficient High-Grade Serous Ovarian Carcinomas. J Proteome Res 2017; 16:3704-3710. [PMID: 28866885 DOI: 10.1021/acs.jproteome.7b00405] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Approximately 20% of high-grade serous ovarian cancers are homologous-recombination (HR)-deficient due to genetic and epigenetic mutations of HR pathway genes including the tumor suppressor genes BRCA1 and 2. HR deficiency (HRD) compromises cells' ability to efficiently repair DNA damage, but it also increases sensitivity to chemotherapeutic treatment strategies; however, not all ovarian cancer patients with HRD tumors exhibit positive responses to chemotherapy. Our previous iTRAQ-based comprehensive proteomic characterization of high-grade serous ovarian carcinomas found that lower levels of histone H4 acetylation at Lys12 and Lys16 (H4-K12acK16ac) were associated with HRD tumors compared with non-HRD tumors. In the current study, we developed and validated an H4-K12acK16ac parallel-reaction-monitoring (PRM)-targeted mass-spectrometry-based assay to analyze acetylation changes of histone H4 and to determine the association of these changes with total H4, histone acetyltransferase, and histone deacetylase (HDAC) levels. Whereas the levels of H4 and histone acetyltransferases were stable irrespective of HRD status, the levels of histone H4 acetylation and one HDAC, HDAC6, were elevated in the HRD tumors. Relative H4 acetylation levels were also analyzed by an antibody-based approach in additional ovarian tumors. It is possible that specific H4 acetylation at Lys12 and Lys16 associated with HRD could inform chemotherapeutic treatment modalities to improve ovarian cancer patients' treatment response.
Collapse
Affiliation(s)
- Stefani N Thomas
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , Baltimore, Maryland 21231, United States
| | - Lijun Chen
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , Baltimore, Maryland 21231, United States
| | - Yang Liu
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , Baltimore, Maryland 21231, United States
| | - Naseruddin Höti
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , Baltimore, Maryland 21231, United States
| | - Hui Zhang
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , Baltimore, Maryland 21231, United States
| |
Collapse
|
25
|
Voigt A, Nowick K, Almaas E. A composite network of conserved and tissue specific gene interactions reveals possible genetic interactions in glioma. PLoS Comput Biol 2017; 13:e1005739. [PMID: 28957313 PMCID: PMC5634634 DOI: 10.1371/journal.pcbi.1005739] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 10/10/2017] [Accepted: 08/24/2017] [Indexed: 02/08/2023] Open
Abstract
Differential co-expression network analyses have recently become an important step in the investigation of cellular differentiation and dysfunctional gene-regulation in cell and tissue disease-states. The resulting networks have been analyzed to identify and understand pathways associated with disorders, or to infer molecular interactions. However, existing methods for differential co-expression network analysis are unable to distinguish between various forms of differential co-expression. To close this gap, here we define the three different kinds (conserved, specific, and differentiated) of differential co-expression and present a systematic framework, CSD, for differential co-expression network analysis that incorporates these interactions on an equal footing. In addition, our method includes a subsampling strategy to estimate the variance of co-expressions. Our framework is applicable to a wide variety of cases, such as the study of differential co-expression networks between healthy and disease states, before and after treatments, or between species. Applying the CSD approach to a published gene-expression data set of cerebral cortex and basal ganglia samples from healthy individuals, we find that the resulting CSD network is enriched in genes associated with cognitive function, signaling pathways involving compounds with well-known roles in the central nervous system, as well as certain neurological diseases. From the CSD analysis, we identify a set of prominent hubs of differential co-expression, whose neighborhood contains a substantial number of genes associated with glioblastoma. The resulting gene-sets identified by our CSD analysis also contain many genes that so far have not been recognized as having a role in glioblastoma, but are good candidates for further studies. CSD may thus aid in hypothesis-generation for functional disease-associations.
Collapse
Affiliation(s)
- André Voigt
- Network Systems Biology Group, Department of Biotechnology, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Katja Nowick
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
- Bioinformatics, Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
- Human Biology, Institute for Biology, Free University Berlin, Berlin, Germany
| | - Eivind Almaas
- Network Systems Biology Group, Department of Biotechnology, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and General Practice, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
26
|
Meng S, Liu G, Su L, Sun L, Wu D, Wang L, Zheng Z. Functional clusters analysis and research based on differential coexpression networks. BIOTECHNOL BIOTEC EQ 2017. [DOI: 10.1080/13102818.2017.1358669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Shuai Meng
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| | - Guixia Liu
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| | - Lingtao Su
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| | - Liyan Sun
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| | - Di Wu
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| | - Lingwei Wang
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| | - Zhao Zheng
- College of Computer Science and Technology, Jilin University, Changchun, PR China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, PR China
| |
Collapse
|
27
|
Anderson WD, DeCicco D, Schwaber JS, Vadigepalli R. A data-driven modeling approach to identify disease-specific multi-organ networks driving physiological dysregulation. PLoS Comput Biol 2017; 13:e1005627. [PMID: 28732007 PMCID: PMC5521738 DOI: 10.1371/journal.pcbi.1005627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/14/2017] [Indexed: 02/02/2023] Open
Abstract
Multiple physiological systems interact throughout the development of a complex disease. Knowledge of the dynamics and connectivity of interactions across physiological systems could facilitate the prevention or mitigation of organ damage underlying complex diseases, many of which are currently refractory to available therapeutics (e.g., hypertension). We studied the regulatory interactions operating within and across organs throughout disease development by integrating in vivo analysis of gene expression dynamics with a reverse engineering approach to infer data-driven dynamic network models of multi-organ gene regulatory influences. We obtained experimental data on the expression of 22 genes across five organs, over a time span that encompassed the development of autonomic nervous system dysfunction and hypertension. We pursued a unique approach for identification of continuous-time models that jointly described the dynamics and structure of multi-organ networks by estimating a sparse subset of ∼12,000 possible gene regulatory interactions. Our analyses revealed that an autonomic dysfunction-specific multi-organ sequence of gene expression activation patterns was associated with a distinct gene regulatory network. We analyzed the model structures for adaptation motifs, and identified disease-specific network motifs involving genes that exhibited aberrant temporal dynamics. Bioinformatic analyses identified disease-specific single nucleotide variants within or near transcription factor binding sites upstream of key genes implicated in maintaining physiological homeostasis. Our approach illustrates a novel framework for investigating the pathogenesis through model-based analysis of multi-organ system dynamics and network properties. Our results yielded novel candidate molecular targets driving the development of cardiovascular disease, metabolic syndrome, and immune dysfunction. Complex diseases such as hypertension often involve maladaptive autonomic nervous system control over the cardiovascular, renal, hepatic, immune, and endocrine systems. We studied the pathogenesis of physiological homeostasis by examining the temporal dynamics of gene expression levels from multiple organs in an animal model of autonomic dysfunction characterized by cardiovascular disease, metabolic dysregulation, and immune system aberrations. We employed a data-driven modeling approach to jointly predict continuous gene expression dynamics and gene regulatory interactions across organs in the disease and control phenotypes. We combined our analyses of multi-organ gene regulatory network dynamics and connectivity with bioinformatic analyses of genetic mutations that could regulate gene expression. Our multi-organ modeling approach to investigate the mechanisms of complex disease pathogenesis revealed novel candidates for therapeutic interventions against the development and progression of complex diseases involving autonomic nervous system dysfunction.
