1
|
Iyer KR, Clarke SL, Guarischi‐Sousa R, Gjoni K, Heath AS, Young EP, Stitziel NO, Laurie C, Broome JG, Khan AT, Lewis JP, Xu H, Montasser ME, Ashley KE, Hasbani NR, Boerwinkle E, Morrison AC, Chami N, Do R, Rocheleau G, Lloyd‐Jones DM, Lemaitre RN, Bis JC, Floyd JS, Kinney GL, Bowden DW, Palmer ND, Benjamin EJ, Nayor M, Yanek LR, Kral BG, Becker LC, Kardia SLR, Smith JA, Bielak LF, Norwood AF, Min Y, Carson AP, Post WS, Rich SS, Herrington D, Guo X, Taylor KD, Manson JE, Franceschini N, Pollard KS, Mitchell BD, Loos RJF, Fornage M, Hou L, Psaty BM, Young KA, Regan EA, Freedman BI, Vasan RS, Levy D, Mathias RA, Peyser PA, Raffield LM, Kooperberg C, Reiner AP, Rotter JI, Jun G, de Vries PS, Assimes TL. Unveiling the Genetic Landscape of Coronary Artery Disease Through Common and Rare Structural Variants. J Am Heart Assoc 2025; 14:e036499. [PMID: 39950338 PMCID: PMC12074758 DOI: 10.1161/jaha.124.036499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/21/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND Genome-wide association studies have identified several hundred susceptibility single nucleotide variants for coronary artery disease (CAD). Despite single nucleotide variant-based genome-wide association studies improving our understanding of the genetics of CAD, the contribution of structural variants (SVs) to the risk of CAD remains largely unclear. METHOD AND RESULTS We leveraged SVs detected from high-coverage whole genome sequencing data in a diverse group of participants from the National Heart Lung and Blood Institute's Trans-Omics for Precision Medicine program. Single variant tests were performed on 58 706 SVs in a study sample of 11 556 CAD cases and 42 907 controls. Additionally, aggregate tests using sliding windows were performed to examine rare SVs. One genome-wide significant association was identified for a common biallelic intergenic duplication on chromosome 6q21 (P=1.54E-09, odds ratio=1.34). The sliding window-based aggregate tests found 1 region on chromosome 17q25.3, overlapping USP36, to be significantly associated with coronary artery disease (P=1.03E-10). USP36 is highly expressed in arterial and adipose tissues while broadly affecting several cardiometabolic traits. CONCLUSIONS Our results suggest that SVs, both common and rare, may influence the risk of coronary artery disease.
Collapse
Affiliation(s)
- Kruthika R. Iyer
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| | - Shoa L. Clarke
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
- Department of Medicine, Stanford Prevention Research CenterStanford University School of MedicineStanfordCAUSA
| | - Rodrigo Guarischi‐Sousa
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| | - Ketrin Gjoni
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCAUSA
| | - Adam S. Heath
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Erica P. Young
- Department of Medicine, Division of CardiologyWashington University School of MedicineSaint LouisMOUSA
- McDonnell Genome Institute, Washington University School of MedicineSaint LouisMOUSA
| | - Nathan O. Stitziel
- Department of Medicine, Division of CardiologyWashington University School of MedicineSaint LouisMOUSA
- McDonnell Genome Institute, Washington University School of MedicineSaint LouisMOUSA
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Cecelia Laurie
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Jai G. Broome
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
- Department of Medicine, Division of Internal MedicineUniversity of WashingtonSeattleWAUSA
| | - Alyna T. Khan
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Joshua P. Lewis
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Huichun Xu
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - May E. Montasser
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Kellan E. Ashley
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Natalie R. Hasbani
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Human Genome Sequencing CenterBaylor College of MedicineHoustonTXUSA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Ron Do
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Rozenn N. Lemaitre
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
| | - Joshua C. Bis
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
| | - James S. Floyd
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
| | - Gregory L. Kinney
- Department of EpidemiologyColorado School of Public HealthAuroraCOUSA
| | - Donald W. Bowden
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Nicholette D. Palmer
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Emelia J. Benjamin
- Department of Medicine, Cardiovascular Medicine, Boston Medical CenterBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMAUSA
| | - Matthew Nayor
- Department of Medicine, Cardiovascular MedicineBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
- Department of Medicine, Preventive Medicine & EpidemiologyBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
| | - Lisa R. Yanek
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Brian G. Kral
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Lewis C. Becker
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Sharon L. R. Kardia
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Jennifer A. Smith
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
- Institute for Social ResearchSurvey Research Center, University of MichiganAnn ArborMIUSA
| | - Lawrence F. Bielak
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Arnita F. Norwood
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Yuan‐I Min
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - April P. Carson
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Wendy S. Post
- Department of Medicine, Division of CardiologyJohns Hopkins UniversityBaltimoreMDUSA
| | - Stephen S. Rich
- Department of Genome SciencesUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - David Herrington
- Department of MedicineWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Kent D. Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - JoAnn E. Manson
- Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - Nora Franceschini
- Department of EpidemiologyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Katherine S. Pollard
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCAUSA
- Chan Zuckerberg BiohubSan FranciscoCAUSA
| | - Braxton D. Mitchell
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
- Geriatric Research and Education Clinical CenterBaltimore Veterans Administration Medical CenterBaltimoreMDUSA
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Myriam Fornage
- Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - Lifang Hou
- Department of Preventive MedicineNorthwestern UniversityChicagoILUSA
| | - Bruce M. Psaty
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
- Department of Health Systems and Population HealthUniversity of WashingtonSeattleWAUSA
| | - Kendra A. Young
- Department of EpidemiologyColorado School of Public HealthAuroraCOUSA
