151
|
Gonzalez-Valbuena EE, Treviño V. Metrics to estimate differential co-expression networks. BioData Min 2017; 10:32. [PMID: 29151892 PMCID: PMC5681815 DOI: 10.1186/s13040-017-0152-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Detecting the differences in gene expression data is important for understanding the underlying molecular mechanisms. Although the differentially expressed genes are a large component, differences in correlation are becoming an interesting approach to achieving deeper insights. However, diverse metrics have been used to detect differential correlation, making selection and use of a single metric difficult. In addition, available implementations are metric-specific, complicating their use in different contexts. Moreover, because the analyses in the literature have been performed on real data, there are uncertainties regarding the performance of metrics and procedures. RESULTS In this work, we compare four novel and two previously proposed metrics to detect differential correlations. We generated well-controlled datasets into which differences in correlations were carefully introduced by controlled multivariate normal correlation networks and addition of noise. The comparisons were performed on three datasets derived from real tumor data. Our results show that metrics differ in their detection performance and computational time. No single metric was the best in all datasets, but trends show that three metrics are highly correlated and are very good candidates for real data analysis. In contrast, other metrics proposed in the literature seem to show low performance and different detections. Overall, our results suggest that metrics that do not filter correlations perform better. We also show an additional analysis of TCGA breast cancer subtypes. CONCLUSIONS We show a methodology to generate controlled datasets for the objective evaluation of differential correlation pipelines, and compare the performance of several metrics. We implemented in R a package called DifCoNet that can provide easy-to-use functions for differential correlation analyses.
Collapse
Affiliation(s)
| | - Víctor Treviño
- Cátedra de Bioinformática, Escuela de Medicina, Tecnológico de Monterrey, 64710 Monterrey, Nuevo León Mexico
| |
Collapse
|
152
|
Uddin R, Singh SM. Gene Network Construction from Microarray Data Identifies a Key Network Module and Several Candidate Hub Genes in Age-Associated Spatial Learning Impairment. Front Syst Neurosci 2017; 11:75. [PMID: 29066959 PMCID: PMC5641338 DOI: 10.3389/fnsys.2017.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/22/2017] [Indexed: 01/06/2023] Open
Abstract
As humans age many suffer from a decrease in normal brain functions including spatial learning impairments. This study aimed to better understand the molecular mechanisms in age-associated spatial learning impairment (ASLI). We used a mathematical modeling approach implemented in Weighted Gene Co-expression Network Analysis (WGCNA) to create and compare gene network models of young (learning unimpaired) and aged (predominantly learning impaired) brains from a set of exploratory datasets in rats in the context of ASLI. The major goal was to overcome some of the limitations previously observed in the traditional meta- and pathway analysis using these data, and identify novel ASLI related genes and their networks based on co-expression relationship of genes. This analysis identified a set of network modules in the young, each of which is highly enriched with genes functioning in broad but distinct GO functional categories or biological pathways. Interestingly, the analysis pointed to a single module that was highly enriched with genes functioning in “learning and memory” related functions and pathways. Subsequent differential network analysis of this “learning and memory” module in the aged (predominantly learning impaired) rats compared to the young learning unimpaired rats allowed us to identify a set of novel ASLI candidate hub genes. Some of these genes show significant repeatability in networks generated from independent young and aged validation datasets. These hub genes are highly co-expressed with other genes in the network, which not only show differential expression but also differential co-expression and differential connectivity across age and learning impairment. The known function of these hub genes indicate that they play key roles in critical pathways, including kinase and phosphatase signaling, in functions related to various ion channels, and in maintaining neuronal integrity relating to synaptic plasticity and memory formation. Taken together, they provide a new insight and generate new hypotheses into the molecular mechanisms responsible for age associated learning impairment, including spatial learning.
Collapse
Affiliation(s)
- Raihan Uddin
- Department of Biology, University of Western Ontario, London, ON, Canada
| | - Shiva M Singh
- Department of Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
153
|
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
|
154
|
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
|
155
|
Ghanat Bari M, Ung CY, Zhang C, Zhu S, Li H. Machine Learning-Assisted Network Inference Approach to Identify a New Class of Genes that Coordinate the Functionality of Cancer Networks. Sci Rep 2017; 7:6993. [PMID: 28765560 PMCID: PMC5539301 DOI: 10.1038/s41598-017-07481-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/27/2017] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence indicates the existence of a new class of cancer genes that act as "signal linkers" coordinating oncogenic signals between mutated and differentially expressed genes. While frequently mutated oncogenes and differentially expressed genes, which we term Class I cancer genes, are readily detected by most analytical tools, the new class of cancer-related genes, i.e., Class II, escape detection because they are neither mutated nor differentially expressed. Given this hypothesis, we developed a Machine Learning-Assisted Network Inference (MALANI) algorithm, which assesses all genes regardless of expression or mutational status in the context of cancer etiology. We used 8807 expression arrays, corresponding to 9 cancer types, to build more than 2 × 108 Support Vector Machine (SVM) models for reconstructing a cancer network. We found that ~3% of ~19,000 not differentially expressed genes are Class II cancer gene candidates. Some Class II genes that we found, such as SLC19A1 and ATAD3B, have been recently reported to associate with cancer outcomes. To our knowledge, this is the first study that utilizes both machine learning and network biology approaches to uncover Class II cancer genes in coordinating functionality in cancer networks and will illuminate our understanding of how genes are modulated in a tissue-specific network contribute to tumorigenesis and therapy development.
Collapse
Affiliation(s)
- Mehrab Ghanat Bari
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| |
Collapse
|
156
|
Zhang XF, Ou-Yang L, Yan H. Node-based differential network analysis in genomics. Comput Biol Chem 2017; 69:194-201. [DOI: 10.1016/j.compbiolchem.2017.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 12/26/2022]
|
157
|
Ou-Yang L, Yan H, Zhang XF. Identifying differential networks based on multi-platform gene expression data. MOLECULAR BIOSYSTEMS 2017; 13:183-192. [PMID: 27868129 DOI: 10.1039/c6mb00619a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exploring how the structure of a gene regulatory network differs between two different disease states is fundamental for understanding the biological mechanisms behind disease development and progression. Recently, with rapid advances in microarray technologies, gene expression profiles of the same patients can be collected from multiple microarray platforms. However, previous differential network analysis methods were usually developed based on a single type of platform, which could not utilize the common information shared across different platforms. In this study, we introduce a multi-view differential network analysis model to infer the differential network between two different patient groups based on gene expression profiles collected from multiple platforms. Unlike previous differential network analysis models that need to analyze each platform separately, our model can draw support from multiple data platforms to jointly estimate the differential networks and produce more accurate and reliable results. Our simulation studies demonstrate that our method consistently outperforms other available differential network analysis methods. We also applied our method to identify network rewiring associated with platinum resistance using TCGA ovarian cancer samples. The experimental results demonstrate that the hub genes in our identified differential networks on the PI3K/AKT/mTOR pathway play an important role in drug resistance.
Collapse
Affiliation(s)
- Le Ou-Yang
- College of Information Engineering, Shenzhen University, Shenzhen, China and Department of Electronic and Engineering, City University of Hong Kong, Hong Kong, China
| | - Hong Yan
- Department of Electronic and Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics & Hubei Key Laboratory of Mathematical Sciences, Central China Normal University, Wuhan, China.
| |
Collapse
|
158
|
Gov E, Arga KY. Differential co-expression analysis reveals a novel prognostic gene module in ovarian cancer. Sci Rep 2017; 7:4996. [PMID: 28694494 PMCID: PMC5504034 DOI: 10.1038/s41598-017-05298-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 05/26/2017] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is one of the most significant disease among gynecological disorders that women suffered from over the centuries. However, disease-specific and effective biomarkers were still not available, since studies have focused on individual genes associated with ovarian cancer, ignoring the interactions and associations among the gene products. Here, ovarian cancer differential co-expression networks were reconstructed via meta-analysis of gene expression data and co-expressed gene modules were identified in epithelial cells from ovarian tumor and healthy ovarian surface epithelial samples to propose ovarian cancer associated genes and their interactions. We propose a novel, highly interconnected, differentially co-expressed, and co-regulated gene module in ovarian cancer consisting of 84 prognostic genes. Furthermore, the specificity of the module to ovarian cancer was shown through analyses of datasets in nine other cancers. These observations underscore the importance of transcriptome based systems biomarkers research in deciphering the elusive pathophysiology of ovarian cancer, and here, we present reciprocal interplay between candidate ovarian cancer genes and their transcriptional regulatory dynamics. The corresponding gene module might provide new insights on ovarian cancer prognosis and treatment strategies that continue to place a significant burden on global health.
Collapse
Affiliation(s)
- Esra Gov
- Department of Bioengineering, Marmara University, 34722, Goztepe, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, 34722, Goztepe, Istanbul, Turkey.
| |
Collapse
|
159
|
Bhattacharya A, Cui Y. A GPU-accelerated algorithm for biclustering analysis and detection of condition-dependent coexpression network modules. Sci Rep 2017. [PMID: 28646174 PMCID: PMC5482832 DOI: 10.1038/s41598-017-04070-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
In the analysis of large-scale gene expression data, it is important to identify groups of genes with common expression patterns under certain conditions. Many biclustering algorithms have been developed to address this problem. However, comprehensive discovery of functionally coherent biclusters from large datasets remains a challenging problem. Here we propose a GPU-accelerated biclustering algorithm, based on searching for the largest Condition-dependent Correlation Subgroups (CCS) for each gene in the gene expression dataset. We compared CCS with thirteen widely used biclustering algorithms. CCS consistently outperformed all the thirteen biclustering algorithms on both synthetic and real gene expression datasets. As a correlation-based biclustering method, CCS can also be used to find condition-dependent coexpression network modules. We implemented the CCS algorithm using C and implemented the parallelized CCS algorithm using CUDA C for GPU computing. The source code of CCS is available from https://github.com/abhatta3/Condition-dependent-Correlation-Subgroups-CCS.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- Department of Microbiology, Immunology and Biochemistry, Memphis, TN, 38163, USA. .,Center for Integrative and Translational Genomics, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Computer Science and Engineering, University of California, San Diego, CA, 92093, USA.