Collapse
Affiliation(s)
- Warren D. Anderson
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Danielle DeCicco
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - James S. Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- * E-mail:
| |
Collapse
|
28
|
Arro J, Cuenca J, Yang Y, Liang Z, Cousins P, Zhong GY. A transcriptome analysis of two grapevine populations segregating for tendril phyllotaxy. HORTICULTURE RESEARCH 2017; 4:17032. [PMID: 28713572 PMCID: PMC5506248 DOI: 10.1038/hortres.2017.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/16/2017] [Accepted: 06/07/2017] [Indexed: 06/01/2023]
Abstract
The shoot structure of cultivated grapevine Vitis vinifera L. typically exhibits a three-node modular repetitive pattern, two sequential leaf-opposed tendrils followed by a tendril-free node. In this study, we investigated the molecular basis of this pattern by characterizing differentially expressed genes in 10 bulk samples of young tendril tissue from two grapevine populations showing segregation of mutant or wild-type shoot/tendril phyllotaxy. One population was the selfed progeny and the other one, an outcrossed progeny of a Vitis hybrid, 'Roger's Red'. We analyzed 13 375 expressed genes and carried out in-depth analyses of 324 of them, which were differentially expressed with a minimum of 1.5-fold changes between the mutant and wild-type bulk samples in both selfed and cross populations. A significant portion of these genes were direct cis-binding targets of 14 transcription factor families that were themselves differentially expressed. Network-based dependency analysis further revealed that most of the significantly rewired connections among the 10 most connected hub genes involved at least one transcription factor. TCP3 and MYB12, which were known important for plant-form development, were among these transcription factors. More importantly, TCP3 and MYB12 were found in this study to be involved in regulating the lignin gene PRX52, which is important to plant-form development. A further support evidence for the roles of TCP3-MYB12-PRX52 in contributing to tendril phyllotaxy was the findings of two other lignin-related genes uniquely expressed in the mutant phyllotaxy background.
Collapse
Affiliation(s)
- Jie Arro
- USDA-Agricultural Research Service, Grape Genetics Research Unit, Geneva, NY 14456, USA
| | - Jose Cuenca
- USDA-Agricultural Research Service, Grape Genetics Research Unit, Geneva, NY 14456, USA
| | - Yingzhen Yang
- USDA-Agricultural Research Service, Grape Genetics Research Unit, Geneva, NY 14456, USA
| | - Zhenchang Liang
- Beijing Key Laboratory of Grape Science and Enology and Key Laboratory of Plant Resource, Institute of Botany, the Chinese Academy of Sciences, Beijing 100093, People’s Republic of China
| | | | - Gan-Yuan Zhong
- USDA-Agricultural Research Service, Grape Genetics Research Unit, Geneva, NY 14456, USA
| |
Collapse
|
29
|
Incorporating prior biological knowledge for network-based differential gene expression analysis using differentially weighted graphical LASSO. BMC Bioinformatics 2017; 18:99. [PMID: 28187708 PMCID: PMC5303311 DOI: 10.1186/s12859-017-1515-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 01/31/2017] [Indexed: 11/10/2022] Open
Abstract
Background Conventional differential gene expression analysis by methods such as student’s t-test, SAM, and Empirical Bayes often searches for statistically significant genes without considering the interactions among them. Network-based approaches provide a natural way to study these interactions and to investigate the rewiring interactions in disease versus control groups. In this paper, we apply weighted graphical LASSO (wgLASSO) algorithm to integrate a data-driven network model with prior biological knowledge (i.e., protein-protein interactions) for biological network inference. We propose a novel differentially weighted graphical LASSO (dwgLASSO) algorithm that builds group-specific networks and perform network-based differential gene expression analysis to select biomarker candidates by considering their topological differences between the groups. Results Through simulation, we showed that wgLASSO can achieve better performance in building biologically relevant networks than purely data-driven models (e.g., neighbor selection, graphical LASSO), even when only a moderate level of information is available as prior biological knowledge. We evaluated the performance of dwgLASSO for survival time prediction using two microarray breast cancer datasets previously reported by Bild et al. and van de Vijver et al. Compared with the top 10 significant genes selected by conventional differential gene expression analysis method, the top 10 significant genes selected by dwgLASSO in the dataset from Bild et al. led to a significantly improved survival time prediction in the independent dataset from van de Vijver et al. Among the 10 genes selected by dwgLASSO, UBE2S, SALL2, XBP1 and KIAA0922 have been confirmed by literature survey to be highly relevant in breast cancer biomarker discovery study. Additionally, we tested dwgLASSO on TCGA RNA-seq data acquired from patients with hepatocellular carcinoma (HCC) on tumors samples and their corresponding non-tumorous liver tissues. Improved sensitivity, specificity and area under curve (AUC) were observed when comparing dwgLASSO with conventional differential gene expression analysis method. Conclusions The proposed network-based differential gene expression analysis algorithm dwgLASSO can achieve better performance than conventional differential gene expression analysis methods by integrating information at both gene expression and network topology levels. The incorporation of prior biological knowledge can lead to the identification of biologically meaningful genes in cancer biomarker studies. Electronic supplementary material The online version of this article (doi:10.1186/s12859-017-1515-1) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Speyer G, Mahendra D, Tran HJ, Kiefer J, Schreiber SL, Clemons PA, Dhruv H, Berens M, Kim S. DIFFERENTIAL PATHWAY DEPENDENCY DISCOVERY ASSOCIATED WITH DRUG RESPONSE ACROSS CANCER CELL LINES. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2017; 22:497-508. [PMID: 27897001 PMCID: PMC5180601 DOI: 10.1142/9789813207813_0046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effort to personalize treatment plans for cancer patients involves the identification of drug treatments that can effectively target the disease while minimizing the likelihood of adverse reactions. In this study, the gene-expression profile of 810 cancer cell lines and their response data to 368 small molecules from the Cancer Therapeutics Research Portal (CTRP) are analyzed to identify pathways with significant rewiring between genes, or differential gene dependency, between sensitive and non-sensitive cell lines. Identified pathways and their corresponding differential dependency networks are further analyzed to discover essentiality and specificity mediators of cell line response to drugs/compounds. For analysis we use the previously published method EDDY (Evaluation of Differential DependencY). EDDY first constructs likelihood distributions of gene-dependency networks, aided by known genegene interaction, for two given conditions, for example, sensitive cell lines vs. non-sensitive cell lines. These sets of networks yield a divergence value between two distributions of network likelihoods that can be assessed for significance using permutation tests. Resulting differential dependency networks are then further analyzed to identify genes, termed mediators, which may play important roles in biological signaling in certain cell lines that are sensitive or non-sensitive to the drugs. Establishing statistical correspondence between compounds and mediators can improve understanding of known gene dependencies associated with drug response while also discovering new dependencies. Millions of compute hours resulted in thousands of these statistical discoveries. EDDY identified 8,811 statistically significant pathways leading to 26,822 compound-pathway-mediator triplets. By incorporating STITCH and STRING databases, we could construct evidence networks for 14,415 compound-pathway-mediator triplets for support. The results of this analysis are presented in a searchable website to aid researchers in studying potential molecular mechanisms underlying cells' drug response as well as in designing experiments for the purpose of personalized treatment regimens.