| | | | - Barry I. Freedman
- Department of Internal Medicine, Section on NephrologyWake Forest University School of MedicineWinston‐SalemNCUSA
| | | | - Daniel Levy
- Division of Intramural Research, Population Sciences BranchNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Rasika A. Mathias
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Patricia A. Peyser
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Laura M. Raffield
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | | | - Alex P. Reiner
- Division of Public HealthFred Hutchinson Cancer CenterSeattleWAUSA
| | - Jerome I. Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Goo Jun
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Paul S. de Vries
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Themistocles L. Assimes
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
- VA Palo Alto Healthcare SystemPalo AltoCAUSA
| |
Collapse
|
2
|
Ni Y, He J, Chalise P. Integration of differential expression and network structure for 'omics data analysis. Comput Biol Med 2022; 150:106133. [PMID: 36179515 DOI: 10.1016/j.compbiomed.2022.106133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/23/2022] [Accepted: 09/18/2022] [Indexed: 11/25/2022]
Abstract
Differential expression (DE) analysis has been routinely used to identify molecular features that are statistically significantly different between distinct biological groups. In recent years, differential network (DN) analysis has emerged as a powerful approach to uncover molecular network structure changes from one biological condition to the other where the molecular features with larger topological changes are selected as biomarkers. Although a large number of DE and a few DN-based methods are available, they have been usually implemented independently. DE analysis ignores the relationship among molecular features while DN analysis does not account for the expression changes at individual level. Therefore, an integrative analysis approach that accounts for both DE and DN is required to identify disease associated key features. Although, a handful of methods have been proposed, there is no method that optimizes the combination of DE and DN. We propose a novel integrative analysis method, DNrank, to identify disease-associated molecular features that leverages the strengths of both DE and DN by calculating a weight using resampling based cross validation scheme within the algorithm. First, differential expression analysis of individual molecular features is carried out. Second, a differential network structure is constructed using the differential partial correlation analysis. Third, the molecular features are ranked in the order of their significances by integrating their DE measures and DN structure using the modified Google's PageRank algorithm. In the algorithm, the optimum combination of DE and DN analyses is achieved by evaluating the prediction performance of top-ranked features utilizing support vector machine classifier with Monte Carlo cross validation. The proposed method is illustrated using both simulated data and three real data sets. The results show that the proposed method has a better performance in identifying important molecular features with respect to predictive discrimination. Also, as compared to existing feature selection methods, the top-ranked features selected by our method had a higher stability in selection. DNrank allows the researchers to identify the disease-associated features by utilizing both expression and network topology changes between two groups.
Collapse
Affiliation(s)
- Yonghui Ni
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Jianghua He
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| |
Collapse
|
3
|
Kim E, Novak LC, Lin C, Colic M, Bertolet LL, Gheorghe V, Bristow CA, Hart T. Dynamic rewiring of biological activity across genotype and lineage revealed by context-dependent functional interactions. Genome Biol 2022; 23:140. [PMID: 35768873 PMCID: PMC9241233 DOI: 10.1186/s13059-022-02712-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 06/17/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Coessentiality networks derived from CRISPR screens in cell lines provide a powerful framework for identifying functional modules in the cell and for inferring the roles of uncharacterized genes. However, these networks integrate signal across all underlying data and can mask strong interactions that occur in only a subset of the cell lines analyzed. RESULTS Here, we decipher dynamic functional interactions by identifying significant cellular contexts, primarily by oncogenic mutation, lineage, and tumor type, and discovering coessentiality relationships that depend on these contexts. We recapitulate well-known gene-context interactions such as oncogene-mutation, paralog buffering, and tissue-specific essential genes, show how mutation rewires known signal transduction pathways, including RAS/RAF and IGF1R-PIK3CA, and illustrate the implications for drug targeting. We further demonstrate how context-dependent functional interactions can elucidate lineage-specific gene function, as illustrated by the maturation of proreceptors IGF1R and MET by proteases FURIN and CPD. CONCLUSIONS This approach advances our understanding of context-dependent interactions and how they can be gleaned from these data. We provide an online resource to explore these context-dependent interactions at diffnet.hart-lab.org.
Collapse
Affiliation(s)
- Eiru Kim
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Present Address: Novartis Institutes for BioMedical Research (NIBR), San Diego, CA, USA
| | - Lance C Novak
- TRACTION, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chenchu Lin
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Medina Colic
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lori L Bertolet
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Veronica Gheorghe
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher A Bristow
- TRACTION, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Traver Hart
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
Li C, Gao Z, Su B, Xu G, Lin X. Data analysis methods for defining biomarkers from omics data. Anal Bioanal Chem 2021; 414:235-250. [PMID: 34951658 DOI: 10.1007/s00216-021-03813-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 02/01/2023]
Abstract
Omics mainly includes genomics, epigenomics, transcriptomics, proteomics and metabolomics. The rapid development of omics technology has opened up new ways to study disease diagnosis and prognosis and to define prospective information of complex diseases. Since omics data are usually large and complex, the method used to analyze the data and to define important information is crucial in omics study. In this review, we focus on advances in biomarker discovery methods based on omics data in the last decade, and categorize them as individual feature analysis, combinatorial feature analysis and network analysis. We also discuss the challenges and perspectives in this field.