| | - Yan Cui
- Department of Microbiology, Immunology and Biochemistry, Memphis, TN, 38163, USA. .,Center for Integrative and Translational Genomics, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
160
|
Huo M, Wang Z, Wu D, Zhang Y, Qiao Y. Using Coexpression Protein Interaction Network Analysis to Identify Mechanisms of Danshensu Affecting Patients with Coronary Heart Disease. Int J Mol Sci 2017. [PMID: 28629174 PMCID: PMC5486119 DOI: 10.3390/ijms18061298] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Salvia miltiorrhiza, known as Danshen, has attracted worldwide interest for its substantial effects on coronary heart disease (CHD). Danshensu (DSS) is one of the main active ingredients of Danshen on CHD. Although it has been proven to have a good clinical effect on CHD, the action mechanisms remain elusive. In the current study, a coexpression network-based approach was used to illustrate the beneficial properties of DSS in the context of CHD. By integrating the gene expression profile data and protein-protein interactions (PPIs) data, two coexpression protein interaction networks (CePIN) in a CHD state (CHD CePIN) and a non-CHD state (non-CHD CePIN) were generated. Then, shared nodes and unique nodes in CHD CePIN were attained by conducting a comparison between CHD CePIN and non-CHD CePIN. By calculating the topological parameters of each shared node and unique node in the networks, and comparing the differentially expressed genes, target proteins involved in disease regulation were attained. Then, Gene Ontology (GO) enrichment was utilized to identify biological processes associated to target proteins. Consequently, it turned out that the treatment of CHD with DSS may be partly attributed to the regulation of immunization and blood circulation. Also, it indicated that sodium/hydrogen exchanger 3 (SLC9A3), Prostaglandin G/H synthase 2 (PTGS2), Oxidized low-density lipoprotein receptor 1 (OLR1), and fibrinogen gamma chain (FGG) may be potential therapeutic targets for CHD. In summary, this study provided a novel coexpression protein interaction network approach to provide an explanation of the mechanisms of DSS on CHD and identify key proteins which maybe the potential therapeutic targets for CHD.
Collapse
Affiliation(s)
- Mengqi Huo
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine; School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Zhixin Wang
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine; School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Dongxue Wu
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine; School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Yanling Zhang
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine; School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Yanjiang Qiao
- Key Laboratory of Traditional Chinese Medicine Information Engineer of State Administration of Traditional Chinese Medicine; School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
161
|
Mechanisms of action of sacubitril/valsartan on cardiac remodeling: a systems biology approach. NPJ Syst Biol Appl 2017. [PMID: 28649439 PMCID: PMC5460292 DOI: 10.1038/s41540-017-0013-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sacubitril/Valsartan, proved superiority over other conventional heart failure management treatments, but its mechanisms of action remains obscure. In this study, we sought to explore the mechanistic details for Sacubitril/Valsartan in heart failure and post-myocardial infarction remodeling, using an in silico, systems biology approach. Myocardial transcriptome obtained in response to myocardial infarction in swine was analyzed to address post-infarction ventricular remodeling. Swine transcriptome hits were mapped to their human equivalents using Reciprocal Best (blast) Hits, Gene Name Correspondence, and InParanoid database. Heart failure remodeling was studied using public data available in gene expression omnibus (accession GSE57345, subseries GSE57338), processed using the GEO2R tool. Using the Therapeutic Performance Mapping System technology, dedicated mathematical models trained to fit a set of molecular criteria, defining both pathologies and including all the information available on Sacubitril/Valsartan, were generated. All relationships incorporated into the biological network were drawn from public resources (including KEGG, REACTOME, INTACT, BIOGRID, and MINT). An artificial neural network analysis revealed that Sacubitril/Valsartan acts synergistically against cardiomyocyte cell death and left ventricular extracellular matrix remodeling via eight principal synergistic nodes. When studying each pathway independently, Valsartan was found to improve cardiac remodeling by inhibiting members of the guanine nucleotide-binding protein family, while Sacubitril attenuated cardiomyocyte cell death, hypertrophy, and impaired myocyte contractility by inhibiting PTEN. The complex molecular mechanisms of action of Sacubitril/Valsartan upon post-myocardial infarction and heart failure cardiac remodeling were delineated using a systems biology approach. Further, this dataset provides pathophysiological rationale for the use of Sacubitril/Valsartan to prevent post-infarct remodeling. The new wonder drug in heart failure management, Sacubitril/Valsartan, rejuvenates the heart by preventing its dilation. Using data from myocardial infarction and heart failure samples, we generated a mathematical model to better understand how Sacubitril/Valsartan modulates pathological heart resize and the combined effect of the drug. Our analysis revealed that Sacubitril/Valsartan mainly acts by blocking both, cell death and the pathological makeover of the outer-membrane of the cardiac cells. These two major processes occur after a heart attack. Most importantly, we discovered a core of 8 proteins that emerge as key players in this process. A better understanding of the mechanism of novel cardiovascular drugs at the most basic level may help decipher future therapies and indications.
Collapse
|
162
|
Zhang XF, Ou-Yang L, Yan H. Incorporating prior information into differential network analysis using non-paranormal graphical models. Bioinformatics 2017; 33:2436-2445. [DOI: 10.1093/bioinformatics/btx208] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/05/2017] [Indexed: 02/02/2023] Open
Affiliation(s)
- Xiao-Fei Zhang
- Department of Statistics, School of Mathematics and Statistics & Hubei Key Laboratory of Mathematical Sciences, Central China Normal University, Wuhan, China
- Department of Electronic Engineering, City University of Hong Kong, Hong Kong, China
| | - Le Ou-Yang
- Department of Electronic Engineering, College of Information Engineering, Shenzhen University, Shenzhen, China
| | - Hong Yan
- Department of Electronic Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
163
|
Will T, Helms V. Rewiring of the inferred protein interactome during blood development studied with the tool PPICompare. BMC SYSTEMS BIOLOGY 2017; 11:44. [PMID: 28376810 PMCID: PMC5379774 DOI: 10.1186/s12918-017-0400-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/26/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Differential analysis of cellular conditions is a key approach towards understanding the consequences and driving causes behind biological processes such as developmental transitions or diseases. The progress of whole-genome expression profiling enabled to conveniently capture the state of a cell's transcriptome and to detect the characteristic features that distinguish cells in specific conditions. In contrast, mapping the physical protein interactome for many samples is experimentally infeasible at the moment. For the understanding of the whole system, however, it is equally important how the interactions of proteins are rewired between cellular states. To overcome this deficiency, we recently showed how condition-specific protein interaction networks that even consider alternative splicing can be inferred from transcript expression data. Here, we present the differential network analysis tool PPICompare that was specifically designed for isoform-sensitive protein interaction networks. RESULTS Besides detecting significant rewiring events between the interactomes of grouped samples, PPICompare infers which alterations to the transcriptome caused each rewiring event and what is the minimal set of alterations necessary to explain all between-group changes. When applied to the development of blood cells, we verified that a reasonable amount of rewiring events were reported by the tool and found that differential gene expression was the major determinant of cellular adjustments to the interactome. Alternative splicing events were consistently necessary in each developmental step to explain all significant alterations and were especially important for rewiring in the context of transcriptional control. CONCLUSIONS Applying PPICompare enabled us to investigate the dynamics of the human protein interactome during developmental transitions. A platform-independent implementation of the tool PPICompare is available at https://sourceforge.net/projects/ppicompare/ .
Collapse
Affiliation(s)
- Thorsten Will
- Center for Bioinformatics, Saarland University, Campus E2.1, Saarbrücken, 66123 Germany
- Graduate School of Computer Science, Saarland University, Campus E1.3, Saarbrücken, 66123 Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Campus E2.1, Saarbrücken, 66123 Germany
| |
Collapse
|
164
|
Martins AJ, Narayanan M, Prüstel T, Fixsen B, Park K, Gottschalk RA, Lu Y, Andrews-Pfannkoch C, Lau WW, Wendelsdorf KV, Tsang JS. Environment Tunes Propagation of Cell-to-Cell Variation in the Human Macrophage Gene Network. Cell Syst 2017; 4:379-392.e12. [PMID: 28365150 DOI: 10.1016/j.cels.2017.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/15/2016] [Accepted: 03/01/2017] [Indexed: 01/22/2023]
Abstract
Cell-to-cell variation in gene expression and the propagation of such variation (PoV or "noise propagation") from one gene to another in the gene network, as reflected by gene-gene correlation across single cells, are commonly observed in single-cell transcriptomic studies and can shape the phenotypic diversity of cell populations. While gene network "rewiring" is known to accompany cellular adaptation to different environments, how PoV changes between environments and its underlying regulatory mechanisms are less understood. Here, we systematically explored context-dependent PoV among genes in human macrophages, utilizing different cytokines as natural perturbations of multiple molecular parameters that may influence PoV. Our single-cell, epigenomic, computational, and stochastic simulation analyses reveal that environmental adaptation can tune PoV to potentially shape cellular heterogeneity by changing parameters such as the degree of phosphorylation and transcription factor-chromatin interactions. This quantitative tuning of PoV may be a widespread, yet underexplored, property of cellular adaptation to distinct environments.
Collapse
Affiliation(s)
- Andrew J Martins
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manikandan Narayanan
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thorsten Prüstel
- Computational Biology Section, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bethany Fixsen
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kyemyung Park
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA; Biophysics Program, University of Maryland-NIH Graduate Partnership Program, University of Maryland, College Park, MD 20742, USA
| | - Rachel A Gottschalk
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong Lu
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cynthia Andrews-Pfannkoch
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - William W Lau
- Office of Intramural Research, Center for Information Technology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine V Wendelsdorf
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - John S Tsang
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institutes of Health, Bethesda, MD 20892, USA; Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
165
|
Muntané G, Santpere G, Verendeev A, Seeley WW, Jacobs B, Hopkins WD, Navarro A, Sherwood CC. Interhemispheric gene expression differences in the cerebral cortex of humans and macaque monkeys. Brain Struct Funct 2017; 222:3241-3254. [PMID: 28317062 DOI: 10.1007/s00429-017-1401-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/05/2017] [Indexed: 11/25/2022]
Abstract
Handedness and language are two well-studied examples of asymmetrical brain function in humans. Approximately 90% of humans exhibit a right-hand preference, and the vast majority shows left-hemisphere dominance for language function. Although genetic models of human handedness and language have been proposed, the actual gene expression differences between cerebral hemispheres in humans remain to be fully defined. In the present study, gene expression profiles were examined in both hemispheres of three cortical regions involved in handedness and language in humans and their homologues in rhesus macaques: ventrolateral prefrontal cortex, posterior superior temporal cortex (STC), and primary motor cortex. Although the overall pattern of gene expression was very similar between hemispheres in both humans and macaques, weighted gene correlation network analysis revealed gene co-expression modules associated with hemisphere, which are different among the three cortical regions examined. Notably, a receptor-enriched gene module in STC was particularly associated with hemisphere and showed different expression levels between hemispheres only in humans.
