351
|
Applications of comparative evolution to human disease genetics. Curr Opin Genet Dev 2015; 35:16-24. [PMID: 26338499 DOI: 10.1016/j.gde.2015.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 12/15/2022]
Abstract
Direct comparison of human diseases with model phenotypes allows exploration of key areas of human biology which are often inaccessible for practical or ethical reasons. We review recent developments in comparative evolutionary approaches for finding models for genetic disease, including high-throughput generation of gene/phenotype relationship data, the linking of orthologous genes and phenotypes across species, and statistical methods for linking human diseases to model phenotypes.
Collapse
|
352
|
|
353
|
Wu X, Chen H, Xu H. The genomic landscape of human immune-mediated diseases. J Hum Genet 2015; 60:675-81. [PMID: 26290150 DOI: 10.1038/jhg.2015.99] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/06/2015] [Accepted: 07/16/2015] [Indexed: 02/06/2023]
Abstract
As the methodology of genetic detection has developed rapidly in recent years, through techniques such as genome-wide association studies (GWAS) and the secondary generation of sequencing, we are able to view the genomic landscape more clearly. It is well known that genes have a vital role in the pathogenesis of immune-mediated diseases (IMDs), which could provide important insight into new clinical therapeutic targets. Here, we review the genomic landscape of IMDs and analyse overlapping loci between diseases. There may be a need for more epigenetics studies to aid in the understanding of the transition from genotype to phenotype.
Collapse
Affiliation(s)
- Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Haiyan Chen
- Department of Rheumatology, Zhongda Hospital, Southeast University, Shanghai, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
354
|
Kim J, Kwon EY, Park S, Kim JR, Choi SW, Choi MS, Kim SJ. Integrative systems analysis of diet-induced obesity identified a critical transition in the transcriptomes of the murine liver and epididymal white adipose tissue. Int J Obes (Lond) 2015; 40:338-45. [PMID: 26268884 DOI: 10.1038/ijo.2015.147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/24/2015] [Accepted: 07/26/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND It is well known that high-fat diet (HFD) can cause immune system-related pathological alterations after a significant body weight gain. The mechanisms of the delayed pathological alterations during the development of diet-induced obesity (DIO) are not fully understood. METHODS To elucidate the mechanisms underlying DIO development, we analyzed time-course microarray data obtained from a previous study. First, differentially expressed genes (DEGs) were identified at each time point by comparing the hepatic transcriptome of mice fed HFD with that of mice fed normal diet. Next, we clustered the union of DEGs and identified annotations related to each cluster. Finally, we constructed an 'integrated obesity-associated gene regulatory network (GRN) in murine liver'. We analyzed the epididymal white adipose tissue (eWAT) transcriptome usig the same procedure. RESULTS Based on time-course microarray data, we found that the genes associated with immune responses were upregulated with an oscillating expression pattern between weeks 2 and 8, relatively downregulated between weeks 12 and 16, and eventually upregulated after week 20 in the liver of the mice fed HFD. The genes associated with immune responses were also upregulated at late stage, in the eWAT of the mice fed HFD. These results suggested that a critical transition occurred in the immune system-related transcriptomes of the liver and eWAT around week 16 of the DIO development, and this may be associated with the delayed pathological alterations. The GRN analysis suggested that Maff may be a key transcription factor for the immune system-related critical transition thatoccurred at week 16. We found that transcription factors associated with immune responses were centrally located in the integrated obesity-associated GRN in the liver. CONCLUSIONS In this study, systems analysis identified regulatory network modules underlying the delayed immune system-related pathological changes during the development of DIO and could suggest possible therapeutic targets.
Collapse
Affiliation(s)
- J Kim
- CHA Cancer Institute, CHA University, Seongnam City, Kyunggi-do, Republic of Korea
| | - E-Y Kwon
- Center for Food and Nutritional Genomics Research, Department of Food Science and Nutrition, Kyungpook National University, Daegu, Republic of Korea
| | - S Park
- CHA Cancer Institute, CHA University, Seongnam City, Kyunggi-do, Republic of Korea
| | - J-R Kim
- Department of Mathematics, University of Seoul, Seoul, Republic of Korea
| | - S-W Choi
- CHA Cancer Institute, CHA University, Seongnam City, Kyunggi-do, Republic of Korea.,Chaum Life Center, CHA University, School of Medicine, Seoul, Republic of Korea
| | - M-S Choi
- Center for Food and Nutritional Genomics Research, Department of Food Science and Nutrition, Kyungpook National University, Daegu, Republic of Korea
| | - S-J Kim
- CHA Cancer Institute, CHA University, Seongnam City, Kyunggi-do, Republic of Korea.,Department of Biomedical Sciences, College of Life Sciences, CHA University, Seongnam City, Kyunggi-do, Republic of Korea
| |
Collapse
|
355
|
Zhu F, Panwar B, Guan Y. Algorithms for modeling global and context-specific functional relationship networks. Brief Bioinform 2015; 17:686-95. [PMID: 26254431 DOI: 10.1093/bib/bbv065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Indexed: 02/07/2023] Open
Abstract
Functional genomics has enormous potential to facilitate our understanding of normal and disease-specific physiology. In the past decade, intensive research efforts have been focused on modeling functional relationship networks, which summarize the probability of gene co-functionality relationships. Such modeling can be based on either expression data only or heterogeneous data integration. Numerous methods have been deployed to infer the functional relationship networks, while most of them target the global (non-context-specific) functional relationship networks. However, it is expected that functional relationships consistently reprogram under different tissues or biological processes. Thus, advanced methods have been developed targeting tissue-specific or developmental stage-specific networks. This article brings together the state-of-the-art functional relationship network modeling methods, emphasizes the need for heterogeneous genomic data integration and context-specific network modeling and outlines future directions for functional relationship networks.
Collapse
|
356
|
Krogan NJ, Lippman S, Agard DA, Ashworth A, Ideker T. The cancer cell map initiative: defining the hallmark networks of cancer. Mol Cell 2015; 58:690-8. [PMID: 26000852 PMCID: PMC5359018 DOI: 10.1016/j.molcel.2015.05.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Progress in DNA sequencing has revealed the startling complexity of cancer genomes, which typically carry thousands of somatic mutations. However, it remains unclear which are the key driver mutations or dependencies in a given cancer and how these influence pathogenesis and response to therapy. Although tumors of similar types and clinical outcomes can have patterns of mutations that are strikingly different, it is becoming apparent that these mutations recurrently hijack the same hallmark molecular pathways and networks. For this reason, it is likely that successful interpretation of cancer genomes will require comprehensive knowledge of the molecular networks under selective pressure in oncogenesis. Here we announce the creation of a new effort, The Cancer Cell Map Initiative (CCMI), aimed at systematically detailing these complex interactions among cancer genes and how they differ between diseased and healthy states. We discuss recent progress that enables creation of these cancer cell maps across a range of tumor types and how they can be used to target networks disrupted in individual patients, significantly accelerating the development of precision medicine.