Collapse
Affiliation(s)
- Gil Speyer
- The Translational Genomics Research Institute, Phoenix, AZ 85004, U.S.A.,
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Developing improved approaches for diagnosis, treatment, and prevention of diseases is a major goal of biomedical research. Therefore, the discovery of biomarker signatures from high-throughput "omics" data is an active research topic in the field of bioinformatics and systems medicine. A major issue is the low reproducibility and the limited biological interpretability of candidate biomarker signatures identified from high-throughput data. This impedes the use of discovered biomarker signatures into clinical applications. Currently, much focus is placed on developing strategies to improve reproducibility and interpretability. Researchers have fruitfully started to incorporate prior knowledge derived from pathways and molecular networks into the process of biomarker identification. In this chapter, after giving a general introduction to the problem of disease classification and biomarker discovery, we will review two types of network-assisted approaches: (1) approaches inferring activity scores for specific pathways which are subsequently used for classification and (2) approaches identifying subnetworks or modules of molecular networks by differential network analysis which can serve as biomarker signatures.
Collapse
|
32
|
A powerful weighted statistic for detecting group differences of directed biological networks. Sci Rep 2016; 6:34159. [PMID: 27686331 PMCID: PMC5054825 DOI: 10.1038/srep34159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/08/2016] [Indexed: 12/15/2022] Open
Abstract
Complex disease is largely determined by a number of biomolecules interwoven into networks, rather than a single biomolecule. Different physiological conditions such as cases and controls may manifest as different networks. Statistical comparison between biological networks can provide not only new insight into the disease mechanism but statistical guidance for drug development. However, the methods developed in previous studies are inadequate to capture the changes in both the nodes and edges, and often ignore the network structure. In this study, we present a powerful weighted statistical test for group differences of directed biological networks, which is independent of the network attributes and can capture the changes in both the nodes and edges, as well as simultaneously accounting for the network structure through putting more weights on the difference of nodes locating on relatively more important position. Simulation studies illustrate that this method had better performance than previous ones under various sample sizes and network structures. One application to GWAS of leprosy successfully identifies the specific gene interaction network contributing to leprosy. Another real data analysis significantly identifies a new biological network, which is related to acute myeloid leukemia. One potential network responsible for lung cancer has also been significantly detected. The source R code is available on our website.
Collapse
|
33
|
Hilakivi-Clarke L, Wärri A, Bouker KB, Zhang X, Cook KL, Jin L, Zwart A, Nguyen N, Hu R, Cruz MI, de Assis S, Wang X, Xuan J, Wang Y, Wehrenberg B, Clarke R. Effects of In Utero Exposure to Ethinyl Estradiol on Tamoxifen Resistance and Breast Cancer Recurrence in a Preclinical Model. J Natl Cancer Inst 2016; 109:2905688. [PMID: 27609189 DOI: 10.1093/jnci/djw188] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 07/19/2016] [Indexed: 12/13/2022] Open
Abstract
Background Responses to endocrine therapies vary among patients with estrogen receptor (ER+) breast cancer. We studied whether in utero exposure to endocrine-disrupting compounds might explain these variations. Methods We describe a novel ER+ breast cancer model to study de novo and acquired tamoxifen (TAM) resistance. Pregnant Sprague Dawley rats were exposed to 0 or 0.1 ppm ethinyl estradiol (EE2), and the response of 9,12-dimethylbenz[a]anthracene (DMBA)-induced mammary tumors to 15 mg/kg TAM, with (n = 17 tumors in the controls and n = 20 tumors in EE2 offspring) or without 1.2 g/kg valproic acid and 5 mg/kg hydralazine (n = 24 tumors in the controls and n = 32 tumors in EE2 offspring) in the female offspring, was assessed. One-sided Chi2 tests were used to calculate P values. Comparisons of differentially expressed genes between mammary tumors in in utero EE2-exposed and control rats, and between anti-estrogen-resistant LCC9 and -sensitive LCC1 human breast cancer cells, were also performed. Results In our preclinical model, 54.2% of mammary tumors in the control rats exhibited a complete response to TAM, of which 23.1% acquired resistance with continued anti-estrogen treatment and recurred. Mammary tumors in the EE2 offspring were statistically significantly less likely to respond to TAM (P = .047) and recur (P = .007). In the EE2 offspring, but not in controls, adding valproic acid and hydralazine to TAM prevented recurrence (P < .001). Three downregulated and hypermethylated genes (KLF4, LGALS3, MICB) and one upregulated gene (ETV4) were identified in EE2 tumors and LCC9 breast cancer cells, and valproic acid and hydralazine normalized the altered expression of all four genes. Conclusions Resistance to TAM may be preprogrammed by in utero exposure to high estrogen levels and mediated through reversible epigenetic alterations in genes associated with epithelial-mesenchymal transition and tumor immune responses.