Collapse
Affiliation(s)
- Chao Li
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| | - Zhenbo Gao
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China
| | - Benzhe Su
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| | - Xiaohui Lin
- School of Computer Science and Technology, Dalian University of Technology, Dalian, 116024, Liaoning, China.
| |
Collapse
|
5
|
Charwudzi A, Meng Y, Hu L, Ding C, Pu L, Li Q, Xu M, Zhai Z, Xiong S. Integrated bioinformatics analysis reveals dynamic candidate genes and signaling pathways involved in the progression and prognosis of diffuse large B-cell lymphoma. PeerJ 2021; 9:e12394. [PMID: 34760386 PMCID: PMC8570165 DOI: 10.7717/peerj.12394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023] Open
Abstract
Background Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous malignancy with varied outcomes. However, the fundamental mechanisms remain to be fully defined. Aim We aimed to identify core differentially co-expressed hub genes and perturbed pathways relevant to the pathogenesis and prognosis of DLBCL. Methods We retrieved the raw gene expression profile and clinical information of GSE12453 from the Gene Expression Omnibus (GEO) database. We used integrated bioinformatics analysis to identify differentially co-expressed genes. The CIBERSORT analysis was also applied to predict tumor-infiltrating immune cells (TIICs) in the GSE12453 dataset. We performed survival and ssGSEA (single-sample Gene Set Enrichment Analysis) (for TIICs) analyses and validated the hub genes using GEPIA2 and an independent GSE31312 dataset. Results We identified 46 differentially co-expressed hub genes in the GSE12453 dataset. Gene expression levels and survival analysis found 15 differentially co-expressed core hub genes. The core genes prognostic values and expression levels were further validated in the GEPIA2 database and GSE31312 dataset to be reliable (p < 0.01). The core genes’ main KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichments were Ribosome and Coronavirus disease-COVID-19. High expressions of the 15 core hub genes had prognostic value in DLBCL. The core genes showed significant predictive accuracy in distinguishing DLBCL cases from non-tumor controls, with the area under the curve (AUC) ranging from 0.992 to 1.00. Finally, CIBERSORT analysis on GSE12453 revealed immune cells, including activated memory CD4+ T cells and M0, M1, and M2-macrophages as the infiltrates in the DLBCL microenvironment. Conclusion Our study found differentially co-expressed core hub genes and relevant pathways involved in ribosome and COVID-19 disease that may be potential targets for prognosis and novel therapeutic intervention in DLBCL.
Collapse
Affiliation(s)
- Alice Charwudzi
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ye Meng
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Linhui Hu
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chen Ding
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lianfang Pu
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qian Li
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengling Xu
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhimin Zhai
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shudao Xiong
- Department of Hematology/Hematological Lab, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
6
|
Guillaume B, Jérôme T, Philippe L, Eduardo C, François-Joseph L, Eric B. Aging at evolutionary crossroads: longitudinal gene co-expression network analyses of proximal and ultimate causes of aging in bats. Mol Biol Evol 2021; 39:6400255. [PMID: 34662394 PMCID: PMC8763092 DOI: 10.1093/molbev/msab302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
How, when, and why do organisms, their tissues, and their cells age remain challenging issues, although researchers have identified multiple mechanistic causes of aging, and three major evolutionary theories have been developed to unravel the ultimate causes of organismal aging. A central hypothesis of these theories is that the strength of natural selection decreases with age. However, empirical evidence on when, why, and how organisms age is phylogenetically limited, especially in natural populations. Here, we developed generic comparisons of gene co-expression networks that quantify and dissect the heterogeneity of gene co-expression in conspecific individuals from different age-classes to provide topological evidence about some mechanical and fundamental causes of organismal aging. We applied this approach to investigate the complexity of some proximal and ultimate causes of aging phenotypes in a natural population of the greater mouse-eared bat Myotis myotis, a remarkably long-lived species given its body size and metabolic rate, with available longitudinal blood transcriptomes. M. myotis gene co-expression networks become increasingly fragmented with age, suggesting an erosion of the strength of natural selection and a general dysregulation of gene co-expression in aging bats. However, selective pressures remain sufficiently strong to allow successive emergence of homogeneous age-specific gene co-expression patterns, for at least 7 years. Thus, older individuals from long-lived species appear to sit at an evolutionary crossroad: as they age, they experience both a decrease in the strength of natural selection and a targeted selection for very specific biological processes, further inviting to refine a central hypothesis in evolutionary aging theories.