Collapse
Affiliation(s)
- Nevan J Krogan
- California Institute for Quantitative Biosciences (QB3), University of California, San Francisco, San Francisco, CA 94143, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA; J. David Gladstone Institutes, San Francisco, CA 94143, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Scott Lippman
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA; Moores Cancer Center, University of California, San Diego, San Diego, CA 92093, USA
| | - David A Agard
- California Institute for Quantitative Biosciences (QB3), University of California, San Francisco, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 92093, USA
| | - Alan Ashworth
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 92093, USA
| | - Trey Ideker
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA; Moores Cancer Center, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
357
|
Park S, Lehner B. Cancer type-dependent genetic interactions between cancer driver alterations indicate plasticity of epistasis across cell types. Mol Syst Biol 2015; 11:824. [PMID: 26227665 PMCID: PMC4547852 DOI: 10.15252/msb.20156102] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancers, like many diseases, are normally caused by combinations of genetic alterations rather than by changes affecting single genes. It is well established that the genetic alterations that drive cancer often interact epistatically, having greater or weaker consequences in combination than expected from their individual effects. In a stringent statistical analysis of data from > 3,000 tumors, we find that the co-occurrence and mutual exclusivity relationships between cancer driver alterations change quite extensively in different types of cancer. This cannot be accounted for by variation in tumor heterogeneity or unrecognized cancer subtypes. Rather, it suggests that how genomic alterations interact cooperatively or partially redundantly to driver cancer changes in different types of cancers. This re-wiring of epistasis across cell types is likely to be a basic feature of genetic architecture, with important implications for understanding the evolution of multicellularity and human genetic diseases. In addition, if this plasticity of epistasis across cell types is also true for synthetic lethal interactions, a synthetic lethal strategy to kill cancer cells may frequently work in one type of cancer but prove ineffective in another.
Collapse
Affiliation(s)
- Solip Park
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra, Barcelona, Spain
| | - Ben Lehner
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), Barcelona, Spain Universitat Pompeu Fabra, Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
358
|
Ricchiuto P, Iwata H, Yabusaki K, Yamada I, Pieper B, Sharma A, Aikawa M, Singh SA. mIMT-visHTS: A novel method for multiplexing isobaric mass tagged datasets with an accompanying visualization high throughput screening tool for protein profiling. J Proteomics 2015; 128:132-40. [PMID: 26232111 DOI: 10.1016/j.jprot.2015.07.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 06/26/2015] [Accepted: 07/20/2015] [Indexed: 12/12/2022]
Abstract
Isobaric mass tagging (IMT) methods enable the analysis of thousands of proteins simultaneously. We used tandem mass tagging reagents (TMT™) to monitor the relative changes in the proteome of the mouse macrophage cell line RAW264.7 at the same six time points after no stimulation (baseline phenotype), stimulation with interferon gamma (pro-inflammatory phenotype) or stimulation with interleukin-4 (anti-inflammatory phenotype). The combined TMT datasets yielded nearly 12,000 protein profiles for comparison. To facilitate this large analysis, we developed a novel method that combines or multiplexes the separate IMT (mIMT) datasets into a single super dataset for subsequent model-based clustering and co-regulation analysis. Specially designed visual High Throughput Screening (visHTS) software screened co-regulated proteins. visHTS generates an interactive and visually intuitive color-coded bullseye plot that enables users to browse the cluster outputs and identify co-regulated proteins.
Collapse
Affiliation(s)
- Piero Ricchiuto
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katsumi Yabusaki
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Iwao Yamada
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Brett Pieper
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Amitabh Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
359
|
Kim S, Sael L, Yu H. A mutation profile for top-k patient search exploiting Gene-Ontology and orthogonal non-negative matrix factorization. Bioinformatics 2015. [PMID: 26209432 PMCID: PMC4672174 DOI: 10.1093/bioinformatics/btv409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
MOTIVATION As the quantity of genomic mutation data increases, the likelihood of finding patients with similar genomic profiles, for various disease inferences, increases. However, so does the difficulty in identifying them. Similarity search based on patient mutation profiles can solve various translational bioinformatics tasks, including prognostics and treatment efficacy predictions for better clinical decision making through large volume of data. However, this is a challenging problem due to heterogeneous and sparse characteristics of the mutation data as well as their high dimensionality. RESULTS To solve this problem we introduce a compact representation and search strategy based on Gene-Ontology and orthogonal non-negative matrix factorization. Statistical significance between the identified cancer subtypes and their clinical features are computed for validation; results show that our method can identify and characterize clinically meaningful tumor subtypes comparable or better in most datasets than the recently introduced Network-Based Stratification method while enabling real-time search. To the best of our knowledge, this is the first attempt to simultaneously characterize and represent somatic mutational data for efficient search purposes. AVAILABILITY The implementations are available at: https://sites.google.com/site/postechdm/research/implementation/orgos. CONTACT sael@cs.stonybrook.edu or hwanjoyu@postech.ac.kr SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sungchul Kim
- Department of Computer Science and Engineering, POSTECH, Pohang, South Korea and
| | - Lee Sael
- Department of Computer Science, Stony Brook University, Stony Brook, USA
| | - Hwanjo Yu
- Department of Computer Science and Engineering, POSTECH, Pohang, South Korea and
| |
Collapse
|
360
|
Paredes-Sánchez FA, Sifuentes-Rincón AM, Segura Cabrera A, García Pérez CA, Parra Bracamonte GM, Ambriz Morales P. Associations of SNPs located at candidate genes to bovine growth traits, prioritized with an interaction networks construction approach. BMC Genet 2015. [PMID: 26198337 PMCID: PMC4511253 DOI: 10.1186/s12863-015-0247-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background For most domestic animal species, including bovines, it is difficult to identify causative genetic variants involved in economically relevant traits. The candidate gene approach is efficient because it investigates genes that are expected to be associated with the expression of a trait and defines whether the genetic variation present in a population is associated with phenotypic diversity. A potential limitation of this approach is the identification of candidates. This study used a bioinformatics approach to identify candidate genes via a search guided by a functional interaction network. Results A functional interaction network tool, BosNet, was constructed for Bos taurus. Predictions for candidate genes were performed using the guilt-by-association principle in BosNet. Association analyses identified five novel markers within BosNet-prioritized genes that had significant effects on different growth traits in Charolais and Brahman cattle. Conclusions BosNet is an excellent tool for the identification of single nucleotide polymorphisms that are potentially associated with complex traits.