Collapse
Affiliation(s)
- Gerard Muntané
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, 20052, USA.
- Institut Biologia Evolutiva, Universitat Pompeu Fabra-CSIC, 08003, Barcelona, Spain.
| | - Gabriel Santpere
- Institut Biologia Evolutiva, Universitat Pompeu Fabra-CSIC, 08003, Barcelona, Spain
| | - Andrey Verendeev
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, 20052, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA, 94158, USA
| | - Bob Jacobs
- Laboratory of Quantitative Neuromorphology, Neuroscience Program, Colorado College, Colorado Springs, CO, 80903, USA
| | - William D Hopkins
- Neuroscience Institute and the Language Research Center, Georgia State University, Atlanta, GA, 30302, USA
| | - Arcadi Navarro
- Institut Biologia Evolutiva, Universitat Pompeu Fabra-CSIC, 08003, Barcelona, Spain
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, 20052, USA
| |
Collapse
|
166
|
Li Q, Li J, Dai W, Li YX, Li YY. Differential regulation analysis reveals dysfunctional regulatory mechanism involving transcription factors and microRNAs in gastric carcinogenesis. Artif Intell Med 2017; 77:12-22. [PMID: 28545608 DOI: 10.1016/j.artmed.2017.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/23/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) is one of the most incident malignancies in the world. Although lots of featured genes and microRNAs (miRNAs) have been identified to be associated with gastric carcinogenesis, underlying regulatory mechanisms still remain unclear. In order to explore the dysfunctional mechanisms of GC, we developed a novel approach to identify carcinogenesis relevant regulatory relationships, which is characterized by quantifying the difference of regulatory relationships between stages. Firstly, we applied the strategy of differential coexpression analysis (DCEA) to transcriptomic datasets including paired mRNA and miRNA of gastric samples to identify a set of genes/miRNAs related to gastric cancer progression. Based on these genes/miRNAs, we constructed conditional combinatorial gene regulatory networks (cGRNs) involving both transcription factors (TFs) and miRNAs. Enrichment of known cancer genes/miRNAs and predicted prognostic genes/miRNAs was observed in each cGRN. Then we designed a quantitative method to measure differential regulation level of every regulatory relationship between normal and cancer, and the known cancer genes/miRNAs proved to be ranked significantly higher. Meanwhile, we defined differentially regulated link (DRL) by combining differential regulation, differential expression and the regulation contribution of the regulator to the target. By integrating survival analysis and DRL identification, three master regulators TCF7L1, TCF4, and MEIS1 were identified and testable hypotheses of dysfunctional mechanisms underlying gastric carcinogenesis related to them were generated. The fine-tuning effects of miRNAs were also observed. We propose that this differential regulation network analysis framework is feasible to gain insights into dysregulated mechanisms underlying tumorigenesis and other phenotypic changes.
Collapse
Affiliation(s)
- Quanxue Li
- School of biotechnology, East China University of Science and Technology, Shanghai, China; Shanghai Center for Bioinformation Technology, Shanghai, China
| | - Junyi Li
- Shanghai Center for Bioinformation Technology, Shanghai, China; Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wentao Dai
- Shanghai Center for Bioinformation Technology, Shanghai, China; Shanghai Industrial Technology Institute, Shanghai, China; Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China
| | - Yi-Xue Li
- School of biotechnology, East China University of Science and Technology, Shanghai, China; Shanghai Center for Bioinformation Technology, Shanghai, China; Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Shanghai Industrial Technology Institute, Shanghai, China; Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China.
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, Shanghai, China; Shanghai Industrial Technology Institute, Shanghai, China; Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China.
| |
Collapse
|
167
|
Abstract
Background The drug discovery and development pipeline is a long and arduous process that inevitably hampers rapid drug development. Therefore, strategies to improve the efficiency of drug development are urgently needed to enable effective drugs to enter the clinic. Precision medicine has demonstrated that genetic features of cancer cells can be used for predicting drug response, and emerging evidence suggest that gene-drug connections could be predicted more accurately by exploring the cumulative effects of many genes simultaneously. Results We developed DeSigN, a web-based tool for predicting drug efficacy against cancer cell lines using gene expression patterns. The algorithm correlates phenotype-specific gene signatures derived from differentially expressed genes with pre-defined gene expression profiles associated with drug response data (IC50) from 140 drugs. DeSigN successfully predicted the right drug sensitivity outcome in four published GEO studies. Additionally, it predicted bosutinib, a Src/Abl kinase inhibitor, as a sensitive inhibitor for oral squamous cell carcinoma (OSCC) cell lines. In vitro validation of bosutinib in OSCC cell lines demonstrated that indeed, these cell lines were sensitive to bosutinib with IC50 of 0.8–1.2 μM. As further confirmation, we demonstrated experimentally that bosutinib has anti-proliferative activity in OSCC cell lines, demonstrating that DeSigN was able to robustly predict drug that could be beneficial for tumour control. Conclusions DeSigN is a robust method that is useful for the identification of candidate drugs using an input gene signature obtained from gene expression analysis. This user-friendly platform could be used to identify drugs with unanticipated efficacy against cancer cell lines of interest, and therefore could be used for the repurposing of drugs, thus improving the efficiency of drug development. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3260-7) contains supplementary material, which is available to authorized users.
Collapse
|
168
|
Li Z, Yao Q, Zhao S, Wang Z, Li Y. Protein coding gene CRNKL1 as a potential prognostic biomarker in esophageal adenocarcinoma. Artif Intell Med 2017; 76:1-6. [PMID: 28363284 DOI: 10.1016/j.artmed.2017.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/12/2017] [Accepted: 01/19/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is one of the most aggressive gastroesophageal cancers. PTGS2, EGFR, ERBB2 and TP53 are the traditional EAC prognostic biomarkers, but they are still limited in their ability to effectively predict the overall survival. OBJECTIVES To identify an improved biomarker for predicting the prognosis of EAC by using the expression profile. MATERIALS AND METHODS Differential co-expression analysis and differential expression analysis were performed to identify the related genes of EAC. The 5-fold cross-validation was used to select a prognostic biomarker from the 532 EAC related genes. RESULTS CRNKL1 was identified as a prognostic biomarker to predict the survival of EAC patients. It could significantly stratify EAC patients into high-risk and low-risk groups and was much better than the traditional biomarkers. Furthermore, ROC curve also verified that CRNKL1 with the highest area under the curve (AUC), reaching a sensitivity of 83.33% and a specificity of 78.57%. CONCLUSIONS Our research proposed that CRNKL1 might be a novel prognostic biomarker with better predictive ability by comparing with the traditional biomarkers, which provided a preferable opportunity in the clinical applications of EAC.
Collapse
Affiliation(s)
- Zhen Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, China
| | - Qianlan Yao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, China
| | - Songjian Zhao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, China
| | - Zhen Wang
- Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yixue Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, China; Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
169
|
Siska C, Kechris K. Differential correlation for sequencing data. BMC Res Notes 2017; 10:54. [PMID: 28103954 PMCID: PMC5244536 DOI: 10.1186/s13104-016-2331-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/10/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Several methods have been developed to identify differential correlation (DC) between pairs of molecular features from -omics studies. Most DC methods have only been tested with microarrays and other platforms producing continuous and Gaussian-like data. Sequencing data is in the form of counts, often modeled with a negative binomial distribution making it difficult to apply standard correlation metrics. We have developed an R package for identifying DC called Discordant which uses mixture models for correlations between features and the Expectation Maximization (EM) algorithm for fitting parameters of the mixture model. Several correlation metrics for sequencing data are provided and tested using simulations. Other extensions in the Discordant package include additional modeling for different types of differential correlation, and faster implementation, using a subsampling routine to reduce run-time and address the assumption of independence between molecular feature pairs. RESULTS With simulations and breast cancer miRNA-Seq and RNA-Seq data, we find that Spearman's correlation has the best performance among the tested correlation methods for identifying differential correlation. Application of Spearman's correlation in the Discordant method demonstrated the most power in ROC curves and sensitivity/specificity plots, and improved ability to identify experimentally validated breast cancer miRNA. We also considered including additional types of differential correlation, which showed a slight reduction in power due to the additional parameters that need to be estimated, but more versatility in applications. Finally, subsampling within the EM algorithm considerably decreased run-time with negligible effect on performance. CONCLUSIONS A new method and R package called Discordant is presented for identifying differential correlation with sequencing data. Based on comparisons with different correlation metrics, this study suggests Spearman's correlation is appropriate for sequencing data, but other correlation metrics are available to the user depending on the application and data type. The Discordant method can also be extended to investigate additional DC types and subsampling with the EM algorithm is now available for reduced run-time. These extensions to the R package make Discordant more robust and versatile for multiple -omics studies.