Collapse
Affiliation(s)
- Bernard Guillaume
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, 75005, France
| | - Teulière Jérôme
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, 75005, France
| | - Lopez Philippe
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, 75005, France
| | - Corel Eduardo
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, 75005, France
| | - Lapointe François-Joseph
- Département de sciences biologiques, Complexe des sciences, 1375 avenue Thérèse-Lavoie-Roux, Université de Montréal, Montréal, Québec), H2V 0B3, Canada (
| | - Bapteste Eric
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, 75005, France
| |
Collapse
|
7
|
Petti M, Verrienti A, Paci P, Farina L. SEaCorAl: Identifying and contrasting the regulation-correlation bias in RNA-Seq paired expression data of patient groups. Comput Biol Med 2021; 135:104567. [PMID: 34174761 DOI: 10.1016/j.compbiomed.2021.104567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 01/11/2023]
Abstract
The Cancer Genome Atlas database offers the possibility of analyzing genome-wide expression RNA-Seq cancer data using paired counts, that is, studies where expression data are collected in pairs of normal and cancer cells, by taking samples from the same individual. Correlation of gene expression profiles is the most common analysis to study co-expression groups, which is used to find biological interpretation of -omics big data. The aim of the paper is threefold: firstly we show for the first time, the presence of a "regulation-correlation bias" in RNA-Seq paired expression data, that is an artifactual link between the expression status (up- or down-regulation) of a gene pair and the sign of the corresponding correlation coefficient. Secondly, we provide a statistical model able to theoretically explain the reasons for the presence of such a bias. Thirdly, we present a bias-removal algorithm, called SEaCorAl, able to effectively reduce bias effects and improve the biological significance of correlation analysis. Validation of the SEaCorAl algorithm is performed by showing a significant increase in the ability to detect biologically meaningful associations of positive correlations and a significant increase of the modularity of the resulting unbiased correlation network.
Collapse
Affiliation(s)
- Manuela Petti
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Italy
| | - Antonella Verrienti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Paola Paci
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Italy
| | - Lorenzo Farina
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Italy.
| |
Collapse
|
8
|
Oh M, Kim K, Sun H. Covariance thresholding to detect differentially co-expressed genes from microarray gene expression data. J Bioinform Comput Biol 2021; 18:2050002. [PMID: 32336254 DOI: 10.1142/s021972002050002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Gene set analysis aims to identify differentially expressed or co-expressed genes within a biological pathway between two experimental conditions, so that it can eventually reveal biological processes and pathways involved in disease development. In the last few decades, various statistical and computational methods have been proposed to improve statistical power of gene set analysis. In recent years, much attention has been paid to differentially co-expressed genes since they can be potentially disease-related genes without significant difference in average expression levels between two conditions. In this paper, we propose a new statistical method to identify differentially co-expressed genes from microarray gene expression data. The proposed method first estimates co-expression levels of paired genes using covariance regularization by thresholding, and then significance of difference in covariance estimation between two conditions is evaluated. We demonstrated that the proposed method is more powerful than the existing main-stream methods to detect co-expressed genes through extensive simulation studies. Also, we applied it to various microarray gene expression datasets related with mutant p53 transcriptional activity, and epithelium and stroma breast cancer.
Collapse
Affiliation(s)
- Mingyu Oh
- Department of Statistics, Pusan National University, Busan, 46241, Korea
| | - Kipoong Kim
- Department of Statistics, Pusan National University, Busan, 46241, Korea
| | - Hokeun Sun
- Department of Statistics, Pusan National University, Busan, 46241, Korea
| |
Collapse
|
9
|
Johnson TS, Xiang S, Dong T, Huang Z, Cheng M, Wang T, Yang K, Ni D, Huang K, Zhang J. Combinatorial analyses reveal cellular composition changes have different impacts on transcriptomic changes of cell type specific genes in Alzheimer's Disease. Sci Rep 2021; 11:353. [PMID: 33432017 PMCID: PMC7801680 DOI: 10.1038/s41598-020-79740-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/09/2020] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) brains are characterized by progressive neuron loss and gliosis. Previous studies of gene expression using bulk tissue samples often fail to consider changes in cell-type composition when comparing AD versus control, which can lead to differences in expression levels that are not due to transcriptional regulation. We mined five large transcriptomic AD datasets for conserved gene co-expression module, then analyzed differential expression and differential co-expression within the modules between AD samples and controls. We performed cell-type deconvolution analysis to determine whether the observed differential expression was due to changes in cell-type proportions in the samples or to transcriptional regulation. Our findings were validated using four additional datasets. We discovered that the increased expression of microglia modules in the AD samples can be explained by increased microglia proportions in the AD samples. In contrast, decreased expression and perturbed co-expression within neuron modules in the AD samples was likely due in part to altered regulation of neuronal pathways. Several transcription factors that are differentially expressed in AD might account for such altered gene regulation. Similarly, changes in gene expression and co-expression within astrocyte modules could be attributed to combined effects of astrogliosis and astrocyte gene activation. Gene expression in the astrocyte modules was also strongly correlated with clinicopathological biomarkers. Through this work, we demonstrated that combinatorial analysis can delineate the origins of transcriptomic changes in bulk tissue data and shed light on key genes and pathways involved in AD.
Collapse
Affiliation(s)
- Travis S Johnson
- Department of Biostatistics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Shunian Xiang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Tianhan Dong
- Department of Pharmacology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Zhi Huang
- Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Cheng
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Kai Yang
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Dong Ni
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen, 518060, China.
| | - Kun Huang
- Department of Medicine, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA.