Collapse
Affiliation(s)
| | - Anni Wärri
- Department of Oncology, Georgetown University, Washington, DC.,Institute of Biomedicine, University of Turku Medical Faculty, Turku, Finland
| | - Kerrie B Bouker
- Department of Oncology, Georgetown University, Washington, DC
| | - Xiyuan Zhang
- Department of Oncology, Georgetown University, Washington, DC
| | - Katherine L Cook
- Department of Oncology, Georgetown University, Washington, DC.,Department of Surgery, Wake Forest University, Winston-Salem, NC
| | - Lu Jin
- Department of Oncology, Georgetown University, Washington, DC
| | - Alan Zwart
- Department of Oncology, Georgetown University, Washington, DC
| | - Nguyen Nguyen
- Department of Oncology, Georgetown University, Washington, DC
| | - Rong Hu
- Department of Oncology, Georgetown University, Washington, DC
| | - M Idalia Cruz
- Department of Oncology, Georgetown University, Washington, DC
| | - Sonia de Assis
- Department of Oncology, Georgetown University, Washington, DC
| | - Xiao Wang
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA
| | - Jason Xuan
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA
| | | | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, DC
| |
Collapse
|
34
|
Yan W, Xue W, Chen J, Hu G. Biological Networks for Cancer Candidate Biomarkers Discovery. Cancer Inform 2016; 15:1-7. [PMID: 27625573 PMCID: PMC5012434 DOI: 10.4137/cin.s39458] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/06/2016] [Accepted: 06/16/2016] [Indexed: 12/16/2022] Open
Abstract
Due to its extraordinary heterogeneity and complexity, cancer is often proposed as a model case of a systems biology disease or network disease. There is a critical need of effective biomarkers for cancer diagnosis and/or outcome prediction from system level analyses. Methods based on integrating omics data into networks have the potential to revolutionize the identification of cancer biomarkers. Deciphering the biological networks underlying cancer is undoubtedly important for understanding the molecular mechanisms of the disease and identifying effective biomarkers. In this review, the networks constructed for cancer biomarker discovery based on different omics level data are described and illustrated from recent advances in the field.
Collapse
Affiliation(s)
- Wenying Yan
- Center for Systems Biology, Soochow University, Suzhou, Jiangsu, China
| | - Wenjin Xue
- Department of Electrical Engineering, Technician College of Taizhou, Taizhou, Jiangsu, China
| | - Jiajia Chen
- School of Chemistry, Biology and Material Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Guang Hu
- Center for Systems Biology, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
35
|
Zuo Y, Cui Y, Di Poto C, Varghese RS, Yu G, Li R, Ressom HW. INDEED: Integrated differential expression and differential network analysis of omic data for biomarker discovery. Methods 2016; 111:12-20. [PMID: 27592383 DOI: 10.1016/j.ymeth.2016.08.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 01/03/2023] Open
Abstract
Differential expression (DE) analysis is commonly used to identify biomarker candidates that have significant changes in their expression levels between distinct biological groups. One drawback of DE analysis is that it only considers the changes on single biomolecule level. Recently, differential network (DN) analysis has become popular due to its capability to measure the changes on biomolecular pair level. In DN analysis, network is typically built based on correlation and biomarker candidates are selected by investigating the network topology. However, correlation tends to generate over-complicated networks and the selection of biomarker candidates purely based on network topology ignores the changes on single biomolecule level. In this paper, we propose a novel approach, INDEED, that builds sparse differential network based on partial correlation and integrates DE and DN analyses for biomarker discovery. We applied this approach on real proteomic and glycomic data generated by liquid chromatography coupled with mass spectrometry for hepatocellular carcinoma (HCC) biomarker discovery study. For each omic data, we used one dataset to select biomarker candidates, built a disease classifier and evaluated the performance of the classifier on an independent dataset. The biomarker candidates, selected by INDEED, were more reproducible across independent datasets, and led to a higher classification accuracy in predicting HCC cases and cirrhotic controls compared with those selected by separate DE and DN analyses. INDEED also identified some candidates previously reported to be relevant to HCC, such as intercellular adhesion molecule 2 (ICAM2) and c4b-binding protein alpha chain (C4BPA), which were missed by both DE and DN analyses. In addition, we applied INDEED for survival time prediction based on transcriptomic data acquired by analysis of samples from breast cancer patients. We selected biomarker candidates and built a regression model for survival time prediction based on a gene expression dataset and patients' survival records. We evaluated the performance of the regression model on an independent dataset. Compared with the biomarker candidates selected by DE and DN analyses, those selected through INDEED led to more accurate survival time prediction.
Collapse
Affiliation(s)
- Yiming Zuo
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA; Department of Radiation Oncology, Stanford University, Palo Alto, CA 94304, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA.
| | - Yi Cui
- Department of Radiation Oncology, Stanford University, Palo Alto, CA 94304, USA.
| | - Cristina Di Poto
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA.
| | - Rency S Varghese
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA.
| | - Guoqiang Yu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA.
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University, Palo Alto, CA 94304, USA.
| | - Habtom W Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA.
| |
Collapse
|
36
|
Differential Regulatory Analysis Based on Coexpression Network in Cancer Research. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4241293. [PMID: 27597964 PMCID: PMC4997028 DOI: 10.1155/2016/4241293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 12/15/2022]
Abstract
With rapid development of high-throughput techniques and accumulation of big transcriptomic data, plenty of computational methods and algorithms such as differential analysis and network analysis have been proposed to explore genome-wide gene expression characteristics. These efforts are aiming to transform underlying genomic information into valuable knowledges in biological and medical research fields. Recently, tremendous integrative research methods are dedicated to interpret the development and progress of neoplastic diseases, whereas differential regulatory analysis (DRA) based on gene coexpression network (GCN) increasingly plays a robust complement to regular differential expression analysis in revealing regulatory functions of cancer related genes such as evading growth suppressors and resisting cell death. Differential regulatory analysis based on GCN is prospective and shows its essential role in discovering the system properties of carcinogenesis features. Here we briefly review the paradigm of differential regulatory analysis based on GCN. We also focus on the applications of differential regulatory analysis based on GCN in cancer research and point out that DRA is necessary and extraordinary to reveal underlying molecular mechanism in large-scale carcinogenesis studies.