Collapse
Affiliation(s)
- Charlotte Siska
- Computational Bioscience Program, Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
170
|
Gambardella G, Carissimo A, Chen A, Cutillo L, Nowakowski TJ, di Bernardo D, Blelloch R. The impact of microRNAs on transcriptional heterogeneity and gene co-expression across single embryonic stem cells. Nat Commun 2017; 8:14126. [PMID: 28102192 PMCID: PMC5253645 DOI: 10.1038/ncomms14126] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs act posttranscriptionally to suppress multiple target genes within a cell population. To what extent this multi-target suppression occurs in individual cells and how it impacts transcriptional heterogeneity and gene co-expression remains unknown. Here we used single-cell sequencing combined with introduction of individual microRNAs. miR-294 and let-7c were introduced into otherwise microRNA-deficient Dgcr8 knockout mouse embryonic stem cells. Both microRNAs induce suppression and correlated expression of their respective gene targets. The two microRNAs had opposing effects on transcriptional heterogeneity within the cell population, with let-7c increasing and miR-294 decreasing the heterogeneity between cells. Furthermore, let-7c promotes, whereas miR-294 suppresses, the phasing of cell cycle genes. These results show at the individual cell level how a microRNA simultaneously has impacts on its many targets and how that in turn can influence a population of cells. The findings have important implications in the understanding of how microRNAs influence the co-expression of genes and pathways, and thus ultimately cell fate. MicroRNAs can posttranscriptionally repress multiple targets in a cell population. Here the authors use single-cell sequencing to investigate the effects of an individual miRNA on transcriptional heterogeneity and gene co-expression
Collapse
Affiliation(s)
| | | | - Amy Chen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA.,Department of Urology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Luisa Cutillo
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy
| | - Tomasz J Nowakowski
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy.,Department of Chemical, Materials and Industrial Engineering, University of Naples 'Federico II', 80125 Naples, Italy
| | - Robert Blelloch
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA.,Department of Urology, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
171
|
Identifying a gene expression signature of cluster headache in blood. Sci Rep 2017; 7:40218. [PMID: 28074859 PMCID: PMC5225606 DOI: 10.1038/srep40218] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 12/05/2016] [Indexed: 12/12/2022] Open
Abstract
Cluster headache is a relatively rare headache disorder, typically characterized by multiple daily, short-lasting attacks of excruciating, unilateral (peri-)orbital or temporal pain associated with autonomic symptoms and restlessness. To better understand the pathophysiology of cluster headache, we used RNA sequencing to identify differentially expressed genes and pathways in whole blood of patients with episodic (n = 19) or chronic (n = 20) cluster headache in comparison with headache-free controls (n = 20). Gene expression data were analysed by gene and by module of co-expressed genes with particular attention to previously implicated disease pathways including hypocretin dysregulation. Only moderate gene expression differences were identified and no associations were found with previously reported pathogenic mechanisms. At the level of functional gene sets, associations were observed for genes involved in several brain-related mechanisms such as GABA receptor function and voltage-gated channels. In addition, genes and modules of co-expressed genes showed a role for intracellular signalling cascades, mitochondria and inflammation. Although larger study samples may be required to identify the full range of involved pathways, these results indicate a role for mitochondria, intracellular signalling and inflammation in cluster headache.
Collapse
|
172
|
Dong LY, Zhou WZ, Ni JW, Xiang W, Hu WH, Yu C, Li HY. Identifying the optimal gene and gene set in hepatocellular carcinoma based on differential expression and differential co-expression algorithm. Oncol Rep 2016; 37:1066-1074. [DOI: 10.3892/or.2016.5333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/10/2016] [Indexed: 11/06/2022] Open
|
173
|
He F, Maslov S. Pan- and core- network analysis of co-expression genes in a model plant. Sci Rep 2016; 6:38956. [PMID: 27982071 PMCID: PMC5159811 DOI: 10.1038/srep38956] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/14/2016] [Indexed: 01/18/2023] Open
Abstract
Genome-wide gene expression experiments have been performed using the model plant Arabidopsis during the last decade. Some studies involved construction of coexpression networks, a popular technique used to identify groups of co-regulated genes, to infer unknown gene functions. One approach is to construct a single coexpression network by combining multiple expression datasets generated in different labs. We advocate a complementary approach in which we construct a large collection of 134 coexpression networks based on expression datasets reported in individual publications. To this end we reanalyzed public expression data. To describe this collection of networks we introduced concepts of 'pan-network' and 'core-network' representing union and intersection between a sizeable fractions of individual networks, respectively. We showed that these two types of networks are different both in terms of their topology and biological function of interacting genes. For example, the modules of the pan-network are enriched in regulatory and signaling functions, while the modules of the core-network tend to include components of large macromolecular complexes such as ribosomes and photosynthetic machinery. Our analysis is aimed to help the plant research community to better explore the information contained within the existing vast collection of gene expression data in Arabidopsis.
Collapse
Affiliation(s)
- Fei He
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Sergei Maslov
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
- Department of Bioengineering, Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
174
|
Kayano M, Higaki S, Satoh JI, Matsumoto K, Matsubara E, Takikawa O, Niida S. Plasma microRNA biomarker detection for mild cognitive impairment using differential correlation analysis. Biomark Res 2016; 4:22. [PMID: 27999671 PMCID: PMC5151129 DOI: 10.1186/s40364-016-0076-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/22/2016] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Mild cognitive impairment (MCI) is an intermediate state between normal aging and dementia including Alzheimer's disease. Early detection of dementia, and MCI, is a crucial issue in terms of secondary prevention. Blood biomarker detection is a possible way for early detection of MCI. Although disease biomarkers are detected by, in general, using single molecular analysis such as t-test, another possible approach is based on interaction between molecules. RESULTS Differential correlation analysis, which detects difference on correlation of two variables in case/control study, was carried out to plasma microRNA (miRNA) expression profiles of 30 age- and race-matched controls and 23 Japanese MCI patients. The 20 pairs of miRNAs, which consist of 20 miRNAs, were selected as MCI markers. Two pairs of miRNAs (hsa-miR-191 and hsa-miR-101, and hsa-miR-103 and hsa-miR-222) out of 20 attained the highest area under the curve (AUC) value of 0.962 for MCI detection. Other two miRNA pairs that include hsa-miR-191 and hsa-miR-125b also attained high AUC value of ≥ 0.95. Pathway analysis was performed to the MCI markers for further understanding of biological implications. As a result, collapsed correlation on hsa-miR-191 and emerged correlation on hsa-miR-125b might have key role in MCI and dementia progression. CONCLUSION Differential correlation analysis, a bioinformatics tool to elucidate complicated and interdependent biological systems behind diseases, detects effective MCI markers that cannot be found by single molecule analysis such as t-test.
Collapse
Affiliation(s)
- Mitsunori Kayano
- Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Sayuri Higaki
- Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Jun-ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Etsuro Matsubara
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Osamu Takikawa
- Innovation Center for Clinical Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Shumpei Niida
- Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| |
Collapse
|
175
|
Differentially Coexpressed Disease Gene Identification Based on Gene Coexpression Network. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3962761. [PMID: 28042568 PMCID: PMC5155124 DOI: 10.1155/2016/3962761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/26/2016] [Indexed: 11/17/2022]
Abstract
Screening disease-related genes by analyzing gene expression data has become a popular theme. Traditional disease-related gene selection methods always focus on identifying differentially expressed gene between case samples and a control group. These traditional methods may not fully consider the changes of interactions between genes at different cell states and the dynamic processes of gene expression levels during the disease progression. However, in order to understand the mechanism of disease, it is important to explore the dynamic changes of interactions between genes in biological networks at different cell states. In this study, we designed a novel framework to identify disease-related genes and developed a differentially coexpressed disease-related gene identification method based on gene coexpression network (DCGN) to screen differentially coexpressed genes. We firstly constructed phase-specific gene coexpression network using time-series gene expression data and defined the conception of differential coexpression of genes in coexpression network. Then, we designed two metrics to measure the value of gene differential coexpression according to the change of local topological structures between different phase-specific networks. Finally, we conducted meta-analysis of gene differential coexpression based on the rank-product method. Experimental results demonstrated the feasibility and effectiveness of DCGN and the superior performance of DCGN over other popular disease-related gene selection methods through real-world gene expression data sets.
Collapse
|
176
|
Thomas LD, Vyshenska D, Shulzhenko N, Yambartsev A, Morgun A. Differentially correlated genes in co-expression networks control phenotype transitions. F1000Res 2016; 5:2740. [PMID: 28163897 PMCID: PMC5247791 DOI: 10.12688/f1000research.9708.1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2016] [Indexed: 01/06/2023] Open
Abstract
Background: Co-expression networks are a tool widely used for analysis of “Big Data” in biology that can range from transcriptomes to proteomes, metabolomes and more recently even microbiomes. Several methods were proposed to answer biological questions interrogating these networks. Differential co-expression analysis is a recent approach that measures how gene interactions change when a biological system transitions from one state to another. Although the importance of differentially co-expressed genes to identify dysregulated pathways has been noted, their role in gene regulation is not well studied. Herein we investigated differentially co-expressed genes in a relatively simple mono-causal process (B lymphocyte deficiency) and in a complex multi-causal system (cervical cancer). Methods: Co-expression networks of B cell deficiency (Control and BcKO) were reconstructed using Pearson correlation coefficient for two
mus musculus datasets: B10.A strain (12 normal, 12 BcKO) and BALB/c strain (10 normal, 10 BcKO). Co-expression networks of cervical cancer (normal and cancer) were reconstructed using local partial correlation method for five datasets (total of 64 normal, 148 cancer). Differentially correlated pairs were identified along with the location of their genes in BcKO and in cancer networks. Minimum Shortest Path and Bi-partite Betweenness Centrality where statistically evaluated for differentially co-expressed genes in corresponding networks. Results: We show that in B cell deficiency the differentially co-expressed genes are highly enriched with immunoglobulin genes (causal genes). In cancer we found that differentially co-expressed genes act as “bottlenecks” rather than causal drivers with most flows that come from the key driver genes to the peripheral genes passing through differentially co-expressed genes. Using
in vitro knockdown experiments for two out of 14 differentially co-expressed genes found in cervical cancer (FGFR2 and CACYBP), we showed that they play regulatory roles in cancer cell growth. Conclusion: Identifying differentially co-expressed genes in co-expression networks is an important tool in detecting regulatory genes involved in alterations of phenotype.