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
10
|
Savino A, Provero P, Poli V. Differential Co-Expression Analyses Allow the Identification of Critical Signalling Pathways Altered during Tumour Transformation and Progression. Int J Mol Sci 2020; 21:E9461. [PMID: 33322692 PMCID: PMC7764314 DOI: 10.3390/ijms21249461] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 02/02/2023] Open
Abstract
Biological systems respond to perturbations through the rewiring of molecular interactions, organised in gene regulatory networks (GRNs). Among these, the increasingly high availability of transcriptomic data makes gene co-expression networks the most exploited ones. Differential co-expression networks are useful tools to identify changes in response to an external perturbation, such as mutations predisposing to cancer development, and leading to changes in the activity of gene expression regulators or signalling. They can help explain the robustness of cancer cells to perturbations and identify promising candidates for targeted therapy, moreover providing higher specificity with respect to standard co-expression methods. Here, we comprehensively review the literature about the methods developed to assess differential co-expression and their applications to cancer biology. Via the comparison of normal and diseased conditions and of different tumour stages, studies based on these methods led to the definition of pathways involved in gene network reorganisation upon oncogenes' mutations and tumour progression, often converging on immune system signalling. A relevant implementation still lagging behind is the integration of different data types, which would greatly improve network interpretability. Most importantly, performance and predictivity evaluation of the large variety of mathematical models proposed would urgently require experimental validations and systematic comparisons. We believe that future work on differential gene co-expression networks, complemented with additional omics data and experimentally tested, will considerably improve our insights into the biology of tumours.
Collapse
Affiliation(s)
- Aurora Savino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Corso Massimo D’Ázeglio 52, 10126 Turin, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Valeria Poli
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
11
|
Chowdhury HA, Bhattacharyya DK, Kalita JK. (Differential) Co-Expression Analysis of Gene Expression: A Survey of Best Practices. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1154-1173. [PMID: 30668502 DOI: 10.1109/tcbb.2019.2893170] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Analysis of gene expression data is widely used in transcriptomic studies to understand functions of molecules inside a cell and interactions among molecules. Differential co-expression analysis studies diseases and phenotypic variations by finding modules of genes whose co-expression patterns vary across conditions. We review the best practices in gene expression data analysis in terms of analysis of (differential) co-expression, co-expression network, differential networking, and differential connectivity considering both microarray and RNA-seq data along with comparisons. We highlight hurdles in RNA-seq data analysis using methods developed for microarrays. We include discussion of necessary tools for gene expression analysis throughout the paper. In addition, we shed light on scRNA-seq data analysis by including preprocessing and scRNA-seq in co-expression analysis along with useful tools specific to scRNA-seq. To get insights, biological interpretation and functional profiling is included. Finally, we provide guidelines for the analyst, along with research issues and challenges which should be addressed.
Collapse
|
12
|
Liu W, Gan C, Wang W, Liao L, Li C, Xu L, Li E. Identification of lncRNA-associated differential subnetworks in oesophageal squamous cell carcinoma by differential co-expression analysis. J Cell Mol Med 2020; 24:4804-4818. [PMID: 32164040 PMCID: PMC7176870 DOI: 10.1111/jcmm.15159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
Differential expression analysis has led to the identification of important biomarkers in oesophageal squamous cell carcinoma (ESCC). Despite enormous contributions, it has not harnessed the full potential of gene expression data, such as interactions among genes. Differential co-expression analysis has emerged as an effective tool that complements differential expression analysis to provide better insight of dysregulated mechanisms and indicate key driver genes. Here, we analysed the differential co-expression of lncRNAs and protein-coding genes (PCGs) between normal oesophageal tissue and ESCC tissues, and constructed a lncRNA-PCG differential co-expression network (DCN). DCN was characterized as a scale-free, small-world network with modular organization. Focusing on lncRNAs, a total of 107 differential lncRNA-PCG subnetworks were identified from the DCN by integrating both differential expression and differential co-expression. These differential subnetworks provide a valuable source for revealing lncRNA functions and the associated dysfunctional regulatory networks in ESCC. Their consistent discrimination suggests that they may have important roles in ESCC and could serve as robust subnetwork biomarkers. In addition, two tumour suppressor genes (AL121899.1 and ELMO2), identified in the core modules, were validated by functional experiments. The proposed method can be easily used to investigate differential subnetworks of other molecules in other cancers.
Collapse
Affiliation(s)
- Wei Liu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouChina
- Department of MathematicsHeilongjiang Institute of TechnologyHarbinChina
| | - Cai‐Yan Gan
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouChina
| | - Wei Wang
- Department of MathematicsHeilongjiang Institute of TechnologyHarbinChina
| | - Lian‐Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Institute of Oncologic PathologyShantou University Medical CollegeShantouChina
| | - Chun‐Quan Li
- Department of Medical InformaticsHarbin Medical University‐DaqingDaqingChina
| | - Li‐Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Institute of Oncologic PathologyShantou University Medical CollegeShantouChina
| | - En‐Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouChina
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouChina
| |
Collapse
|
13
|
Kakati T, Bhattacharyya DK, Barah P, Kalita JK. Comparison of Methods for Differential Co-expression Analysis for Disease Biomarker Prediction. Comput Biol Med 2019; 113:103380. [PMID: 31415946 DOI: 10.1016/j.compbiomed.2019.103380] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/01/2019] [Accepted: 08/03/2019] [Indexed: 01/23/2023]
Abstract
In the recent past, a number of methods have been developed for analysis of biological data. Among these methods, gene co-expression networks have the ability to mine functionally related genes with similar co-expression patterns, because of which such networks have been most widely used. However, gene co-expression networks cannot identify genes, which undergo condition specific changes in their relationships with other genes. In contrast, differential co-expression analysis enables finding co-expressed genes exhibiting significant changes across disease conditions. In this paper, we present some significant outcomes of a comparative study of four co-expression network module detection techniques, namely, THD-Module Extractor, DiffCoEx, MODA, and WGCNA, which can perform differential co-expression analysis on both gene and miRNA expression data (microarray and RNA-seq) and discuss the applications to Alzheimer's disease and Parkinson's disease research. Our observations reveal that compared to other methods, THD-Module Extractor is the most effective in finding modules with higher functional relevance and biological significance.