Collapse
|
37
|
Wan S, Mak MW, Kung SY. Mem-mEN: Predicting Multi-Functional Types of Membrane Proteins by Interpretable Elastic Nets. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2016; 13:706-718. [PMID: 26336143 DOI: 10.1109/tcbb.2015.2474407] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Membrane proteins play important roles in various biological processes within organisms. Predicting the functional types of membrane proteins is indispensable to the characterization of membrane proteins. Recent studies have extended to predicting single- and multi-type membrane proteins. However, existing predictors perform poorly and more importantly, they are often lack of interpretability. To address these problems, this paper proposes an efficient predictor, namely Mem-mEN, which can produce sparse and interpretable solutions for predicting membrane proteins with single- and multi-label functional types. Given a query membrane protein, its associated gene ontology (GO) information is retrieved by searching a compact GO-term database with its homologous accession number, which is subsequently classified by a multi-label elastic net (EN) classifier. Experimental results show that Mem-mEN significantly outperforms existing state-of-the-art membrane-protein predictors. Moreover, by using Mem-mEN, 338 out of more than 7,900 GO terms are found to play more essential roles in determining the functional types. Based on these 338 essential GO terms, Mem-mEN can not only predict the functional type of a membrane protein, but also explain why it belongs to that type. For the reader's convenience, the Mem-mEN server is available online at http://bioinfo.eie.polyu.edu.hk/MemmENServer/.
Collapse
|
38
|
Integrated Proteogenomic Characterization of Human High-Grade Serous Ovarian Cancer. Cell 2016; 166:755-765. [PMID: 27372738 PMCID: PMC4967013 DOI: 10.1016/j.cell.2016.05.069] [Citation(s) in RCA: 702] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/05/2016] [Accepted: 05/19/2016] [Indexed: 12/22/2022]
Abstract
To provide a detailed analysis of the molecular components and underlying mechanisms associated with ovarian cancer, we performed a comprehensive mass-spectrometry-based proteomic characterization of 174 ovarian tumors previously analyzed by The Cancer Genome Atlas (TCGA), of which 169 were high-grade serous carcinomas (HGSCs). Integrating our proteomic measurements with the genomic data yielded a number of insights into disease, such as how different copy-number alternations influence the proteome, the proteins associated with chromosomal instability, the sets of signaling pathways that diverse genome rearrangements converge on, and the ones most associated with short overall survival. Specific protein acetylations associated with homologous recombination deficiency suggest a potential means for stratifying patients for therapy. In addition to providing a valuable resource, these findings provide a view of how the somatic genome drives the cancer proteome and associations between protein and post-translational modification levels and clinical outcomes in HGSC. VIDEO ABSTRACT.
Collapse
|
39
|
Grechkin M, Logsdon BA, Gentles AJ, Lee SI. Identifying Network Perturbation in Cancer. PLoS Comput Biol 2016; 12:e1004888. [PMID: 27145341 PMCID: PMC4856318 DOI: 10.1371/journal.pcbi.1004888] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/25/2016] [Indexed: 01/08/2023] Open
Abstract
We present a computational framework, called DISCERN (DIfferential SparsE Regulatory Network), to identify informative topological changes in gene-regulator dependence networks inferred on the basis of mRNA expression datasets within distinct biological states. DISCERN takes two expression datasets as input: an expression dataset of diseased tissues from patients with a disease of interest and another expression dataset from matching normal tissues. DISCERN estimates the extent to which each gene is perturbed-having distinct regulator connectivity in the inferred gene-regulator dependencies between the disease and normal conditions. This approach has distinct advantages over existing methods. First, DISCERN infers conditional dependencies between candidate regulators and genes, where conditional dependence relationships discriminate the evidence for direct interactions from indirect interactions more precisely than pairwise correlation. Second, DISCERN uses a new likelihood-based scoring function to alleviate concerns about accuracy of the specific edges inferred in a particular network. DISCERN identifies perturbed genes more accurately in synthetic data than existing methods to identify perturbed genes between distinct states. In expression datasets from patients with acute myeloid leukemia (AML), breast cancer and lung cancer, genes with high DISCERN scores in each cancer are enriched for known tumor drivers, genes associated with the biological processes known to be important in the disease, and genes associated with patient prognosis, in the respective cancer. Finally, we show that DISCERN can uncover potential mechanisms underlying network perturbation by explaining observed epigenomic activity patterns in cancer and normal tissue types more accurately than alternative methods, based on the available epigenomic data from the ENCODE project.
Collapse
Affiliation(s)
- Maxim Grechkin
- Department of Computer Science & Engineering, University of Washington, Seattle, Washington, United States of America
| | | | - Andrew J. Gentles
- Center for Cancer Systems Biology, Department of Radiology, Stanford University, Stanford, California, United States of America
| | - Su-In Lee
- Department of Computer Science & Engineering, University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
40
|
Wan S, Mak MW, Kung SY. Sparse regressions for predicting and interpreting subcellular localization of multi-label proteins. BMC Bioinformatics 2016; 17:97. [PMID: 26911432 PMCID: PMC4765148 DOI: 10.1186/s12859-016-0940-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 01/27/2016] [Indexed: 11/10/2022] Open
Abstract
Background Predicting protein subcellular localization is indispensable for inferring protein functions. Recent studies have been focusing on predicting not only single-location proteins, but also multi-location proteins. Almost all of the high performing predictors proposed recently use gene ontology (GO) terms to construct feature vectors for classification. Despite their high performance, their prediction decisions are difficult to interpret because of the large number of GO terms involved. Results This paper proposes using sparse regressions to exploit GO information for both predicting and interpreting subcellular localization of single- and multi-location proteins. Specifically, we compared two multi-label sparse regression algorithms, namely multi-label LASSO (mLASSO) and multi-label elastic net (mEN), for large-scale predictions of protein subcellular localization. Both algorithms can yield sparse and interpretable solutions. By using the one-vs-rest strategy, mLASSO and mEN identified 87 and 429 out of more than 8,000 GO terms, respectively, which play essential roles in determining subcellular localization. More interestingly, many of the GO terms selected by mEN are from the biological process and molecular function categories, suggesting that the GO terms of these categories also play vital roles in the prediction. With these essential GO terms, not only where a protein locates can be decided, but also why it resides there can be revealed. Conclusions Experimental results show that the output of both mEN and mLASSO are interpretable and they perform significantly better than existing state-of-the-art predictors. Moreover, mEN selects more features and performs better than mLASSO on a stringent human benchmark dataset. For readers’ convenience, an online server called SpaPredictor for both mLASSO and mEN is available at http://bioinfo.eie.polyu.edu.hk/SpaPredictorServer/.