Collapse
Affiliation(s)
- Lina D Thomas
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Anatoly Yambartsev
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, Brazil
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, USA
| |
Collapse
|
177
|
Troy NM, Bosco A. Respiratory viral infections and host responses; insights from genomics. Respir Res 2016; 17:156. [PMID: 27871304 PMCID: PMC5117516 DOI: 10.1186/s12931-016-0474-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/10/2016] [Indexed: 01/23/2023] Open
Abstract
Respiratory viral infections are a leading cause of disease and mortality. The severity of these illnesses can vary markedly from mild or asymptomatic upper airway infections to severe wheezing, bronchiolitis or pneumonia. In this article, we review the viral sensing pathways and organizing principles that govern the innate immune response to infection. Then, we reconstruct the molecular networks that differentiate symptomatic from asymptomatic respiratory viral infections, and identify the underlying molecular drivers of these networks. Finally, we discuss unique aspects of the biology and pathogenesis of infections with respiratory syncytial virus, rhinovirus and influenza, drawing on insights from genomics.
Collapse
Affiliation(s)
- Niamh M Troy
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia.
| |
Collapse
|
178
|
Wang D, Wang J, Jiang Y, Liang Y, Xu D. BFDCA: A Comprehensive Tool of Using Bayes Factor for Differential Co-Expression Analysis. J Mol Biol 2016; 429:446-453. [PMID: 27984044 DOI: 10.1016/j.jmb.2016.10.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/22/2016] [Accepted: 10/23/2016] [Indexed: 10/20/2022]
Abstract
Comparing the gene-expression profiles between biological conditions is useful for understanding gene regulation underlying complex phenotypes. Along this line, analysis of differential co-expression (DC) has gained attention in the recent years, where genes under one condition have different co-expression patterns compared with another. We developed an R package Bayes Factor approach for Differential Co-expression Analysis (BFDCA) for DC analysis. BFDCA is unique in integrating various aspects of DC patterns (including Shift, Cross, and Re-wiring) into one uniform Bayes factor. We tested BFDCA using simulation data and experimental data. Simulation results indicate that BFDCA outperforms existing methods in accuracy and robustness of detecting DC pairs and DC modules. Results of using experimental data suggest that BFDCA can cluster disease-related genes into functional DC subunits and estimate the regulatory impact of disease-related genes well. BFDCA also achieves high accuracy in predicting case-control phenotypes by using significant DC gene pairs as markers. BFDCA is publicly available at http://dx.doi.org/10.17632/jdz4vtvnm3.1.
Collapse
Affiliation(s)
- Duolin Wang
- College of Computer Science and Technology, Jilin University, Changchun, China 130012; Department of Computer Science and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Juexin Wang
- Department of Computer Science and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Yuexu Jiang
- College of Computer Science and Technology, Jilin University, Changchun, China 130012; Department of Computer Science and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Yanchun Liang
- College of Computer Science and Technology, Jilin University, Changchun, China 130012; Department of Computer Science and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Dong Xu
- College of Computer Science and Technology, Jilin University, Changchun, China 130012; Department of Computer Science and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
179
|
He FQ, Ollert M. Network-Guided Key Gene Discovery for a Given Cellular Process. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016. [PMID: 27783134 DOI: 10.1007/10_2016_39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Identification of key genes for a given physiological or pathological process is an essential but still very challenging task for the entire biomedical research community. Statistics-based approaches, such as genome-wide association study (GWAS)- or quantitative trait locus (QTL)-related analysis have already made enormous contributions to identifying key genes associated with a given disease or phenotype, the success of which is however very much dependent on a huge number of samples. Recent advances in network biology, especially network inference directly from genome-scale data and the following-up network analysis, opens up new avenues to predict key genes driving a given biological process or cellular function. Here we review and compare the current approaches in predicting key genes, which have no chances to stand out by classic differential expression analysis, from gene-regulatory, protein-protein interaction, or gene expression correlation networks. We elaborate these network-based approaches mainly in the context of immunology and infection, and urge more usage of correlation network-based predictions. Such network-based key gene discovery approaches driven by information-enriched 'omics' data should be very useful for systematic key gene discoveries for any given biochemical process or cellular function, and also valuable for novel drug target discovery and novel diagnostic, prognostic and therapeutic-efficiency marker prediction for a specific disease or disorder.
Collapse
Affiliation(s)
- Feng Q He
- Department of Infection and Immunity, Group of Immune Systems Biology, Luxembourg Institute of Health, 29, rue Henri Koch, 4354, Esch-sur-Alzette, Luxembourg.
| | - Markus Ollert
- Department of Infection and Immunity, Group of Allergy and Clinical Immunology, Luxembourg Institute of Health, 29, rue Henri Koch, 4354, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000, Odense C, Denmark
| |
Collapse
|
180
|
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
|
181
|
Differential Coexpression Analysis Reveals Extensive Rewiring of Arabidopsis Gene Coexpression in Response to Pseudomonas syringae Infection. Sci Rep 2016; 6:35064. [PMID: 27721457 PMCID: PMC5056366 DOI: 10.1038/srep35064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/23/2016] [Indexed: 01/21/2023] Open
Abstract
Plant defense responses to pathogens involve massive transcriptional reprogramming. Recently, differential coexpression analysis has been developed to study the rewiring of gene networks through microarray data, which is becoming an important complement to traditional differential expression analysis. Using time-series microarray data of Arabidopsis thaliana infected with Pseudomonas syringae, we analyzed Arabidopsis defense responses to P. syringae through differential coexpression analysis. Overall, we found that differential coexpression was a common phenomenon of plant immunity. Genes that were frequently involved in differential coexpression tend to be related to plant immune responses. Importantly, many of those genes have similar average expression levels between normal plant growth and pathogen infection but have different coexpression partners. By integrating the Arabidopsis regulatory network into our analysis, we identified several transcription factors that may be regulators of differential coexpression during plant immune responses. We also observed extensive differential coexpression between genes within the same metabolic pathways. Several metabolic pathways, such as photosynthesis light reactions, exhibited significant changes in expression correlation between normal growth and pathogen infection. Taken together, differential coexpression analysis provides a new strategy for analyzing transcriptional data related to plant defense responses and new insights into the understanding of plant-pathogen interactions.
Collapse
|
182
|
Sinha NR, Rowland SD, Ichihashi Y. Using gene networks in EvoDevo analyses. CURRENT OPINION IN PLANT BIOLOGY 2016; 33:133-139. [PMID: 27455887 DOI: 10.1016/j.pbi.2016.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 06/06/2023]
Abstract
An intricate web of regulatory relationships between DNA, RNA, proteins and metabolites regulates how organisms achieve form and function. Genome sequencing combined with computational methods has allowed us to look at diverse readouts and generate a comprehensive framework for how molecules generate morphological phenotypes. RNAseq has evolved and proved useful for identifying links between transcription factor activity and transcript abundance, and for the generation of transcriptomes in non-model species through de novo assembly. Gene coexpression networks, combined with differential correlation analysis, offer the most versatile gene interaction exploratory tools, using gene expression patterns to determine potential associations and modularity of gene interactions and the differential associations between networks. Networks incorporating different types of biological data can help reduce the complexity of the entirety of biological information into discrete and interpretable pieces of data that can be the starting point for hypothesis construction and testing.
Collapse
Affiliation(s)
- Neelima R Sinha
- Department of Plant Biology, University of California at Davis, Davis, CA 95616, USA.
| | - Steven D Rowland
- Department of Plant Biology, University of California at Davis, Davis, CA 95616, USA
| | - Yasunori Ichihashi
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
183
|
Differential network analysis from cross-platform gene expression data. Sci Rep 2016; 6:34112. [PMID: 27677586 PMCID: PMC5039701 DOI: 10.1038/srep34112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/07/2016] [Indexed: 01/18/2023] Open
Abstract
Understanding how the structure of gene dependency network changes between two patient-specific groups is an important task for genomic research. Although many computational approaches have been proposed to undertake this task, most of them estimate correlation networks from group-specific gene expression data independently without considering the common structure shared between different groups. In addition, with the development of high-throughput technologies, we can collect gene expression profiles of same patients from multiple platforms. Therefore, inferring differential networks by considering cross-platform gene expression profiles will improve the reliability of network inference. We introduce a two dimensional joint graphical lasso (TDJGL) model to simultaneously estimate group-specific gene dependency networks from gene expression profiles collected from different platforms and infer differential networks. TDJGL can borrow strength across different patient groups and data platforms to improve the accuracy of estimated networks. Simulation studies demonstrate that TDJGL provides more accurate estimates of gene networks and differential networks than previous competing approaches. We apply TDJGL to the PI3K/AKT/mTOR pathway in ovarian tumors to build differential networks associated with platinum resistance. The hub genes of our inferred differential networks are significantly enriched with known platinum resistance-related genes and include potential platinum resistance-related genes.
Collapse
|
184
|
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: 23] [Impact Index Per Article: 2.6] [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
|
185
|
Wu S, Li J, Cao M, Yang J, Li YX, Li YY. A novel integrated gene coexpression analysis approach reveals a prognostic three-transcription-factor signature for glioma molecular subtypes. BMC SYSTEMS BIOLOGY 2016; 10 Suppl 3:71. [PMID: 27586240 PMCID: PMC5009532 DOI: 10.1186/s12918-016-0315-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Glioma is the most common brain tumor and it has very high mortality rate due to its infiltration and heterogeneity. Precise classification of glioma subtype is essential for proper therapeutic treatment and better clinical prognosis. However, the molecular mechanism of glioma is far from clear and the classical classification methods based on traditional morphologic and histopathologic knowledge are subjective and inconsistent. Recently, classification methods based on molecular characteristics are developed with rapid progress of high throughput technology. Methods In the present study, we designed a novel integrated gene coexpression analysis approach, which involves differential coexpression and differential regulation analysis (DCEA and DRA), to investigate glioma prognostic biomarkers and molecular subtypes based on six glioma transcriptome data sets. Results We revealed a novel three-transcription-factor signature including AHR, NFIL3 and ZNF423 for glioma molecular subtypes. This three-TF signature clusters glioma patients into three major subtypes (ZG, NG and IG subtypes) which are significantly different in patient survival as well as transcriptomic patterns. Notably, ZG subtype is featured with higher expression of ZNF423 and has better prognosis with younger age at diagnosis. NG subtype is associated with higher expression of NFIL3 and AHR, and has worse prognosis with elder age at diagnosis. According to our inferred differential networking information and previously reported signalling knowledge, we suggested testable hypotheses on the roles of AHR and NFIL3 in glioma carcinogenesis. Conclusions With so far the least biomarkers, our approach not only provides a novel glioma prognostic molecular classification scheme, but also helps to explore its dysregulation mechanisms. Our work is extendable to prognosis-related classification and signature identification in other cancer researches. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0315-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sujuan Wu
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.,Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, China
| | - Junyi Li
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, China.,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mushui Cao
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, China.,School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| | - Jing Yang
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.,Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, China
| | - Yi-Xue Li
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China. .,Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, China. .,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China. .,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, China. .,School of Life Science and Technology, Tongji University, Shanghai, 200092, China. .,Shanghai Engineering Research Center of Pharmaceutical Translation, 1278 Keyuan Road, Shanghai, 201203, China.