Collapse
Affiliation(s)
- Francisco Alejandro Paredes-Sánchez
- Laboratorio de Biotecnología Animal, Centro de Biotecnología Genómica. IPN, Boulevard del Maestro esq. Elías Piña, Col. Narciso Mendoza, Cd. Reynosa, Tam, C.P. 88710, Mexico.
| | - Ana María Sifuentes-Rincón
- Laboratorio de Biotecnología Animal, Centro de Biotecnología Genómica. IPN, Boulevard del Maestro esq. Elías Piña, Col. Narciso Mendoza, Cd. Reynosa, Tam, C.P. 88710, Mexico.
| | - Aldo Segura Cabrera
- Red de Estudios Moleculares Avanzados, Instituto de Ecología, A.C., Xalapa, Mexico.
| | - Carlos Armando García Pérez
- Laboratorio de Bioinformática, Centro de Biotecnología Genómica. IPN, Boulevard del Maestro esq. Elías Piña, Col. Narciso Mendoza, Cd. Reynosa, Tam, C.P. 88710, Mexico.
| | - Gaspar Manuel Parra Bracamonte
- Laboratorio de Biotecnología Animal, Centro de Biotecnología Genómica. IPN, Boulevard del Maestro esq. Elías Piña, Col. Narciso Mendoza, Cd. Reynosa, Tam, C.P. 88710, Mexico.
| | - Pascuala Ambriz Morales
- Laboratorio de Biotecnología Animal, Centro de Biotecnología Genómica. IPN, Boulevard del Maestro esq. Elías Piña, Col. Narciso Mendoza, Cd. Reynosa, Tam, C.P. 88710, Mexico.
| |
Collapse
|
361
|
|
362
|
Lee CH, Koyejo O, Ghosh J. Identifying candidate disease genes using a trace norm constrained bipartite raking model. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2013:3459-62. [PMID: 24110473 DOI: 10.1109/embc.2013.6610286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Computational prediction of genes that play roles in human diseases remains an important but challenging task. In this work, we formulate candidate gene prediction as a bipartite ranking problem combining a task-wise ordered observation model with a latent multitask regression function using the matrix-variate Gaussian process (MV-GP). We then use a trace-norm constrained variational inference approach to obtain the bipartite ranking model variables and the parameters of the underlying multitask regression model. We use this model to predict candidate genes from two gene-disease association data sets and show that our model outperforms current state-of-the-art methods. Finally, we demonstrate the practical utility of our method by successfully recovering well characterized gene-disease associations hidden in our training data.
Collapse
|
363
|
Shim JE, Hwang S, Lee I. Pathway-Dependent Effectiveness of Network Algorithms for Gene Prioritization. PLoS One 2015; 10:e0130589. [PMID: 26091506 PMCID: PMC4474432 DOI: 10.1371/journal.pone.0130589] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 05/22/2015] [Indexed: 01/18/2023] Open
Abstract
A network-based approach has proven useful for the identification of novel genes associated with complex phenotypes, including human diseases. Because network-based gene prioritization algorithms are based on propagating information of known phenotype-associated genes through networks, the pathway structure of each phenotype might significantly affect the effectiveness of algorithms. We systematically compared two popular network algorithms with distinct mechanisms – direct neighborhood which propagates information to only direct network neighbors, and network diffusion which diffuses information throughout the entire network – in prioritization of genes for worm and human phenotypes. Previous studies reported that network diffusion generally outperforms direct neighborhood for human diseases. Although prioritization power is generally measured for all ranked genes, only the top candidates are significant for subsequent functional analysis. We found that high prioritizing power of a network algorithm for all genes cannot guarantee successful prioritization of top ranked candidates for a given phenotype. Indeed, the majority of the phenotypes that were more efficiently prioritized by network diffusion showed higher prioritizing power for top candidates by direct neighborhood. We also found that connectivity among pathway genes for each phenotype largely determines which network algorithm is more effective, suggesting that the network algorithm used for each phenotype should be chosen with consideration of pathway gene connectivity.
Collapse
Affiliation(s)
- Jung Eun Shim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Sohyun Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
364
|
TRRUST: a reference database of human transcriptional regulatory interactions. Sci Rep 2015; 5:11432. [PMID: 26066708 PMCID: PMC4464350 DOI: 10.1038/srep11432] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/22/2015] [Indexed: 11/09/2022] Open
Abstract
The reconstruction of transcriptional regulatory networks (TRNs) is a long-standing challenge in human genetics. Numerous computational methods have been developed to infer regulatory interactions between human transcriptional factors (TFs) and target genes from high-throughput data, and their performance evaluation requires gold-standard interactions. Here we present a database of literature-curated human TF-target interactions, TRRUST (transcriptional regulatory relationships unravelled by sentence-based text-mining, http://www.grnpedia.org/trrust), which currently contains 8,015 interactions between 748 TF genes and 1,975 non-TF genes. A sentence-based text-mining approach was employed for efficient manual curation of regulatory interactions from approximately 20 million Medline abstracts. To the best of our knowledge, TRRUST is the largest publicly available database of literature-curated human TF-target interactions to date. TRRUST also has several useful features: i) information about the mode-of-regulation; ii) tests for target modularity of a query TF; iii) tests for TF cooperativity of a query target; iv) inferences about cooperating TFs of a query TF; and v) prioritizing associated pathways and diseases with a query TF. We observed high enrichment of TF-target pairs in TRRUST for top-scored interactions inferred from high-throughput data, which suggests that TRRUST provides a reliable benchmark for the computational reconstruction of human TRNs.
Collapse
|
365
|
Zhong X, Yang H, Zhao S, Shyr Y, Li B. Network-based stratification analysis of 13 major cancer types using mutations in panels of cancer genes. BMC Genomics 2015; 16 Suppl 7:S7. [PMID: 26099277 PMCID: PMC4474538 DOI: 10.1186/1471-2164-16-s7-s7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Cancers are complex diseases with heterogeneous genetic causes and clinical outcomes. It is critical to classify patients into subtypes and associate the subtypes with clinical outcomes for better prognosis and treatment. Large-scale studies have comprehensively identified somatic mutations across multiple tumor types, providing rich datasets for classifying patients based on genomic mutations. One challenge associated with this task is that mutations are rarely shared across patients. Network-based stratification (NBS) approaches have been proposed to overcome this challenge and used to classify tumors based on exome-level mutations. In routine research and clinical applications, however, usually only a small panel of pre-selected genes is screened for mutations. It is unknown whether such small panels are effective in classifying patients into clinically meaningful subtypes. Results In this study, we applied NBS to 13 major cancer types with exome-level mutation data and compared the classification based on the full exome data with those focusing only on small sets of genes. Specifically, we investigated three panels, FoundationOne (240 genes), PanCan (127 genes) and TruSeq (48 genes). We showed that small panels not only are effective in clustering tumors but also often outperform full exome data for most cancer types. We further associated subtypes with clinical data and identified 5 tumor types (CRC-Colorectal carcinoma, HNSC-Head and neck squamous cell carcinoma, KIRC-Kidney renal clear cell carcinoma, LUAD-Lung adenocarcinoma and UCEC-Uterine corpus endometrial carcinoma) whose subtypes are significantly associated with overall survival, all based on small panels. Conclusion Our analyses indicate that effective patient subtyping can be carried out using mutations detected in smaller gene panels, probably due to the enrichment of clinically important genes in such panels.