Collapse
Affiliation(s)
- Shibiao Wan
- Department of Electronic and Information Engineering, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| | - Man-Wai Mak
- Department of Electronic and Information Engineering, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| | - Sun-Yuan Kung
- Department of Electrical Engineering, Princeton University, New Jersey, USA.
| |
Collapse
|
41
|
Ji J, Yuan Z, Zhang X, Xue F. A powerful score-based statistical test for group difference in weighted biological networks. BMC Bioinformatics 2016; 17:86. [PMID: 26867929 PMCID: PMC4751708 DOI: 10.1186/s12859-016-0916-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/29/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Complex disease is largely determined by a number of biomolecules interwoven into networks, rather than a single biomolecule. A key but inadequately addressed issue is how to test possible differences of the networks between two groups. Group-level comparison of network properties may shed light on underlying disease mechanisms and benefit the design of drug targets for complex diseases. We therefore proposed a powerful score-based statistic to detect group difference in weighted networks, which simultaneously capture the vertex changes and edge changes. RESULTS Simulation studies indicated that the proposed network difference measure (NetDifM) was stable and outperformed other methods existed, under various sample sizes and network topology structure. One application to real data about GWAS of leprosy successfully identified the specific gene interaction network contributing to leprosy. For additional gene expression data of ovarian cancer, two candidate subnetworks, PI3K-AKT and Notch signaling pathways, were considered and identified respectively. CONCLUSIONS The proposed method, accounting for the vertex changes and edge changes simultaneously, is valid and powerful to capture the group difference of biological networks.
Collapse
Affiliation(s)
- Jiadong Ji
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| | - Xiaoshuai Zhang
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, PO Box 100, Jinan, 250012, Shandong, China.
| |
Collapse
|
42
|
Van Landeghem S, Van Parys T, Dubois M, Inzé D, Van de Peer Y. Diffany: an ontology-driven framework to infer, visualise and analyse differential molecular networks. BMC Bioinformatics 2016; 17:18. [PMID: 26729218 PMCID: PMC4700732 DOI: 10.1186/s12859-015-0863-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/17/2015] [Indexed: 11/30/2022] Open
Abstract
Background Differential networks have recently been introduced as a powerful way to study the dynamic rewiring capabilities of an interactome in response to changing environmental conditions or stimuli. Currently, such differential networks are generated and visualised using ad hoc methods, and are often limited to the analysis of only one condition-specific response or one interaction type at a time. Results In this work, we present a generic, ontology-driven framework to infer, visualise and analyse an arbitrary set of condition-specific responses against one reference network. To this end, we have implemented novel ontology-based algorithms that can process highly heterogeneous networks, accounting for both physical interactions and regulatory associations, symmetric and directed edges, edge weights and negation. We propose this integrative framework as a standardised methodology that allows a unified view on differential networks and promotes comparability between differential network studies. As an illustrative application, we demonstrate its usefulness on a plant abiotic stress study and we experimentally confirmed a predicted regulator. Availability Diffany is freely available as open-source java library and Cytoscape plugin from http://bioinformatics.psb.ugent.be/supplementary_data/solan/diffany/. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0863-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofie Van Landeghem
- Department of Plant Systems Biology, VIB, Technologiepark 927, Ghent, 9052, Belgium. .,Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, Ghent, 9052, Belgium.
| | - Thomas Van Parys
- Department of Plant Systems Biology, VIB, Technologiepark 927, Ghent, 9052, Belgium. .,Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, Ghent, 9052, Belgium.
| | - Marieke Dubois
- Department of Plant Systems Biology, VIB, Technologiepark 927, Ghent, 9052, Belgium. .,Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, Ghent, 9052, Belgium.
| | - Dirk Inzé
- Department of Plant Systems Biology, VIB, Technologiepark 927, Ghent, 9052, Belgium. .,Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, Ghent, 9052, Belgium.
| | - Yves Van de Peer
- Department of Plant Systems Biology, VIB, Technologiepark 927, Ghent, 9052, Belgium. .,Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, Ghent, 9052, Belgium. .,Genomics Research Institute, University of Pretoria, Private bag X200028, Pretoria, South Africa.
| |
Collapse
|
43
|
Speyer G, Kiefer J, Dhruv H, Berens M, Kim S. KNOWLEDGE-ASSISTED APPROACH TO IDENTIFY PATHWAYS WITH DIFFERENTIAL DEPENDENCIES. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2016; 21:33-44. [PMID: 26776171 PMCID: PMC4721243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
We have previously developed a statistical method to identify gene sets enriched with condition-specific genetic dependencies. The method constructs gene dependency networks from bootstrapped samples in one condition and computes the divergence between distributions of network likelihood scores from different conditions. It was shown to be capable of sensitive and specific identification of pathways with phenotype-specific dysregulation, i.e., rewiring of dependencies between genes in different conditions. We now present an extension of the method by incorporating prior knowledge into the inference of networks. The degree of prior knowledge incorporation has substantial effect on the sensitivity of the method, as the data is the source of condition specificity while prior knowledge incorporation can provide additional support for dependencies that are only partially supported by the data. Use of prior knowledge also significantly improved the interpretability of the results. Further analysis of topological characteristics of gene differential dependency networks provides a new approach to identify genes that could play important roles in biological signaling in a specific condition, hence, promising targets customized to a specific condition. Through analysis of TCGA glioblastoma multiforme data, we demonstrate the method can identify not only potentially promising targets but also underlying biology for new targets.