Collapse
Affiliation(s)
- Tulika Kakati
- Department of Computer Science and Engineering, Tezpur University, Tezpur, Assam, 784028, India
| | - Dhruba K Bhattacharyya
- Department of Computer Science and Engineering, Tezpur University, Tezpur, Assam, 784028, India.
| | - Pankaj Barah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| | - Jugal K Kalita
- Department of Computer Science, University of Colorado, Colorado Springs, CO, 80918, USA
| |
Collapse
|
14
|
Jardim VC, Santos SDS, Fujita A, Buckeridge MS. BioNetStat: A Tool for Biological Networks Differential Analysis. Front Genet 2019; 10:594. [PMID: 31293621 PMCID: PMC6598498 DOI: 10.3389/fgene.2019.00594] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/05/2019] [Indexed: 01/25/2023] Open
Abstract
The study of interactions among biological components can be carried out by using methods grounded on network theory. Most of these methods focus on the comparison of two biological networks (e.g., control vs. disease). However, biological systems often present more than two biological states (e.g., tumor grades). To compare two or more networks simultaneously, we developed BioNetStat, a Bioconductor package with a user-friendly graphical interface. BioNetStat compares correlation networks based on the probability distribution of a feature of the graph (e.g., centrality measures). The analysis of the structural alterations on the network reveals significant modifications in the system. For example, the analysis of centrality measures provides information about how the relevance of the nodes changes among the biological states. We evaluated the performance of BioNetStat in both, toy models and two case studies. The latter related to gene expression of tumor cells and plant metabolism. Results based on simulated scenarios suggest that the statistical power of BioNetStat is less sensitive to the increase of the number of networks than Gene Set Coexpression Analysis (GSCA). Also, besides being able to identify nodes with modified centralities, BioNetStat identified altered networks associated with signaling pathways that were not identified by other methods.
Collapse
Affiliation(s)
- Vinícius Carvalho Jardim
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
- Department of Botany, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Suzana de Siqueira Santos
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
| | - Andre Fujita
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
15
|
Xiang R, McNally J, Bond J, Tucker D, Cameron M, Donaldson AJ, Austin KL, Rowe S, Jonker A, Pinares-Patino CS, McEwan JC, Vercoe PE, Oddy VH, Dalrymple BP. Across-Experiment Transcriptomics of Sheep Rumen Identifies Expression of Lipid/Oxo-Acid Metabolism and Muscle Cell Junction Genes Associated With Variation in Methane-Related Phenotypes. Front Genet 2018; 9:330. [PMID: 30177952 PMCID: PMC6109778 DOI: 10.3389/fgene.2018.00330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/31/2018] [Indexed: 01/03/2023] Open
Abstract
Ruminants are significant contributors to the livestock generated component of the greenhouse gas, methane (CH4). The CH4 is primarily produced by the rumen microbes. Although the composition of the diet and animal intake amount have the largest effect on CH4 production and yield (CH4 production/dry matter intake, DMI), the host also influences CH4 yield. Shorter rumen feed mean retention time (MRT) is associated with higher dry matter intake and lower CH4 yield, but the molecular mechanism(s) by which the host affects CH4 production remain unclear. We integrated rumen wall transcriptome data and CH4 phenotypes from two independent experiments conducted with sheep in Australia (AUS, n = 62) and New Zealand (NZ, n = 24). The inclusion of the AUS data validated the previously identified clusters and gene sets representing rumen epithelial, metabolic and muscular functions. In addition, the expression of the cell cycle genes as a group was consistently positively correlated with acetate and butyrate concentrations (p < 0.05, based on AUS and NZ data together). The expression of a group of metabolic genes showed positive correlations in both AUS and NZ datasets with CH4 production (p < 0.05) and yield (p < 0.01). These genes encode key enzymes in the ketone body synthesis pathway and included members of the poorly characterized aldo-keto reductase 1C (AKR1C) family. Several AKR1C family genes appear to have ruminant specific evolution patterns, supporting their specialized roles in the ruminants. Combining differential gene expression in the rumen wall muscle of the shortest and longest MRT AUS animals (no data available for the NZ animals) with correlation and network analysis, we identified a set of rumen muscle genes involved in cell junctions as potential regulators of MRT, presumably by influencing contraction rates of the smooth muscle component of the rumen wall. Higher rumen expression of these genes, including SYNPO (synaptopodin, p < 0.01) and NEXN (nexilin, p < 0.05), was associated with lower CH4 yield in both AUS and NZ datasets. Unlike the metabolic genes, the variations in the expression of which may reflect the availability of rumen metabolites, the muscle genes are currently our best candidates for causal genes that influence CH4 yield.