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, China. .,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, China. .,Shanghai Engineering Research Center of Pharmaceutical Translation, 1278 Keyuan Road, Shanghai, 201203, China.
| |
Collapse
|
186
|
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
|
187
|
He F, Karve AA, Maslov S, Babst BA. Large-Scale Public Transcriptomic Data Mining Reveals a Tight Connection between the Transport of Nitrogen and Other Transport Processes in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2016; 7:1207. [PMID: 27563305 PMCID: PMC4981021 DOI: 10.3389/fpls.2016.01207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/29/2016] [Indexed: 05/29/2023]
Abstract
Movement of nitrogen to the plant tissues where it is needed for growth is an important contribution to nitrogen use efficiency. However, we have very limited knowledge about the mechanisms of nitrogen transport. Loading of nitrogen into the xylem and/or phloem by transporter proteins is likely important, but there are several families of genes that encode transporters of nitrogenous molecules (collectively referred to as N transporters here), each comprised of many gene members. In this study, we leveraged publicly available microarray data of Arabidopsis to investigate the gene networks of N transporters to elucidate their possible biological roles. First, we showed that tissue-specificity of nitrogen (N) transporters was well reflected among the public microarray data. Then, we built coexpression networks of N transporters, which showed relationships between N transporters and particular aspects of plant metabolism, such as phenylpropanoid biosynthesis and carbohydrate metabolism. Furthermore, genes associated with several biological pathways were found to be tightly coexpressed with N transporters in different tissues. Our coexpression networks provide information at the systems-level that will serve as a resource for future investigation of nitrogen transport systems in plants, including candidate gene clusters that may work together in related biological roles.
Collapse
Affiliation(s)
- Fei He
- Biological, Environmental and Climate Sciences Department, Brookhaven National LaboratoryUpton, NY, USA
| | - Abhijit A. Karve
- Biological, Environmental and Climate Sciences Department, Brookhaven National LaboratoryUpton, NY, USA
- Purdue Research FoundationWest Lafayette, IN, USA
| | - Sergei Maslov
- Biological, Environmental and Climate Sciences Department, Brookhaven National LaboratoryUpton, NY, USA
- Department of Bioengineering, Carl R. Woese Institute for Genomic Biology, National Center for Supercomputing Applications, University of Illinois at Urbana-ChampaignUrbana, IL, USA
| | - Benjamin A. Babst
- Biological, Environmental and Climate Sciences Department, Brookhaven National LaboratoryUpton, NY, USA
- Arkansas Forest Resources Center, The University of Arkansas at MonticelloMonticello, AR, USA
| |
Collapse
|
188
|
Quigley DA, Kandyba E, Huang P, Halliwill KD, Sjölund J, Pelorosso F, Wong CE, Hirst GL, Wu D, Delrosario R, Kumar A, Balmain A. Gene Expression Architecture of Mouse Dorsal and Tail Skin Reveals Functional Differences in Inflammation and Cancer. Cell Rep 2016; 16:1153-1165. [PMID: 27425619 DOI: 10.1016/j.celrep.2016.06.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/16/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
Inherited germline polymorphisms can cause gene expression levels in normal tissues to differ substantially between individuals. We present an analysis of the genetic architecture of normal adult skin from 470 genetically unique mice, demonstrating the effect of germline variants, skin tissue location, and perturbation by exogenous inflammation or tumorigenesis on gene signaling pathways. Gene networks related to specific cell types and signaling pathways, including sonic hedgehog (Shh), Wnt, Lgr family stem cell markers, and keratins, differed at these tissue sites, suggesting mechanisms for the differential susceptibility of dorsal and tail skin to development of skin diseases and tumorigenesis. The Pten tumor suppressor gene network is rewired in premalignant tumors compared to normal tissue, but this response to perturbation is lost during malignant progression. We present a software package for expression quantitative trait loci (eQTL) network analysis and demonstrate how network analysis of whole tissues provides insights into interactions between cell compartments and signaling molecules.
Collapse
Affiliation(s)
- David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo 0310, Norway; K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo 0313, Norway; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Eve Kandyba
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Phillips Huang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Genome Institute of Singapore, 60 Biopolis Street, #02-01 Genome Building, Singapore 138672, Singapore
| | - Kyle D Halliwill
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jonas Sjölund
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
| | - Facundo Pelorosso
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 9(th) Floor, Ciudad Autónoma de Buenos Aires 1121, Argentina
| | - Christine E Wong
- Institute of Surgical Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Gillian L Hirst
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Di Wu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Reyno Delrosario
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Atul Kumar
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
189
|
Chang J, Zhou W, Zhou WX, Wang L. Comparing large covariance matrices under weak conditions on the dependence structure and its application to gene clustering. Biometrics 2016; 73:31-41. [PMID: 27377648 DOI: 10.1111/biom.12552] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 04/01/2016] [Accepted: 05/01/2016] [Indexed: 12/30/2022]
Abstract
Comparing large covariance matrices has important applications in modern genomics, where scientists are often interested in understanding whether relationships (e.g., dependencies or co-regulations) among a large number of genes vary between different biological states. We propose a computationally fast procedure for testing the equality of two large covariance matrices when the dimensions of the covariance matrices are much larger than the sample sizes. A distinguishing feature of the new procedure is that it imposes no structural assumptions on the unknown covariance matrices. Hence, the test is robust with respect to various complex dependence structures that frequently arise in genomics. We prove that the proposed procedure is asymptotically valid under weak moment conditions. As an interesting application, we derive a new gene clustering algorithm which shares the same nice property of avoiding restrictive structural assumptions for high-dimensional genomics data. Using an asthma gene expression dataset, we illustrate how the new test helps compare the covariance matrices of the genes across different gene sets/pathways between the disease group and the control group, and how the gene clustering algorithm provides new insights on the way gene clustering patterns differ between the two groups. The proposed methods have been implemented in an R-package HDtest and are available on CRAN.
Collapse
Affiliation(s)
- Jinyuan Chang
- School of Statistics, Southwestern University of Finance and Economics, Chengdu, Sichuan 611130, China.,School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Wen Zhou
- Department of Statistics, Colorado State University, Fort Collins, Colorado 80523, U.S.A
| | - Wen-Xin Zhou
- Department of Operations Research and Financial Engineering, Princeton University, Princeton, New Jersey 08544, U.S.A
| | - Lan Wang
- School of Statistics, University of Minnesota, Minneapolis, Minnesota 55455, U.S.A
| |
Collapse
|
190
|
Gaiteri C, Mostafavi S, Honey CJ, De Jager PL, Bennett DA. Genetic variants in Alzheimer disease - molecular and brain network approaches. Nat Rev Neurol 2016; 12:413-27. [PMID: 27282653 PMCID: PMC5017598 DOI: 10.1038/nrneurol.2016.84] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetic studies in late-onset Alzheimer disease (LOAD) are aimed at identifying core disease mechanisms and providing potential biomarkers and drug candidates to improve clinical care of AD. However, owing to the complexity of LOAD, including pathological heterogeneity and disease polygenicity, extraction of actionable guidance from LOAD genetics has been challenging. Past attempts to summarize the effects of LOAD-associated genetic variants have used pathway analysis and collections of small-scale experiments to hypothesize functional convergence across several variants. In this Review, we discuss how the study of molecular, cellular and brain networks provides additional information on the effects of LOAD-associated genetic variants. We then discuss emerging combinations of these omic data sets into multiscale models, which provide a more comprehensive representation of the effects of LOAD-associated genetic variants at multiple biophysical scales. Furthermore, we highlight the clinical potential of mechanistically coupling genetic variants and disease phenotypes with multiscale brain models.
Collapse
Affiliation(s)
- Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S Paulina Street, Chicago, Illinois 60612, USA
| | - Sara Mostafavi
- Department of Statistics, and Medical Genetics; Centre for Molecular and Medicine and Therapeutics, University of British Columbia, 950 West 28th Avenue, Vancouver, British Columbia V5Z 4H4, Canada
| | - Christopher J Honey
- Department of Psychology, University of Toronto, 100 St. George Street, 4th Floor Sidney Smith Hall, Toronto, Ontario M5S 3G3, Canada
| | - Philip L De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 75 Francis Street, Boston MA 02115, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S Paulina Street, Chicago, Illinois 60612, USA
| |
Collapse
|
191
|
Xu Y, Yue W, Yao Shugart Y, Li S, Cai L, Li Q, Cheng Z, Wang G, Zhou Z, Jin C, Yuan J, Tian L, Wang J, Zhang K, Zhang K, Liu S, Song Y, Zhang F. Exploring Transcription Factors-microRNAs Co-regulation Networks in Schizophrenia. Schizophr Bull 2016; 42:1037-45. [PMID: 26609121 PMCID: PMC4903044 DOI: 10.1093/schbul/sbv170] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Transcriptional factors (TFs) and microRNAs (miRNAs) have been recognized as 2 classes of principal gene regulators that may be responsible for genome coexpression changes observed in schizophrenia (SZ). METHODS This study aims to (1) identify differentially coexpressed genes (DCGs) in 3 mRNA expression microarray datasets; (2) explore potential interactions among the DCGs, and differentially expressed miRNAs identified in our dataset composed of early-onset SZ patients and healthy controls; (3) validate expression levels of some key transcripts; and (4) explore the druggability of DCGs using the curated database. RESULTS We detected a differential coexpression network associated with SZ and found that 9 out of the 12 regulators were replicated in either of the 2 other datasets. Leveraging the differentially expressed miRNAs identified in our previous dataset, we constructed a miRNA-TF-gene network relevant to SZ, including an EGR1-miR-124-3p-SKIL feed-forward loop. Our real-time quantitative PCR analysis indicated the overexpression of miR-124-3p, the under expression of SKIL and EGR1 in the blood of SZ patients compared with controls, and the direction of change of miR-124-3p and SKIL mRNA levels in SZ cases were reversed after a 12-week treatment cycle. Our druggability analysis revealed that many of these genes have the potential to be drug targets. CONCLUSIONS Together, our results suggest that coexpression network abnormalities driven by combinatorial and interactive action from TFs and miRNAs may contribute to the development of SZ and be relevant to the clinical treatment of the disease.