Collapse
Affiliation(s)
- Gil Speyer
- Integrated Cancer Genomics Division, The Translational Genomics
Research Institute, Phoenix, AZ 85004, U.S.A
| | - Jeff Kiefer
- Integrated Cancer Genomics Division, The Translational Genomics
Research Institute, Phoenix, AZ 85004, U.S.A
| | - Harshil Dhruv
- Cancer Cell Biology Division, The Translational Genomics Research
Institute, Phoenix, AZ 85004, U.S.A
| | - Michael Berens
- Cancer Cell Biology Division, The Translational Genomics Research
Institute, Phoenix, AZ 85004, U.S.A
| | - Seungchan Kim
- Integrated Cancer Genomics Division, The Translational Genomics
Research Institute, Phoenix, AZ 85004, U.S.A
| |
Collapse
|
44
|
Clarke R, Tyson JJ, Dixon JM. Endocrine resistance in breast cancer--An overview and update. Mol Cell Endocrinol 2015; 418 Pt 3:220-34. [PMID: 26455641 PMCID: PMC4684757 DOI: 10.1016/j.mce.2015.09.035] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
Abstract
Tumors that express detectable levels of the product of the ESR1 gene (estrogen receptor-α; ERα) represent the single largest molecular subtype of breast cancer. More women eventually die from ERα+ breast cancer than from either HER2+ disease (almost half of which also express ERα) and/or from triple negative breast cancer (ERα-negative, progesterone receptor-negative, and HER2-negative). Antiestrogens and aromatase inhibitors are largely indistinguishable from each other in their abilities to improve overall survival and almost 50% of ERα+ breast cancers will eventually fail one or more of these endocrine interventions. The precise reasons why these therapies fail in ERα+ breast cancer remain largely unknown. Pharmacogenetic explanations for Tamoxifen resistance are controversial. The role of ERα mutations in endocrine resistance remains unclear. Targeting the growth factors and oncogenes most strongly correlated with endocrine resistance has proven mostly disappointing in their abilities to improve overall survival substantially, particularly in the metastatic setting. Nonetheless, there are new concepts in endocrine resistance that integrate molecular signaling, cellular metabolism, and stress responses including endoplasmic reticulum stress and the unfolded protein response (UPR) that provide novel insights and suggest innovative therapeutic targets. Encouraging evidence that drug combinations with CDK4/CDK6 inhibitors can extend recurrence free survival may yet translate to improvements in overall survival. Whether the improvements seen with immunotherapy in other cancers can be achieved in breast cancer remains to be determined, particularly for ERα+ breast cancers. This review explores the basic mechanisms of resistance to endocrine therapies, concluding with some new insights from systems biology approaches further implicating autophagy and the UPR in detail, and a brief discussion of exciting new avenues and future prospects.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown University Medical Center, Washington DC 20057, USA.
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic and State University, Blacksburg, VA 24061, USA
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| |
Collapse
|
45
|
Wan S, Mak MW, Kung SY. mLASSO-Hum: A LASSO-based interpretable human-protein subcellular localization predictor. J Theor Biol 2015; 382:223-34. [DOI: 10.1016/j.jtbi.2015.06.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 02/03/2023]
|
46
|
Affiliation(s)
- Bai Zhang
- From the Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD (B.Z., Z.Z.); the Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD (Z.Z.); and Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington (Y.T.)
| | - Ye Tian
- From the Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD (B.Z., Z.Z.); the Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD (Z.Z.); and Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington (Y.T.)
| | - Zhen Zhang
- From the Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD (B.Z., Z.Z.); the Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD (Z.Z.); and Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington (Y.T.).
| |
Collapse
|
47
|
Cao MS, Liu BY, Dai WT, Zhou WX, Li YX, Li YY. Differential network analysis reveals dysfunctional regulatory networks in gastric carcinogenesis. Am J Cancer Res 2015; 5:2605-2625. [PMID: 26609471 PMCID: PMC4633893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 08/04/2015] [Indexed: 06/05/2023] Open
Abstract
Gastric Carcinoma is one of the most common cancers in the world. A large number of differentially expressed genes have been identified as being associated with gastric cancer progression, however, little is known about the underlying regulatory mechanisms. To address this problem, we developed a differential networking approach that is characterized by including a nascent methodology, differential coexpression analysis (DCEA), and two novel quantitative methods for differential regulation analysis. We first applied DCEA to a gene expression dataset of gastric normal mucosa, adenoma and carcinoma samples to identify gene interconnection changes during cancer progression, based on which we inferred normal, adenoma, and carcinoma-specific gene regulation networks by using linear regression model. It was observed that cancer genes and drug targets were enriched in each network. To investigate the dynamic changes of gene regulation during carcinogenesis, we then designed two quantitative methods to prioritize differentially regulated genes (DRGs) and gene pairs or links (DRLs) between adjacent stages. It was found that known cancer genes and drug targets are significantly higher ranked. The top 4% normal vs. adenoma DRGs (36 genes) and top 6% adenoma vs. carcinoma DRGs (56 genes) proved to be worthy of further investigation to explore their association with gastric cancer. Out of the 16 DRGs involved in two top-10 DRG lists of normal vs. adenoma and adenoma vs. carcinoma comparisons, 15 have been reported to be gastric cancer or cancer related. Based on our inferred differential networking information and known signaling pathways, we generated testable hypotheses on the roles of GATA6, ESRRG and their signaling pathways in gastric carcinogenesis. Compared with established approaches which build genome-scale GRNs, or sub-networks around differentially expressed genes, the present one proved to be better at enriching cancer genes and drug targets, and prioritizing disease-related genes on the dataset we considered. We propose this extendable differential networking framework as a promising way to gain insights into gene regulatory mechanisms underlying cancer progression and other phenotypic changes.