Collapse
|
366
|
|
367
|
Zhang L, Li H, Hu X, Benedek DM, Fullerton CS, Forsten RD, Naifeh JA, Li X, Wu H, Benevides KN, Le T, Smerin S, Russell DW, Ursano RJ. Mitochondria-focused gene expression profile reveals common pathways and CPT1B dysregulation in both rodent stress model and human subjects with PTSD. Transl Psychiatry 2015; 5:e580. [PMID: 26080315 PMCID: PMC4490278 DOI: 10.1038/tp.2015.65] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/09/2015] [Accepted: 03/24/2015] [Indexed: 12/30/2022] Open
Abstract
Posttraumatic stress disorder (PTSD), a trauma-related mental disorder, is associated with mitochondrial dysfunction in the brain. However, the biologic approach to identifying the mitochondria-focused genes underlying the pathogenesis of PTSD is still in its infancy. Previous research, using a human mitochondria-focused cDNA microarray (hMitChip3) found dysregulated mitochondria-focused genes present in postmortem brains of PTSD patients, indicating that those genes might be PTSD-related biomarkers. To further test this idea, this research examines profiles of mitochondria-focused gene expression in the stressed-rodent model (inescapable tail shock in rats), which shows characteristics of PTSD-like behaviors and also in the blood of subjects with PTSD. This study found that 34 mitochondria-focused genes being upregulated in stressed-rat amygdala. Ten common pathways, including fatty acid metabolism and peroxisome proliferator-activated receptors (PPAR) pathways were dysregulated in the amygdala of the stressed rats. Carnitine palmitoyltransferase 1B (CPT1B), an enzyme in the fatty acid metabolism and PPAR pathways, was significantly over-expressed in the amygdala (P < 0.007) and in the blood (P < 0.01) of stressed rats compared with non-stressed controls. In human subjects with (n = 28) or without PTSD (n = 31), significant over-expression of CPT1B in PTSD was also observed in the two common dysregulated pathways: fatty acid metabolism (P = 0.0027, false discovery rate (FDR) = 0.043) and PPAR (P = 0.006, FDR = 0.08). Quantitative real-time polymerase chain reaction validated the microarray findings and the CPT1B result. These findings indicate that blood can be used as a specimen in the search for PTSD biomarkers in fatty acid metabolism and PPAR pathways, and, in addition, that CPT1B may contribute to the pathology of PTSD.
Collapse
Affiliation(s)
- L Zhang
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA,Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA. E-mail:
| | - H Li
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - X Hu
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - D M Benedek
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - C S Fullerton
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - R D Forsten
- U.S. Army Pacific Command, Hawaiian Islands, HI, USA
| | - J A Naifeh
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - X Li
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - H Wu
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - K N Benevides
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - T Le
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - S Smerin
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - D W Russell
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - R J Ursano
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
368
|
Kachroo AH, Laurent JM, Yellman CM, Meyer AG, Wilke CO, Marcotte EM. Evolution. Systematic humanization of yeast genes reveals conserved functions and genetic modularity. Science 2015; 348:921-5. [PMID: 25999509 PMCID: PMC4718922 DOI: 10.1126/science.aaa0769] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
To determine whether genes retain ancestral functions over a billion years of evolution and to identify principles of deep evolutionary divergence, we replaced 414 essential yeast genes with their human orthologs, assaying for complementation of lethal growth defects upon loss of the yeast genes. Nearly half (47%) of the yeast genes could be successfully humanized. Sequence similarity and expression only partly predicted replaceability. Instead, replaceability depended strongly on gene modules: Genes in the same process tended to be similarly replaceable (e.g., sterol biosynthesis) or not (e.g., DNA replication initiation). Simulations confirmed that selection for specific function can maintain replaceability despite extensive sequence divergence. Critical ancestral functions of many essential genes are thus retained in a pathway-specific manner, resilient to drift in sequences, splicing, and protein interfaces.
Collapse
Affiliation(s)
- Aashiq H Kachroo
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Jon M Laurent
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Christopher M Yellman
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Austin G Meyer
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA. Center for Computational Biology and Bioinformatics, University of Texas at Austin, Austin, TX 78712, USA
| | - Claus O Wilke
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA. Center for Computational Biology and Bioinformatics, University of Texas at Austin, Austin, TX 78712, USA. Department of Integrative Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Edward M Marcotte
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA. Center for Computational Biology and Bioinformatics, University of Texas at Austin, Austin, TX 78712, USA. Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
369
|
Genetic similarity between cancers and comorbid Mendelian diseases identifies candidate driver genes. Nat Commun 2015; 6:7033. [PMID: 25926297 PMCID: PMC4416231 DOI: 10.1038/ncomms8033] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/26/2015] [Indexed: 12/21/2022] Open
Abstract
Despite large-scale cancer genomics studies, key somatic mutations driving cancer, and their functional roles, remain elusive. Here we propose that analysis of comorbidities of Mendelian diseases with cancers provides a novel, systematic way to discover new cancer genes. If germline genetic variation in Mendelian loci predisposes bearers to common cancers, the same loci may harbor cancer-associated somatic variation. Compilations of clinical records spanning over 100 million patients provide an unprecedented opportunity to assess clinical associations between Mendelian diseases and cancers. We systematically compare these comorbidities against recurrent somatic mutations from more than five thousand patients across many cancers. Using multiple measures of genetic similarity, we show that a Mendelian disease and comorbid cancer indeed have genetic alterations of significant functional similarity. This result provides a basis to identify candidate drivers in cancers including melanoma and glioblastoma. Some Mendelian diseases demonstrate “pan-cancer” comorbidity and shared genetics across cancers.
Collapse
|
370
|
Magi A, D'Aurizio R, Palombo F, Cifola I, Tattini L, Semeraro R, Pippucci T, Giusti B, Romeo G, Abbate R, Gensini GF. Characterization and identification of hidden rare variants in the human genome. BMC Genomics 2015; 16:340. [PMID: 25903059 PMCID: PMC4416239 DOI: 10.1186/s12864-015-1481-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 03/23/2015] [Indexed: 12/11/2022] Open
Abstract
Background By examining the genotype calls generated by the 1000 Genomes Project we discovered that the human reference genome GRCh37 contains almost 20,000 loci in which the reference allele has never been observed in healthy individuals and around 70,000 loci in which it has been observed only in the heterozygous state. Results We show that a large fraction of this rare reference allele (RRA) loci belongs to coding, functional and regulatory elements of the genome and could be linked to rare Mendelian disorders as well as cancer. We also demonstrate that classical germline and somatic variant calling tools are not capable to recognize the rare allele when present in these loci. To overcome such limitations, we developed a novel tool, named RAREVATOR, that is able to identify and call the rare allele in these genomic positions. By using a small cancer dataset we compared our tool with two state-of-the-art callers and we found that RAREVATOR identified more than 1,500 germline and 22 somatic RRA variants missed by the two methods and which belong to significantly mutated pathways. Conclusions These results show that, to date, the investigation of around 100,000 loci of the human genome has been missed by re-sequencing experiments based on the GRCh37 assembly and that our tool can fill the gap left by other methods. Moreover, the investigation of the latest version of the human reference genome, GRCh38, showed that although the GRC corrected almost all insertions and a small part of SNVs and deletions, a large number of functionally relevant RRAs still remain unchanged. For this reason, also future resequencing experiments, based on GRCh38, will benefit from RAREVATOR analysis results. RAREVATOR is freely available at http://sourceforge.net/projects/rarevator. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1481-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Romina D'Aurizio
- Laboratory of Integrative Systems Medicine (LISM), Institute of Informatics and Telematics and Institute of Clinical Physiology, National Research Council, Pisa, Italy.
| | - Flavia Palombo
- Medical Genetics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| | - Ingrid Cifola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.
| | - Lorenzo Tattini
- Department of Neuroscience, Pharmacology and Child Health, University of Florence, Florence, Italy.