Collapse
Affiliation(s)
- Yong Xu
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China;,These authors contributed equally to this work
| | - Weihua Yue
- Department of Psychiatry, Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China;,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, China;,These authors contributed equally to this work
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD;,These authors contributed equally to this work
| | - Sheng Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Cai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Li
- Shanghai Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
| | - Zaohuo Cheng
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Guoqiang Wang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Zhenhe Zhou
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Chunhui Jin
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jianmin Yuan
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Lin Tian
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jun Wang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Kai Zhang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Kerang Zhang
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China
| | - Sha Liu
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuqing Song
- Department of Psychiatry, Institute of Mental Health, Sixth Hospital, Peking University, Beijing, China
| | - Fuquan Zhang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| |
Collapse
|
192
|
Wu Y, Fu X, Wang L. Identification of novel biomarkers for preeclampsia on the basis of differential expression network analysis. Exp Ther Med 2016; 12:201-207. [PMID: 27347039 PMCID: PMC4906647 DOI: 10.3892/etm.2016.3261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 02/11/2016] [Indexed: 12/27/2022] Open
Abstract
Preeclampsia (PE) is a severe pregnancy complication, which is a leading cause of maternal and fetal mortality. The present study aimed to screen potential biomarkers for the diagnosis and prediction of PE and to investigate the underlying mechanisms of PE development based on the differential expression network (DEN). The microarray datasets E-GEOD-6573 and E-GEOD-48424 were downloaded from the European Bioinformatics Institute database. Differentially expressed genes (DEGs) between the PE and normal groups were screened by Significant Analysis of Microarrays with the cutoff value of a |log2 fold change| of >2, and a false discovery rate of <0.05. The DEN was constructed based on the differential and non-differential interactions observed. In addition, genes with higher connectivity degrees in the DEN were identified on the basis of centrality analysis, while disease genes were also extracted from the DEN. In order to understand the functional roles of genes in DEN, Gene Ontology (GO) and pathway enrichment analyses were performed. The present results indicated that a total of 225 genes were considered as DEGs in the PE group, while 466 nodes and 314 gene interactions were involved in the DEN. Among these 466 nodes, 4 nodes with higher degrees were identified, including ubiquitin C (UBC), small ubiquitin-like modifier 1 (SUMO1), SUMO2 and RAD21 homolog (S. pombe) (RAD21). Notably, UBC was also found to be a disease gene. UBC, RAD21, SUMO2 and SUMO1 were markedly enriched in the regulation of programmed cell death, as well as in the regulation of apoptosis, cell cycle and chromosomal part. In conclusion, based on these results, we suggest that UBC, RAD21, SUMO2 and SUMO1 may be reliable biomarkers for the prediction of the development and progression of PE.
Collapse
Affiliation(s)
- Yufang Wu
- Department of Gynecology, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Xiuhua Fu
- Department of Obstetrics, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Lin Wang
- Department of Obstetrics and Gynecology, Xijing Hospital, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shanxi 710032, P.R. China
| |
Collapse
|
193
|
Creanza TM, Liguori M, Liuni S, Nuzziello N, Ancona N. Meta-Analysis of Differential Connectivity in Gene Co-Expression Networks in Multiple Sclerosis. Int J Mol Sci 2016; 17:E936. [PMID: 27314336 PMCID: PMC4926469 DOI: 10.3390/ijms17060936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/09/2016] [Accepted: 05/24/2016] [Indexed: 12/20/2022] Open
Abstract
Differential gene expression analyses to investigate multiple sclerosis (MS) molecular pathogenesis cannot detect genes harboring genetic and/or epigenetic modifications that change the gene functions without affecting their expression. Differential co-expression network approaches may capture changes in functional interactions resulting from these alterations. We re-analyzed 595 mRNA arrays from publicly available datasets by studying changes in gene co-expression networks in MS and in response to interferon (IFN)-β treatment. Interestingly, MS networks show a reduced connectivity relative to the healthy condition, and the treatment activates the transcription of genes and increases their connectivity in MS patients. Importantly, the analysis of changes in gene connectivity in MS patients provides new evidence of association for genes already implicated in MS by single-nucleotide polymorphism studies and that do not show differential expression. This is the case of amiloride-sensitive cation channel 1 neuronal (ACCN1) that shows a reduced number of interacting partners in MS networks, and it is known for its role in synaptic transmission and central nervous system (CNS) development. Furthermore, our study confirms a deregulation of the vitamin D system: among the transcription factors that potentially regulate the deregulated genes, we find TCF3 and SP1 that are both involved in vitamin D3-induced p27Kip1 expression. Unveiling differential network properties allows us to gain systems-level insights into disease mechanisms and may suggest putative targets for the treatment.
Collapse
Affiliation(s)
- Teresa Maria Creanza
- Institute of Intelligent Systems for Automation, National Research Council of Italy, 70126 Bari, Italy.
- Center for Complex Systems in Molecular Biology and Medicine, University of Turin, 10123 Turin, Italy.
| | - Maria Liguori
- Institute of Biomedical Technologies, National Research Council of Italy, 70126 Bari, Italy.
| | - Sabino Liuni
- Institute of Biomedical Technologies, National Research Council of Italy, 70126 Bari, Italy.
| | - Nicoletta Nuzziello
- Institute of Biomedical Technologies, National Research Council of Italy, 70126 Bari, Italy.
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, 70126 Bari, Italy.
| | - Nicola Ancona
- Institute of Intelligent Systems for Automation, National Research Council of Italy, 70126 Bari, Italy.
| |
Collapse
|
194
|
Karničar K, Drobnak I, Petek M, Magdevska V, Horvat J, Vidmar R, Baebler Š, Rotter A, Jamnik P, Fujs Š, Turk B, Fonovič M, Gruden K, Kosec G, Petković H. Integrated omics approaches provide strategies for rapid erythromycin yield increase in Saccharopolyspora erythraea. Microb Cell Fact 2016; 15:93. [PMID: 27255285 PMCID: PMC4891893 DOI: 10.1186/s12934-016-0496-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 05/25/2016] [Indexed: 11/24/2022] Open
Abstract
Background Omics approaches have significantly increased our understanding of biological systems. However, they have had limited success in explaining the dramatically increased productivity of commercially important natural products by industrial high-producing strains, such as the erythromycin-producing actinomycete Saccharopolyspora erythraea. Further yield increase is of great importance but requires a better understanding of the underlying physiological processes. Results To reveal the mechanisms related to erythromycin yield increase, we have undertaken an integrated study of the genomic, transcriptomic, and proteomic differences between the wild type strain NRRL2338 (WT) and the industrial high-producing strain ABE1441 (HP) of S. erythraea at multiple time points of a simulated industrial bioprocess. 165 observed mutations lead to differences in gene expression profiles and protein abundance between the two strains, which were most prominent in the initial stages of erythromycin production. Enzymes involved in erythromycin biosynthesis, metabolism of branched chain amino acids and proteolysis were most strongly upregulated in the HP strain. Interestingly, genes related to TCA cycle and DNA-repair were downregulated. Additionally, comprehensive data analysis uncovered significant correlations in expression profiles of the erythromycin-biosynthetic genes, other biosynthetic gene clusters and previously unidentified putative regulatory genes. Based on this information, we demonstrated that overexpression of several genes involved in amino acid metabolism can contribute to increased yield of erythromycin, confirming the validity of our systems biology approach. Conclusions Our comprehensive omics approach, carried out in industrially relevant conditions, enabled the identification of key pathways affecting erythromycin yield and suggests strategies for rapid increase in the production of secondary metabolites in industrial environment. Electronic supplementary material The online version of this article (doi:10.1186/s12934-016-0496-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Igor Drobnak
- Acies Bio, d.o.o., Tehnološki park 21, SI-1000, Ljubljana, Slovenia.,Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1000, Ljubljana, Slovenia
| | - Marko Petek
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 111, SI-1000, Ljubljana, Slovenia
| | | | - Jaka Horvat
- Acies Bio, d.o.o., Tehnološki park 21, SI-1000, Ljubljana, Slovenia
| | - Robert Vidmar
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Jamova cesta 39, SI-1000, Ljubljana, Slovenia.,International Postgraduate School Jožef Stefan, Jamova cesta 39, SI-1000, Ljubljana, Slovenia
| | - Špela Baebler
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 111, SI-1000, Ljubljana, Slovenia
| | - Ana Rotter
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 111, SI-1000, Ljubljana, Slovenia
| | - Polona Jamnik
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000, Ljubljana, Slovenia
| | - Štefan Fujs
- Acies Bio, d.o.o., Tehnološki park 21, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Jamova cesta 39, SI-1000, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Aškerčeva cesta 5, SI-1000, Ljubljana, Slovenia.,Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova cesta 39, SI-1000, Ljubljana, Slovenia
| | - Marko Fonovič
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Jamova cesta 39, SI-1000, Ljubljana, Slovenia.,Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova cesta 39, SI-1000, Ljubljana, Slovenia
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 111, SI-1000, Ljubljana, Slovenia
| | - Gregor Kosec
- Acies Bio, d.o.o., Tehnološki park 21, SI-1000, Ljubljana, Slovenia.