Collapse
Affiliation(s)
- Ruidong Xiang
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, St Lucia, QLD, Australia.,Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia.,Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, Australia
| | - Jody McNally
- F. D. McMaster Laboratory, CSIRO Agriculture & Food, Armidale, NSW, Australia
| | - Jude Bond
- NSW Department of Primary Industries, Extensive Livestock Industries Centre, University of New England, Armidale, NSW, Australia
| | - David Tucker
- NSW Department of Primary Industries, Extensive Livestock Industries Centre, University of New England, Armidale, NSW, Australia
| | - Margaret Cameron
- NSW Department of Primary Industries, Extensive Livestock Industries Centre, University of New England, Armidale, NSW, Australia
| | - Alistair J Donaldson
- NSW Department of Primary Industries, Extensive Livestock Industries Centre, University of New England, Armidale, NSW, Australia
| | - Katie L Austin
- NSW Department of Primary Industries, Extensive Livestock Industries Centre, University of New England, Armidale, NSW, Australia
| | - Suzanne Rowe
- Invermay Agricultural Centre, AgResearch Limited, Mosgiel, New Zealand
| | - Arjan Jonker
- Grasslands Research Centre, AgResearch Limited, Palmerston North, New Zealand
| | - Cesar S Pinares-Patino
- Grasslands Research Centre, AgResearch Limited, Palmerston North, New Zealand.,New Zealand-Peru Dairy Support Project, MINAGRI, Lima, Peru
| | - John C McEwan
- Invermay Agricultural Centre, AgResearch Limited, Mosgiel, New Zealand
| | - Phil E Vercoe
- School of Animal Biology, The University of Western Australia, Crawley, WA, Australia.,Institute of Agriculture, The University of Western Australia, Crawley, WA, Australia
| | - V H Oddy
- NSW Department of Primary Industries, Extensive Livestock Industries Centre, University of New England, Armidale, NSW, Australia
| | - Brian P Dalrymple
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, St Lucia, QLD, Australia.,Institute of Agriculture, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
16
|
Parraga-Alava J, Dorn M, Inostroza-Ponta M. A multi-objective gene clustering algorithm guided by apriori biological knowledge with intensification and diversification strategies. BioData Min 2018; 11:16. [PMID: 30100924 PMCID: PMC6081857 DOI: 10.1186/s13040-018-0178-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/29/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Biologists aim to understand the genetic background of diseases, metabolic disorders or any other genetic condition. Microarrays are one of the main high-throughput technologies for collecting information about the behaviour of genetic information on different conditions. In order to analyse this data, clustering arises as one of the main techniques used, and it aims at finding groups of genes that have some criterion in common, like similar expression profile. However, the problem of finding groups is normally multi dimensional, making necessary to approach the clustering as a multi-objective problem where various cluster validity indexes are simultaneously optimised. They are usually based on criteria like compactness and separation, which may not be sufficient since they can not guarantee the generation of clusters that have both similar expression patterns and biological coherence. METHOD We propose a Multi-Objective Clustering algorithm Guided by a-Priori Biological Knowledge (MOC-GaPBK) to find clusters of genes with high levels of co-expression, biological coherence, and also good compactness and separation. Cluster quality indexes are used to optimise simultaneously gene relationships at expression level and biological functionality. Our proposal also includes intensification and diversification strategies to improve the search process. RESULTS The effectiveness of the proposed algorithm is demonstrated on four publicly available datasets. Comparative studies of the use of different objective functions and other widely used microarray clustering techniques are reported. Statistical, visual and biological significance tests are carried out to show the superiority of the proposed algorithm. CONCLUSIONS Integrating a-priori biological knowledge into a multi-objective approach and using intensification and diversification strategies allow the proposed algorithm to find solutions with higher quality than other microarray clustering techniques available in the literature in terms of co-expression, biological coherence, compactness and separation.
Collapse
Affiliation(s)
- Jorge Parraga-Alava
- Centre for Biotechnology and Bioengineering (CeBiB), Departamento de Ingeniería Informática, Universidad de Santiago de Chile, Av. Ecuador 3659, Santiago, Chile
- Carrera de Computación, Escuela Superior Politécnica Agropecuaria de Manabí Manuel Félix López, Campus Politécnico Sitio El Limón, Calceta, Ecuador
| | - Marcio Dorn
- Instituto de Informatica, Universidade Federal do Rio Grande do Sul, Av. Bento Gonçalves 9500, Porto Alegre, 91501-970 Brasil
| | - Mario Inostroza-Ponta
- Centre for Biotechnology and Bioengineering (CeBiB), Departamento de Ingeniería Informática, Universidad de Santiago de Chile, Av. Ecuador 3659, Santiago, Chile
| |
Collapse
|
17
|
Igolkina AA, Armoskus C, Newman JRB, Evgrafov OV, McIntyre LM, Nuzhdin SV, Samsonova MG. Analysis of Gene Expression Variance in Schizophrenia Using Structural Equation Modeling. Front Mol Neurosci 2018; 11:192. [PMID: 29942251 PMCID: PMC6004421 DOI: 10.3389/fnmol.2018.00192] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/15/2018] [Indexed: 01/02/2023] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder of unknown etiology. There is evidence suggesting that aberrations in neurodevelopment are a significant attribute of schizophrenia pathogenesis and progression. To identify biologically relevant molecular abnormalities affecting neurodevelopment in SCZ we used cultured neural progenitor cells derived from olfactory neuroepithelium (CNON cells). Here, we tested the hypothesis that variance in gene expression differs between individuals from SCZ and control groups. In CNON cells, variance in gene expression was significantly higher in SCZ samples in comparison with control samples. Variance in gene expression was enriched in five molecular pathways: serine biosynthesis, PI3K-Akt, MAPK, neurotrophin and focal adhesion. More than 14% of variance in disease status was explained within the logistic regression model (C-value = 0.70) by predictors accounting for gene expression in 69 genes from these five pathways. Structural equation modeling (SEM) was applied to explore how the structure of these five pathways was altered between SCZ patients and controls. Four out of five pathways showed differences in the estimated relationships among genes: between KRAS and NF1, and KRAS and SOS1 in the MAPK pathway; between PSPH and SHMT2 in serine biosynthesis; between AKT3 and TSC2 in the PI3K-Akt signaling pathway; and between CRK and RAPGEF1 in the focal adhesion pathway. Our analysis provides evidence that variance in gene expression is an important characteristic of SCZ, and SEM is a promising method for uncovering altered relationships between specific genes thus suggesting affected gene regulation associated with the disease. We identified altered gene-gene interactions in pathways enriched for genes with increased variance in expression in SCZ. These pathways and loci were previously implicated in SCZ, providing further support for the hypothesis that gene expression variance plays important role in the etiology of SCZ.