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Tommaso Pippucci
- Medical Genetics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Giovanni Romeo
- Medical Genetics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| | - Rosanna Abbate
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Gian Franco Gensini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
371
|
The clustering of functionally related genes contributes to CNV-mediated disease. Genome Res 2015; 25:802-13. [PMID: 25887030 PMCID: PMC4448677 DOI: 10.1101/gr.184325.114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 04/13/2015] [Indexed: 12/20/2022]
Abstract
Clusters of functionally related genes can be disrupted by a single copy number variant (CNV). We demonstrate that the simultaneous disruption of multiple functionally related genes is a frequent and significant characteristic of de novo CNVs in patients with developmental disorders (P = 1 × 10−3). Using three different functional networks, we identified unexpectedly large numbers of functionally related genes within de novo CNVs from two large independent cohorts of individuals with developmental disorders. The presence of multiple functionally related genes was a significant predictor of a CNV's pathogenicity when compared to CNVs from apparently healthy individuals and a better predictor than the presence of known disease or haploinsufficient genes for larger CNVs. The functionally related genes found in the de novo CNVs belonged to 70% of all clusters of functionally related genes found across the genome. De novo CNVs were more likely to affect functional clusters and affect them to a greater extent than benign CNVs (P = 6 × 10−4). Furthermore, such clusters of functionally related genes are phenotypically informative: Different patients possessing CNVs that affect the same cluster of functionally related genes exhibit more similar phenotypes than expected (P < 0.05). The spanning of multiple functionally similar genes by single CNVs contributes substantially to how these variants exert their pathogenic effects.
Collapse
|
372
|
Zuo X, Sun L, Yin X, Gao J, Sheng Y, Xu J, Zhang J, He C, Qiu Y, Wen G, Tian H, Zheng X, Liu S, Wang W, Li W, Cheng Y, Liu L, Chang Y, Wang Z, Li Z, Li L, Wu J, Fang L, Shen C, Zhou F, Liang B, Chen G, Li H, Cui Y, Xu A, Yang X, Hao F, Xu L, Fan X, Li Y, Wu R, Wang X, Liu X, Zheng M, Song S, Ji B, Fang H, Yu J, Sun Y, Hui Y, Zhang F, Yang R, Yang S, Zhang X. Whole-exome SNP array identifies 15 new susceptibility loci for psoriasis. Nat Commun 2015; 6:6793. [PMID: 25854761 PMCID: PMC4403312 DOI: 10.1038/ncomms7793] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 02/28/2015] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies (GWASs) have reproducibly associated ∼40 susceptibility loci with psoriasis. However, the missing heritability is evident and the contributions of coding variants have not yet been systematically evaluated. Here, we present a large-scale whole-exome array analysis for psoriasis consisting of 42,760 individuals. We discover 16 SNPs within 15 new genes/loci associated with psoriasis, including C1orf141, ZNF683, TMC6, AIM2, IL1RL1, CASR, SON, ZFYVE16, MTHFR, CCDC129, ZNF143, AP5B1, SYNE2, IFNGR2 and 3q26.2-q27 (P<5.00 × 10(-08)). In addition, we also replicate four known susceptibility loci TNIP1, NFKBIA, IL12B and LCE3D-LCE3E. These susceptibility variants identified in the current study collectively account for 1.9% of the psoriasis heritability. The variant within AIM2 is predicted to impact protein structure. Our findings increase the number of genetic risk factors for psoriasis and highlight new and plausible biological pathways in psoriasis.
Collapse
Affiliation(s)
- Xianbo Zuo
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Dermatology, No.2 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Liangdan Sun
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Dermatology, No.2 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Xianyong Yin
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Dermatology, No.2 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Jinping Gao
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Yujun Sheng
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People’s Hospital, Beijing 100044, China
| | - Chundi He
- Department of Dermatology, No.1 Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Ying Qiu
- Department of Dermatology, Jining No. 1 People’s Hospital, Jining, Shandong 272011, China
| | - Guangdong Wen
- Department of Dermatology, Peking University People’s Hospital, Beijing 100044, China
| | - Hongqing Tian
- Shandong Provincial Institute of Dermatology and Venereology, Jinan, Shandong 250022, China
| | - Xiaodong Zheng
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Shengxiu Liu
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Wenjun Wang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Weiran Li
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Yuyan Cheng
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Longdan Liu
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Yan Chang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Zaixing Wang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Zenggang Li
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Longnian Li
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Jianping Wu
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Ling Fang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Changbing Shen
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Fusheng Zhou
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Bo Liang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Gang Chen
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Hui Li
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Yong Cui
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Aie Xu
- The Third People's Hospital of Hangzhou, Hangzhou, Zhejiang 310009, China
| | - Xueqin Yang
- Department of Dermatology, General Hospital of PLA Air Force, Beijing 100036, China
| | - Fei Hao
- Department of Dermatology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Limin Xu
- Department of Dermatology, Tianjin Changzheng Hospital, Tianjin 300106, China
| | - Xing Fan
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Yuzhen Li
- Department of Dermatology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Rina Wu
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical College, Huhehot, Inner Mongolia 010050, China
| | - Xiuli Wang
- Shanghai Skin Diseases and STD Hospital, Shanghai 200050, China
| | - Xiaoming Liu
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, China
| | - Min Zheng
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhenjiang 310009, China
| | - Shunpeng Song
- Department of Dermatology, Dalian Dermatosis Hosptial, Liaoning 116011, China
| | - Bihua Ji
- Department of Dermatology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| | - Hong Fang
- Department of Dermatology, The First Affiliated Hospital of Zhejiang University School of Medicine, Zhenjiang 310006, China
| | - Jianbin Yu
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongxin Sun
- Department of Dermatology, Anshan Tanggangzi hosptial, Liaoning 210300, China
| | - Yan Hui
- Department of Dermatology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang 830054, China
| | - Furen Zhang
- Shandong Provincial Institute of Dermatology and Venereology, Jinan, Shandong 250022, China
| | - Rongya Yang
- Department of Dermatology, General Hospital of Beijing Military Command, Beijing 100010, China
| | - Sen Yang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| | - Xuejun Zhang
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Dermatology, No.2 Hospital, Anhui Medical University, Hefei, Anhui 230022, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui 230032, China
- State Key Lab Incubation of Dermatology, Ministry of Science and Technology, Hefei, Anhui 230032, China
- Key Lab of Dermatology, Ministry of Education, Hefei, Anhui 230032, China
- Key Lab of Gene Resources Utilization for Severe Inherited Disorders, Anhui 230032, China
| |
Collapse
|
373
|
Baglioni M, Russo F, Geraci F, Rizzo M, Rainaldi G, Pellegrini M. A new method for discovering disease-specific MiRNA-target regulatory networks. PLoS One 2015; 10:e0122473. [PMID: 25848944 PMCID: PMC4388573 DOI: 10.1371/journal.pone.0122473] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/11/2015] [Indexed: 12/27/2022] Open
Abstract
Genes and their expression regulation are among the key factors in the comprehension of the genesis and development of complex diseases. In this context, microRNAs (miRNAs) are post-transcriptional regulators that play an important role in gene expression since they are frequently deregulated in pathologies like cardiovascular disease and cancer. In vitro validation of miRNA - targets regulation is often too expensive and time consuming to be carried out for every possible alternative. As a result, a tool able to provide some criteria to prioritize trials is becoming a pressing need. Moreover, before planning in vitro experiments, the scientist needs to evaluate the miRNA-target genes interaction network. In this paper we describe the miRable method whose purpose is to identify new potentially relevant genes and their interaction networks associate to a specific pathology. To achieve this goal miRable follows a system biology approach integrating together general-purpose medical knowledge (literature, Protein-Protein Interaction networks, prediction tools) and pathology specific data (gene expression data). A case study on Prostate Cancer has shown that miRable is able to: 1) find new potential miRNA-targets pairs, 2) highlight novel genes potentially involved in a disease but never or little studied before, 3) reconstruct all possible regulatory subnetworks starting from the literature to expand the knowledge on the regulation of miRNA regulatory mechanisms.