| | - Hrvoje Petković
- Acies Bio, d.o.o., Tehnološki park 21, SI-1000, Ljubljana, Slovenia. .,Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
195
|
Yang J, Wu SJ, Yang SY, Peng JW, Wang SN, Wang FY, Song YX, Qi T, Li YX, Li YY. DNetDB: The human disease network database based on dysfunctional regulation mechanism. BMC SYSTEMS BIOLOGY 2016; 10:36. [PMID: 27209279 PMCID: PMC4875653 DOI: 10.1186/s12918-016-0280-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/05/2016] [Indexed: 11/18/2022]
Abstract
Disease similarity study provides new insights into disease taxonomy, pathogenesis, which plays a guiding role in diagnosis and treatment. The early studies were limited to estimate disease similarities based on clinical manifestations, disease-related genes, medical vocabulary concepts or registry data, which were inevitably biased to well-studied diseases and offered small chance of discovering novel findings in disease relationships. In other words, genome-scale expression data give us another angle to address this problem since simultaneous measurement of the expression of thousands of genes allows for the exploration of gene transcriptional regulation, which is believed to be crucial to biological functions. Although differential expression analysis based methods have the potential to explore new disease relationships, it is difficult to unravel the upstream dysregulation mechanisms of diseases. We therefore estimated disease similarities based on gene expression data by using differential coexpression analysis, a recently emerging method, which has been proved to be more potential to capture dysfunctional regulation mechanisms than differential expression analysis. A total of 1,326 disease relationships among 108 diseases were identified, and the relevant information constituted the human disease network database (DNetDB). Benefiting from the use of differential coexpression analysis, the potential common dysfunctional regulation mechanisms shared by disease pairs (i.e. disease relationships) were extracted and presented. Statistical indicators, common disease-related genes and drugs shared by disease pairs were also included in DNetDB. In total, 1,326 disease relationships among 108 diseases, 5,598 pathways, 7,357 disease-related genes and 342 disease drugs are recorded in DNetDB, among which 3,762 genes and 148 drugs are shared by at least two diseases. DNetDB is the first database focusing on disease similarity from the viewpoint of gene regulation mechanism. It provides an easy-to-use web interface to search and browse the disease relationships and thus helps to systematically investigate etiology and pathogenesis, perform drug repositioning, and design novel therapeutic interventions. Database URL: http://app.scbit.org/DNetDB/#.
Collapse
Affiliation(s)
- Jing Yang
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P.R. China
| | - Su-Juan Wu
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China
| | - Shao-You Yang
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China
| | - Jia-Wei Peng
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China
| | - Shi-Nuo Wang
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China
| | - Fu-Yan Wang
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China
| | - Yu-Xing Song
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China
| | - Ting Qi
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China.,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China
| | - Yi-Xue Li
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China. .,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P.R. China. .,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China. .,Shanghai Engineering Research Center of Pharmaceutical Translation, 1278 Keyuan Road, Shanghai, 201203, P.R. China.
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, Shanghai, 200235, P.R. China. .,Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, P.R. China. .,Shanghai Engineering Research Center of Pharmaceutical Translation, 1278 Keyuan Road, Shanghai, 201203, P.R. China.
| |
Collapse
|
196
|
Cai TT, Liu W. Large-Scale Multiple Testing of Correlations. J Am Stat Assoc 2016; 111:229-240. [PMID: 27284211 PMCID: PMC4894362 DOI: 10.1080/01621459.2014.999157] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 11/01/2014] [Indexed: 10/24/2022]
Abstract
Multiple testing of correlations arises in many applications including gene coexpression network analysis and brain connectivity analysis. In this paper, we consider large scale simultaneous testing for correlations in both the one-sample and two-sample settings. New multiple testing procedures are proposed and a bootstrap method is introduced for estimating the proportion of the nulls falsely rejected among all the true nulls. The properties of the proposed procedures are investigated both theoretically and numerically. It is shown that the procedures asymptotically control the overall false discovery rate and false discovery proportion at the nominal level. Simulation results show that the methods perform well numerically in terms of both the size and power of the test and it significantly outperforms two alternative methods. The two-sample procedure is also illustrated by an analysis of a prostate cancer dataset for the detection of changes in coexpression patterns between gene expression levels.
Collapse
Affiliation(s)
- T Tony Cai
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104 ( )
| | - Weidong Liu
- Department of Mathematics, Institute of Natural Sciences and MOE-LSC, Shanghai Jiao Tong University, Shanghai, China ( ).
| |
Collapse
|
197
|
Jaeger PA, Lucin KM, Britschgi M, Vardarajan B, Huang RP, Kirby ED, Abbey R, Boeve BF, Boxer AL, Farrer LA, Finch N, Graff-Radford NR, Head E, Hofree M, Huang R, Johns H, Karydas A, Knopman DS, Loboda A, Masliah E, Narasimhan R, Petersen RC, Podtelezhnikov A, Pradhan S, Rademakers R, Sun CH, Younkin SG, Miller BL, Ideker T, Wyss-Coray T. Network-driven plasma proteomics expose molecular changes in the Alzheimer's brain. Mol Neurodegener 2016; 11:31. [PMID: 27112350 PMCID: PMC4845325 DOI: 10.1186/s13024-016-0095-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/08/2016] [Indexed: 12/17/2022] Open
Abstract
Background Biological pathways that significantly contribute to sporadic Alzheimer’s disease are largely unknown and cannot be observed directly. Cognitive symptoms appear only decades after the molecular disease onset, further complicating analyses. As a consequence, molecular research is often restricted to late-stage post-mortem studies of brain tissue. However, the disease process is expected to trigger numerous cellular signaling pathways and modulate the local and systemic environment, and resulting changes in secreted signaling molecules carry information about otherwise inaccessible pathological processes. Results To access this information we probed relative levels of close to 600 secreted signaling proteins from patients’ blood samples using antibody microarrays and mapped disease-specific molecular networks. Using these networks as seeds we then employed independent genome and transcriptome data sets to corroborate potential pathogenic pathways. Conclusions We identified Growth-Differentiation Factor (GDF) signaling as a novel Alzheimer’s disease-relevant pathway supported by in vivo and in vitro follow-up experiments, demonstrating the existence of a highly informative link between cellular pathology and changes in circulatory signaling proteins. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0095-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Philipp A Jaeger
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,Institute of Chemistry and Biochemistry, Free University Berlin, Berlin, Germany. .,Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Kurt M Lucin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Present address: Biology Department, Eastern Connecticut State University, Willimantic, CT, USA
| | - Markus Britschgi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Present address: Roche Pharma Research and Early Development, NORD DTA, Roche Innovation, Center Basel, Basel, Switzerland
| | - Badri Vardarajan
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine, Boston, MA, USA
| | - Ruo-Pan Huang
- RayBiotech, Guangzhou, China.,RayBiotech, Norcrosse, GA, USA
| | - Elizabeth D Kirby
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Rachelle Abbey
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Adam L Boxer
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine, Boston, MA, USA.,Departments of Neurology, Ophthalmology, Genetics and Genomics, Epidemiology, and Biostatistics, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - NiCole Finch
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Elizabeth Head
- Departments of Pharmacology and Nutritional Sciences and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Matan Hofree
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Ruochun Huang
- RayBiotech, Guangzhou, China.,RayBiotech, Norcrosse, GA, USA
| | - Hudson Johns
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Anna Karydas
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | | | - Andrey Loboda
- Genetics and Pharmacogenomics, Merck Research Laboratories, West Point, PA, USA
| | - Eliezer Masliah
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Ramya Narasimhan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Suraj Pradhan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Chung-Huan Sun
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Bruce L Miller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Trey Ideker
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,Center for Tissue Regeneration, Repair and Restoration, VA Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
198
|
Padayachee T, Khamiakova T, Shkedy Z, Perola M, Salo P, Burzykowski T. The Detection of Metabolite-Mediated Gene Module Co-Expression Using Multivariate Linear Models. PLoS One 2016; 11:e0150257. [PMID: 26918614 PMCID: PMC4769021 DOI: 10.1371/journal.pone.0150257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 02/11/2016] [Indexed: 12/29/2022] Open
Abstract
Investigating whether metabolites regulate the co-expression of a predefined gene module is one of the relevant questions posed in the integrative analysis of metabolomic and transcriptomic data. This article concerns the integrative analysis of the two high-dimensional datasets by means of multivariate models and statistical tests for the dependence between metabolites and the co-expression of a gene module. The general linear model (GLM) for correlated data that we propose models the dependence between adjusted gene expression values through a block-diagonal variance-covariance structure formed by metabolic-subset specific general variance-covariance blocks. Performance of statistical tests for the inference of conditional co-expression are evaluated through a simulation study. The proposed methodology is applied to the gene expression data of the previously characterized lipid-leukocyte module. Our results show that the GLM approach improves on a previous approach by being less prone to the detection of spurious conditional co-expression.
Collapse
Affiliation(s)
- Trishanta Padayachee
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-Biostat), Hasselt University, Diepenbeek, Belgium
- * E-mail:
| | - Tatsiana Khamiakova
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-Biostat), Hasselt University, Diepenbeek, Belgium
| | - Ziv Shkedy
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-Biostat), Hasselt University, Diepenbeek, Belgium
| | - Markus Perola
- Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Finland
| | - Perttu Salo
- Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Finland
| | - Tomasz Burzykowski
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-Biostat), Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
199
|
Inferring causal molecular networks: empirical assessment through a community-based effort. Nat Methods 2016; 13:310-8. [PMID: 26901648 PMCID: PMC4854847 DOI: 10.1038/nmeth.3773] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 01/21/2016] [Indexed: 01/08/2023]
Abstract
The HPN-DREAM community challenge assessed the ability of computational methods to infer causal molecular networks, focusing specifically on the task of inferring causal protein signaling networks in cancer cell lines. It remains unclear whether causal, rather than merely correlational, relationships in molecular networks can be inferred in complex biological settings. Here we describe the HPN-DREAM network inference challenge, which focused on learning causal influences in signaling networks. We used phosphoprotein data from cancer cell lines as well as in silico data from a nonlinear dynamical model. Using the phosphoprotein data, we scored more than 2,000 networks submitted by challenge participants. The networks spanned 32 biological contexts and were scored in terms of causal validity with respect to unseen interventional data. A number of approaches were effective, and incorporating known biology was generally advantageous. Additional sub-challenges considered time-course prediction and visualization. Our results suggest that learning causal relationships may be feasible in complex settings such as disease states. Furthermore, our scoring approach provides a practical way to empirically assess inferred molecular networks in a causal sense.
Collapse
|
200
|
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
|