Collapse
Affiliation(s)
- Anna A Igolkina
- Institute of Applied Mathematics and Mechanics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Chris Armoskus
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jeremy R B Newman
- Department of Molecular Genetics & Microbiology, Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Oleg V Evgrafov
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Lauren M McIntyre
- Department of Molecular Genetics & Microbiology, Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Sergey V Nuzhdin
- Institute of Applied Mathematics and Mechanics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia.,Molecular and Computation Biology, University of Southern California, Los Angeles, CA, United States
| | - Maria G Samsonova
- Institute of Applied Mathematics and Mechanics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
18
|
Patkar S, Magen A, Sharan R, Hannenhalli S. A network diffusion approach to inferring sample-specific function reveals functional changes associated with breast cancer. PLoS Comput Biol 2017; 13:e1005793. [PMID: 29190299 PMCID: PMC5708603 DOI: 10.1371/journal.pcbi.1005793] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/27/2017] [Indexed: 11/18/2022] Open
Abstract
Guilt-by-association codifies the empirical observation that a gene's function is informed by its neighborhood in a biological network. This would imply that when a gene's network context is altered, for instance in disease condition, so could be the gene's function. Although context-specific changes in biological networks have been explored, the potential changes they may induce on the functional roles of genes are yet to be characterized. Here we analyze, for the first time, the network-induced potential functional changes in breast cancer. Using transcriptomic samples for 1047 breast tumors and 110 healthy breast tissues from TCGA, we derive sample-specific protein interaction networks and assign sample-specific functions to genes via a diffusion strategy. Testing for significant changes in the inferred functions between normal and cancer samples, we find several functions to have significantly gained or lost genes in cancer, not due to differential expression of genes known to perform the function, but rather due to changes in the network topology. Our predicted functional changes are supported by mutational and copy number profiles in breast cancers. Our diffusion-based functional assignment provides a novel characterization of a tumor that is complementary to the standard approach based on functional annotation alone. Importantly, this characterization is effective in predicting patient survival, as well as in predicting several known histopathological subtypes of breast cancer.
Collapse
Affiliation(s)
- Sushant Patkar
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Assaf Magen
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Sridhar Hannenhalli
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| |
Collapse
|
19
|
Tasaki S, Suzuki K, Nishikawa A, Kassai Y, Takiguchi M, Kurisu R, Okuzono Y, Miyazaki T, Takeshita M, Yoshimoto K, Yasuoka H, Yamaoka K, Ikeura K, Tsunoda K, Morita R, Yoshimura A, Toyoshiba H, Takeuchi T. Multiomic disease signatures converge to cytotoxic CD8 T cells in primary Sjögren's syndrome. Ann Rheum Dis 2017; 76:1458-1466. [PMID: 28522454 PMCID: PMC5738597 DOI: 10.1136/annrheumdis-2016-210788] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/28/2017] [Accepted: 04/09/2017] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Multiomics study was conducted to elucidate the crucial molecular mechanisms of primary Sjögren's syndrome (SS) pathology. METHODS We generated multiple data set from well-defined patients with SS, which includes whole-blood transcriptomes, serum proteomes and peripheral immunophenotyping. Based on our newly generated data, we performed an extensive bioinformatic investigation. RESULTS Our integrative analysis identified SS gene signatures (SGS) dysregulated in widespread omics layers, including epigenomes, mRNAs and proteins. SGS predominantly involved the interferon signature and ADAMs substrates. Besides, SGS was significantly overlapped with SS-causing genes indicated by a genome-wide association study and expression trait loci analyses. Combining the molecular signatures with immunophenotypic profiles revealed that cytotoxic CD8 -T cells- were associated with SGS. Further, we observed the activation of SGS in cytotoxic CD8 T cells isolated from patients with SS. CONCLUSIONS Our multiomics investigation identified gene signatures deeply associated with SS pathology and showed the involvement of cytotoxic CD8 T cells. These integrative relations across multiple layers will facilitate our understanding of SS at the system level.
Collapse
Affiliation(s)
- Shinya Tasaki
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
- Rush University Medical Center, Rush Alzheimer’s Disease Center, Chicago, Illinois, USA
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Ayumi Nishikawa
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yoshiaki Kassai
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Maiko Takiguchi
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Rina Kurisu
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Yuumi Okuzono
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Takahiro Miyazaki
- Immunology Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
- Nektar Therapeutics, San Francisco, California, USA
| | - Masaru Takeshita
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Keiko Yoshimoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hidekata Yasuoka
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kunihiro Yamaoka
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kazuhiro Ikeura
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kazuyuki Tsunoda
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroyoshi Toyoshiba
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa City, Kanagawa, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|