Collapse
Affiliation(s)
- Miriam Baglioni
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Francesco Russo
- Laboratory of Integrative Systems Medicine (LISM), Institute of Informatics and Telematics (IIT) and Institute of Clinical Physiology (IFC), National Research Council (CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
- Department of Computer Science, University of Pisa, Largo Bruno Pontecorvo 3, 56127, Pisa, Italy
- * E-mail:
| | - Filippo Geraci
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Milena Rizzo
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Giuseppe Rainaldi
- Laboratory of Integrative Systems Medicine (LISM), Institute of Informatics and Telematics (IIT) and Institute of Clinical Physiology (IFC), National Research Council (CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Marco Pellegrini
- Laboratory of Integrative Systems Medicine (LISM), Institute of Informatics and Telematics (IIT) and Institute of Clinical Physiology (IFC), National Research Council (CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
374
|
Zhu F, Shi L, Engel JD, Guan Y. Regulatory network inferred using expression data of small sample size: application and validation in erythroid system. Bioinformatics 2015; 31:2537-44. [PMID: 25840044 DOI: 10.1093/bioinformatics/btv186] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/27/2015] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Modeling regulatory networks using expression data observed in a differentiation process may help identify context-specific interactions. The outcome of the current algorithms highly depends on the quality and quantity of a single time-course dataset, and the performance may be compromised for datasets with a limited number of samples. RESULTS In this work, we report a multi-layer graphical model that is capable of leveraging many publicly available time-course datasets, as well as a cell lineage-specific data with small sample size, to model regulatory networks specific to a differentiation process. First, a collection of network inference methods are used to predict the regulatory relationships in individual public datasets. Then, the inferred directional relationships are weighted and integrated together by evaluating against the cell lineage-specific dataset. To test the accuracy of this algorithm, we collected a time-course RNA-Seq dataset during human erythropoiesis to infer regulatory relationships specific to this differentiation process. The resulting erythroid-specific regulatory network reveals novel regulatory relationships activated in erythropoiesis, which were further validated by genome-wide TR4 binding studies using ChIP-seq. These erythropoiesis-specific regulatory relationships were not identifiable by single dataset-based methods or context-independent integrations. Analysis of the predicted targets reveals that they are all closely associated with hematopoietic lineage differentiation.
Collapse
Affiliation(s)
- Fan Zhu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | | | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA, Department of Internal Medicine, and Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
375
|
Liu C, Xuan Z. Prioritization of cancer-related genomic variants by SNP association network. Cancer Inform 2015; 14:57-70. [PMID: 25995611 PMCID: PMC4384763 DOI: 10.4137/cin.s17288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/11/2015] [Accepted: 01/13/2015] [Indexed: 12/11/2022] Open
Abstract
We have developed a general framework to construct an association network of single nucleotide polymorphisms (SNPs) (SNP association network, SAN) based on the functional interactions of genes located in the flanking regions of SNPs. SAN, which was constructed based on protein-protein interactions in the Human Protein Reference Database (HPRD), showed significantly enriched signals in both linkage disequilibrium (LD) and long-range chromatin interaction (Hi-C). We used this network to further develop two methods for predicting and prioritizing disease-associated genes from genome-wide association studies (GWASs). We found that random walk with restart (RWR) using SAN (RWR-SAN) can greatly improve the prediction of lung-cancer-associated genes by comparing RWR with the use of network in HPRD (AUC 0.81 vs 0.66). In a reanalysis of the GWAS dataset of age-related macular degeneration (AMD), SAN could identify more potential AMD-associated genes that were previously ranked lower in the GWAS study. The interactions in SAN could facilitate the study of complex diseases.
Collapse
Affiliation(s)
- Changning Liu
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, Texas, USA
- Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Xuan
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
376
|
Lee T, Kim H, Lee I. Network-assisted crop systems genetics: network inference and integrative analysis. CURRENT OPINION IN PLANT BIOLOGY 2015; 24:61-70. [PMID: 25698380 DOI: 10.1016/j.pbi.2015.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 01/15/2015] [Accepted: 02/02/2015] [Indexed: 05/24/2023]
Abstract
Although next-generation sequencing (NGS) technology has enabled the decoding of many crop species genomes, most of the underlying genetic components for economically important crop traits remain to be determined. Network approaches have proven useful for the study of the reference plant, Arabidopsis thaliana, and the success of network-based crop genetics will also require the availability of a genome-scale functional networks for crop species. In this review, we discuss how to construct functional networks and elucidate the holistic view of a crop system. The crop gene network then can be used for gene prioritization and the analysis of resequencing-based genome-wide association study (GWAS) data, the amount of which will rapidly grow in the field of crop science in the coming years.