Collapse
Affiliation(s)
- Mu-Shui Cao
- School of Life Science and Technology, Tongji UniversityShanghai 200092, P. R. China
- Shanghai Center for Bioinformation TechnologyShanghai 200235, P. R. China
- Shanghai Industrial Technology Institute1278 Keyuan Road, Shanghai 201203, P. R. China
| | - Bing-Ya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, P. R. China
| | - Wen-Tao Dai
- Shanghai Center for Bioinformation TechnologyShanghai 200235, P. R. China
- Shanghai Industrial Technology Institute1278 Keyuan Road, Shanghai 201203, P. R. China
| | - Wei-Xin Zhou
- Shanghai Center for Bioinformation TechnologyShanghai 200235, P. R. China
- Shanghai Industrial Technology Institute1278 Keyuan Road, Shanghai 201203, P. R. China
- Shanghai Engineering Research Center of Pharmaceutical Translation1278 Keyuan Road, Shanghai 201203, P. R. China
| | - Yi-Xue Li
- School of Life Science and Technology, Tongji UniversityShanghai 200092, P. R. China
- Shanghai Center for Bioinformation TechnologyShanghai 200235, P. R. China
- Shanghai Industrial Technology Institute1278 Keyuan Road, Shanghai 201203, P. R. China
- Shanghai Engineering Research Center of Pharmaceutical Translation1278 Keyuan Road, Shanghai 201203, P. R. China
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation TechnologyShanghai 200235, P. R. China
- Shanghai Industrial Technology Institute1278 Keyuan Road, Shanghai 201203, P. R. China
- Shanghai Engineering Research Center of Pharmaceutical Translation1278 Keyuan Road, Shanghai 201203, P. R. China
| |
Collapse
|
48
|
Ha MJ, Baladandayuthapani V, Do KA. DINGO: differential network analysis in genomics. Bioinformatics 2015; 31:3413-20. [PMID: 26148744 DOI: 10.1093/bioinformatics/btv406] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 06/26/2015] [Indexed: 12/22/2022] Open
Abstract
MOTIVATION Cancer progression and development are initiated by aberrations in various molecular networks through coordinated changes across multiple genes and pathways. It is important to understand how these networks change under different stress conditions and/or patient-specific groups to infer differential patterns of activation and inhibition. Existing methods are limited to correlation networks that are independently estimated from separate group-specific data and without due consideration of relationships that are conserved across multiple groups. METHOD We propose a pathway-based differential network analysis in genomics (DINGO) model for estimating group-specific networks and making inference on the differential networks. DINGO jointly estimates the group-specific conditional dependencies by decomposing them into global and group-specific components. The delineation of these components allows for a more refined picture of the major driver and passenger events in the elucidation of cancer progression and development. RESULTS Simulation studies demonstrate that DINGO provides more accurate group-specific conditional dependencies than achieved by using separate estimation approaches. We apply DINGO to key signaling pathways in glioblastoma to build differential networks for long-term survivors and short-term survivors in The Cancer Genome Atlas. The hub genes found by mRNA expression, DNA copy number, methylation and microRNA expression reveal several important roles in glioblastoma progression. AVAILABILITY AND IMPLEMENTATION R Package at: odin.mdacc.tmc.edu/∼vbaladan. CONTACT veera@mdanderson.org SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
49
|
Yu X, Zeng T, Wang X, Li G, Chen L. Unravelling personalized dysfunctional gene network of complex diseases based on differential network model. J Transl Med 2015; 13:189. [PMID: 26070628 PMCID: PMC4467679 DOI: 10.1186/s12967-015-0546-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 05/25/2015] [Indexed: 11/10/2022] Open
Abstract
In the conventional analysis of complex diseases, the control and case samples are assumed to be of great purity. However, due to the heterogeneity of disease samples, many disease genes are even not always consistently up-/down-regulated, leading to be under-estimated. This problem will seriously influence effective personalized diagnosis or treatment. The expression variance and expression covariance can address such a problem in a network manner. But, these analyses always require multiple samples rather than one sample, which is generally not available in clinical practice for each individual. To extract the common and specific network characteristics for individual patients in this paper, a novel differential network model, e.g. personalized dysfunctional gene network, is proposed to integrate those genes with different features, such as genes with the differential gene expression (DEG), genes with the differential expression variance (DEVG) and gene-pairs with the differential expression covariance (DECG) simultaneously, to construct personalized dysfunctional networks. This model uses a new statistic-like measurement on differential information, i.e., a differential score (DEVC), to reconstruct the differential expression network between groups of normal and diseased samples; and further quantitatively evaluate different feature genes in the patient-specific network for each individual. This DEVC-based differential expression network (DEVC-net) has been applied to the study of complex diseases for prostate cancer and diabetes. (1) Characterizing the global expression change between normal and diseased samples, the differential gene networks of those diseases were found to have a new bi-coloured topological structure, where their non hub-centred sub-networks are mainly composed of genes/proteins controlling various biological processes. (2) The differential expression variance/covariance rather than differential expression is new informative sources, and can be used to identify genes or gene-pairs with discriminative power, which are ignored by traditional methods. (3) More importantly, DEVC-net is effective to measure the expression state or activity of different feature genes and their network or modules in one sample for an individual. All of these results support that DEVC-net indeed has a clear advantage to effectively extract discriminatively interpretable features of gene/protein network of one sample (i.e. personalized dysfunctional network) even when disease samples are heterogeneous, and thus can provide new features like gene-pairs, in addition to the conventional individual genes, to the analysis of the personalized diagnosis and prognosis, and a better understanding on the underlying biological mechanisms.
Collapse
Affiliation(s)
- Xiangtian Yu
- School of Mathematics, Shandong University, Jinan, 250100, China. .,Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Tao Zeng
- Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiangdong Wang
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Shanghai, China.
| | - Guojun Li
- School of Mathematics, Shandong University, Jinan, 250100, China.
| | - Luonan Chen
- Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China. .,Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
50
|
Chen H, Zhu Z, Zhu Y, Wang J, Mei Y, Cheng Y. Pathway mapping and development of disease-specific biomarkers: protein-based network biomarkers. J Cell Mol Med 2015; 19:297-314. [PMID: 25560835 PMCID: PMC4407592 DOI: 10.1111/jcmm.12447] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/22/2014] [Indexed: 01/06/2023] Open
Abstract
It is known that a disease is rarely a consequence of an abnormality of a single gene, but reflects the interactions of various processes in a complex network. Annotated molecular networks offer new opportunities to understand diseases within a systems biology framework and provide an excellent substrate for network-based identification of biomarkers. The network biomarkers and dynamic network biomarkers (DNBs) represent new types of biomarkers with protein-protein or gene-gene interactions that can be monitored and evaluated at different stages and time-points during development of disease. Clinical bioinformatics as a new way to combine clinical measurements and signs with human tissue-generated bioinformatics is crucial to translate biomarkers into clinical application, validate the disease specificity, and understand the role of biomarkers in clinical settings. In this article, the recent advances and developments on network biomarkers and DNBs are comprehensively reviewed. How network biomarkers help a better understanding of molecular mechanism of diseases, the advantages and constraints of network biomarkers for clinical application, clinical bioinformatics as a bridge to the development of diseases-specific, stage-specific, severity-specific and therapy predictive biomarkers, and the potentials of network biomarkers are also discussed.
Collapse
Affiliation(s)
- Hao Chen
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji University, Shanghai, China
| | | | | | | | | | | |
Collapse
|