Collapse
Affiliation(s)
- Tak Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hyojin Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
377
|
Jiang Y, Qin H, Yang L. Using network clustering to predict copy number variations associated with health disparities. PeerJ 2015; 3:e677. [PMID: 25780754 PMCID: PMC4358638 DOI: 10.7717/peerj.677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 01/08/2015] [Indexed: 11/20/2022] Open
Abstract
Substantial health disparities exist between African Americans and Caucasians in the United States. Copy number variations (CNVs) are one form of human genetic variations that have been linked with complex diseases and often occur at different frequencies among African Americans and Caucasian populations. Here, we aimed to investigate whether CNVs with differential frequencies can contribute to health disparities from the perspective of gene networks. We inferred network clusters from human gene/protein networks based on two different data sources. We then evaluated each network cluster for the occurrences of known pathogenic genes and genes located in CNVs with different population frequencies, and used false discovery rates to rank network clusters. This approach let us identify five clusters enriched with known pathogenic genes and with genes located in CNVs with different frequencies between African Americans and Caucasians. These clustering patterns predict two candidate causal genes located in four population-specific CNVs that play potential roles in health disparities
Collapse
Affiliation(s)
- Yi Jiang
- Department of Computer Science and Engineering, University of Tennessee at Chattanooga , TN , USA
| | - Hong Qin
- Departement of Biology, Spelman College , Atlanta, GA , United States
| | - Li Yang
- Department of Computer Science and Engineering, University of Tennessee at Chattanooga , TN , USA
| |
Collapse
|
378
|
Chen L, Chu C, Kong X, Huang G, Huang T, Cai YD. A hybrid computational method for the discovery of novel reproduction-related genes. PLoS One 2015; 10:e0117090. [PMID: 25768094 PMCID: PMC4358884 DOI: 10.1371/journal.pone.0117090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 12/13/2014] [Indexed: 12/12/2022] Open
Abstract
Uncovering the molecular mechanisms underlying reproduction is of great importance to infertility treatment and to the generation of healthy offspring. In this study, we discovered novel reproduction-related genes with a hybrid computational method, integrating three different types of method, which offered new clues for further reproduction research. This method was first executed on a weighted graph, constructed based on known protein-protein interactions, to search the shortest paths connecting any two known reproduction-related genes. Genes occurring in these paths were deemed to have a special relationship with reproduction. These newly discovered genes were filtered with a randomization test. Then, the remaining genes were further selected according to their associations with known reproduction-related genes measured by protein-protein interaction score and alignment score obtained by BLAST. The in-depth analysis of the high confidence novel reproduction genes revealed hidden mechanisms of reproduction and provided guidelines for further experimental validations.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People’s Republic of China
| | - Chen Chu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People’s Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, People’s Republic of China
| | - Guohua Huang
- Institute of Systems Biology, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, People’s Republic of China
- * E-mail: (TH); (YDC)
| | - Yu-Dong Cai
- Institute of Systems Biology, Shanghai University, Shanghai, 200444, People’s Republic of China
- * E-mail: (TH); (YDC)
| |
Collapse
|
379
|
Integrated genomic characterization of papillary thyroid carcinoma. Cell 2015; 159:676-90. [PMID: 25417114 DOI: 10.1016/j.cell.2014.09.050] [Citation(s) in RCA: 2144] [Impact Index Per Article: 214.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 09/16/2014] [Accepted: 09/23/2014] [Indexed: 02/07/2023]
Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. Here, we describe the genomic landscape of 496 PTCs. We observed a low frequency of somatic alterations (relative to other carcinomas) and extended the set of known PTC driver alterations to include EIF1AX, PPM1D, and CHEK2 and diverse gene fusions. These discoveries reduced the fraction of PTC cases with unknown oncogenic driver from 25% to 3.5%. Combined analyses of genomic variants, gene expression, and methylation demonstrated that different driver groups lead to different pathologies with distinct signaling and differentiation characteristics. Similarly, we identified distinct molecular subgroups of BRAF-mutant tumors, and multidimensional analyses highlighted a potential involvement of oncomiRs in less-differentiated subgroups. Our results propose a reclassification of thyroid cancers into molecular subtypes that better reflect their underlying signaling and differentiation properties, which has the potential to improve their pathological classification and better inform the management of the disease.
Collapse
Affiliation(s)
-
- Cancer Genome Atlas Program Office, National Cancer Institute at NIH, 31 Center Drive, Bldg. 31, Suite 3A20, Bethesda MD 20892, USA.
| |
Collapse
|
380
|
Kim H, Jung KW, Maeng S, Chen YL, Shin J, Shim JE, Hwang S, Janbon G, Kim T, Heitman J, Bahn YS, Lee I. Network-assisted genetic dissection of pathogenicity and drug resistance in the opportunistic human pathogenic fungus Cryptococcus neoformans. Sci Rep 2015; 5:8767. [PMID: 25739925 PMCID: PMC4350084 DOI: 10.1038/srep08767] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/29/2015] [Indexed: 12/12/2022] Open
Abstract
Cryptococcus neoformans is an opportunistic human pathogenic fungus that causes meningoencephalitis. Due to the increasing global risk of cryptococcosis and the emergence of drug-resistant strains, the development of predictive genetics platforms for the rapid identification of novel genes governing pathogenicity and drug resistance of C. neoformans is imperative. The analysis of functional genomics data and genome-scale mutant libraries may facilitate the genetic dissection of such complex phenotypes but with limited efficiency. Here, we present a genome-scale co-functional network for C. neoformans, CryptoNet, which covers ~81% of the coding genome and provides an efficient intermediary between functional genomics data and reverse-genetics resources for the genetic dissection of C. neoformans phenotypes. CryptoNet is the first genome-scale co-functional network for any fungal pathogen. CryptoNet effectively identified novel genes for pathogenicity and drug resistance using guilt-by-association and context-associated hub algorithms. CryptoNet is also the first genome-scale co-functional network for fungi in the basidiomycota phylum, as Saccharomyces cerevisiae belongs to the ascomycota phylum. CryptoNet may therefore provide insights into pathway evolution between two distinct phyla of the fungal kingdom. The CryptoNet web server (www.inetbio.org/cryptonet) is a public resource that provides an interactive environment of network-assisted predictive genetics for C. neoformans.
Collapse
Affiliation(s)
- Hanhae Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Kwang-Woo Jung
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Shinae Maeng
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Ying-Lien Chen
- 1] Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA [2] Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Junha Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Jung Eun Shim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Sohyun Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Guilhem Janbon
- Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, F-75015, Paris, France
| | - Taeyup Kim
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Medicine, and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yong-Sun Bahn
- Department of Biotechnology, Center for Fungal Pathogenesis, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul. 120-749, Korea
| |
Collapse
|
381
|
Kuperstein I, Grieco L, Cohen DPA, Thieffry D, Zinovyev A, Barillot E. The shortest path is not the one you know: application of biological network resources in precision oncology research. Mutagenesis 2015; 30:191-204. [PMID: 25688112 DOI: 10.1093/mutage/geu078] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Several decades of molecular biology research have delivered a wealth of detailed descriptions of molecular interactions in normal and tumour cells. This knowledge has been functionally organised and assembled into dedicated biological pathway resources that serve as an invaluable tool, not only for structuring the information about molecular interactions but also for making it available for biological, clinical and computational studies. With the advent of high-throughput molecular profiling of tumours, close to complete molecular catalogues of mutations, gene expression and epigenetic modifications are available and require adequate interpretation. Taking into account the information about biological signalling machinery in cells may help to better interpret molecular profiles of tumours. Making sense out of these descriptions requires biological pathway resources for functional interpretation of the data. In this review, we describe the available biological pathway resources, their characteristics in terms of construction mode, focus, aims and paradigms of biological knowledge representation. We present a new resource that is focused on cancer-related signalling, the Atlas of Cancer Signalling Networks. We briefly discuss current approaches for data integration, visualisation and analysis, using biological networks, such as pathway scoring, guilt-by-association and network propagation. Finally, we illustrate with several examples the added value of data interpretation in the context of biological networks and demonstrate that it may help in analysis of high-throughput data like mutation, gene expression or small interfering RNA screening and can guide in patients stratification. Finally, we discuss perspectives for improving precision medicine using biological network resources and tools. Taking into account the information about biological signalling machinery in cells may help to better interpret molecular patterns of tumours and enable to put precision oncology into general clinical practice.
Collapse
Affiliation(s)
- Inna Kuperstein
- Institut Curie, 26 rue d'Ulm, Paris F-75248, France, INSERM, U900, Paris F-75248, France, Mines ParisTech, Fontainebleau F-77300, France
| | - Luca Grieco
- Institut Curie, 26 rue d'Ulm, Paris F-75248, France, INSERM, U900, Paris F-75248, France, Mines ParisTech, Fontainebleau F-77300, France, Ecole Normale Supérieure - Département de Biologie, IBENS, 46 rue d'Ulm, Paris F-75005, France, CNRS, UMR8197, Paris F 75005, France and INSERM, U1024, Paris F 75005, France Present address: Clinical Operational Research Unit, University College London, 4 Taviton Street, London WC1H 0BT, UK
| | - David P A Cohen
- Institut Curie, 26 rue d'Ulm, Paris F-75248, France, INSERM, U900, Paris F-75248, France, Mines ParisTech, Fontainebleau F-77300, France
| | - Denis Thieffry
- Ecole Normale Supérieure - Département de Biologie, IBENS, 46 rue d'Ulm, Paris F-75005, France, CNRS, UMR8197, Paris F 75005, France and INSERM, U1024, Paris F 75005, France
| | - Andrei Zinovyev
- Institut Curie, 26 rue d'Ulm, Paris F-75248, France, INSERM, U900, Paris F-75248, France, Mines ParisTech, Fontainebleau F-77300, France
| | - Emmanuel Barillot
- Institut Curie, 26 rue d'Ulm, Paris F-75248, France, INSERM, U900, Paris F-75248, France, Mines ParisTech, Fontainebleau F-77300, France,
| |
Collapse
|
382
|
Menche J, Sharma A, Kitsak M, Ghiassian SD, Vidal M, Loscalzo J, Barabási AL. Disease networks. Uncovering disease-disease relationships through the incomplete interactome. Science 2015; 347:1257601. [PMID: 25700523 PMCID: PMC4435741 DOI: 10.1126/science.1257601] [Citation(s) in RCA: 946] [Impact Index Per Article: 94.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
According to the disease module hypothesis, the cellular components associated with a disease segregate in the same neighborhood of the human interactome, the map of biologically relevant molecular interactions. Yet, given the incompleteness of the interactome and the limited knowledge of disease-associated genes, it is not obvious if the available data have sufficient coverage to map out modules associated with each disease. Here we derive mathematical conditions for the identifiability of disease modules and show that the network-based location of each disease module determines its pathobiological relationship to other diseases. For example, diseases with overlapping network modules show significant coexpression patterns, symptom similarity, and comorbidity, whereas diseases residing in separated network neighborhoods are phenotypically distinct. These tools represent an interactome-based platform to predict molecular commonalities between phenotypically related diseases, even if they do not share primary disease genes.
Collapse
Affiliation(s)
- Jörg Menche
- Center for Complex Networks Research and Department of Physics, Northeastern University, 110 Forsyth Street, 111 Dana Research Center, Boston, MA 02115, USA. Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA. Center for Network Science, Central European University, Nador u. 9, 1051 Budapest, Hungary
| | - Amitabh Sharma
- Center for Complex Networks Research and Department of Physics, Northeastern University, 110 Forsyth Street, 111 Dana Research Center, Boston, MA 02115, USA. Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Maksim Kitsak
- Center for Complex Networks Research and Department of Physics, Northeastern University, 110 Forsyth Street, 111 Dana Research Center, Boston, MA 02115, USA. Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Susan Dina Ghiassian
- Center for Complex Networks Research and Department of Physics, Northeastern University, 110 Forsyth Street, 111 Dana Research Center, Boston, MA 02115, USA. Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA. Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Albert-László Barabási
- Center for Complex Networks Research and Department of Physics, Northeastern University, 110 Forsyth Street, 111 Dana Research Center, Boston, MA 02115, USA. Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA. Center for Network Science, Central European University, Nador u. 9, 1051 Budapest, Hungary. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
383
|
Peng J, Uygun S, Kim T, Wang Y, Rhee SY, Chen J. Measuring semantic similarities by combining gene ontology annotations and gene co-function networks. BMC Bioinformatics 2015; 16:44. [PMID: 25886899 PMCID: PMC4339680 DOI: 10.1186/s12859-015-0474-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 01/26/2015] [Indexed: 01/18/2023] Open
Abstract
Background Gene Ontology (GO) has been used widely to study functional relationships between genes. The current semantic similarity measures rely only on GO annotations and GO structure. This limits the power of GO-based similarity because of the limited proportion of genes that are annotated to GO in most organisms. Results We introduce a novel approach called NETSIM (network-based similarity measure) that incorporates information from gene co-function networks in addition to using the GO structure and annotations. Using metabolic reaction maps of yeast, Arabidopsis, and human, we demonstrate that NETSIM can improve the accuracy of GO term similarities. We also demonstrate that NETSIM works well even for genomes with sparser gene annotation data. We applied NETSIM on large Arabidopsis gene families such as cytochrome P450 monooxygenases to group the members functionally and show that this grouping could facilitate functional characterization of genes in these families. Conclusions Using NETSIM as an example, we demonstrated that the performance of a semantic similarity measure could be significantly improved after incorporating genome-specific information. NETSIM incorporates both GO annotations and gene co-function network data as a priori knowledge in the model. Therefore, functional similarities of GO terms that are not explicitly encoded in GO but are relevant in a taxon-specific manner become measurable when GO annotations are limited. Supplementary information and software are available at http://www.msu.edu/~jinchen/NETSIM. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0474-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiajie Peng
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China. .,Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, 48824, USA.
| | - Sahra Uygun
- Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, 48824, USA. .,Genetics Program, Michigan State University, East Lansing, MI, 48824, USA.
| | - Taehyong Kim
- Department of Plant Biology, Carnegie Institution for Science, 260 Panama St, Stanford, CA, 94305, USA.
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.
| | - Seung Y Rhee
- Department of Plant Biology, Carnegie Institution for Science, 260 Panama St, Stanford, CA, 94305, USA.
| | - Jin Chen
- Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, 48824, USA. .,Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
384
|
Biological interpretation of genome-wide association studies using predicted gene functions. Nat Commun 2015; 6:5890. [PMID: 25597830 DOI: 10.1038/ncomms6890] [Citation(s) in RCA: 540] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/18/2014] [Indexed: 12/17/2022] Open
Abstract
The main challenge for gaining biological insights from genetic associations is identifying which genes and pathways explain the associations. Here we present DEPICT, an integrative tool that employs predicted gene functions to systematically prioritize the most likely causal genes at associated loci, highlight enriched pathways and identify tissues/cell types where genes from associated loci are highly expressed. DEPICT is not limited to genes with established functions and prioritizes relevant gene sets for many phenotypes.
Collapse
|
385
|
Taşan M, Musso G, Hao T, Vidal M, MacRae CA, Roth FP. Selecting causal genes from genome-wide association studies via functionally coherent subnetworks. Nat Methods 2014; 12:154-9. [PMID: 25532137 DOI: 10.1038/nmeth.3215] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 11/24/2014] [Indexed: 12/27/2022]
Abstract
Genome-wide association (GWA) studies have linked thousands of loci to human diseases, but the causal genes and variants at these loci generally remain unknown. Although investigators typically focus on genes closest to the associated polymorphisms, the causal gene is often more distal. Reliance on published work to prioritize candidates is biased toward well-characterized genes. We describe a 'prix fixe' strategy and software that uses genome-scale shared-function networks to identify sets of mutually functionally related genes spanning multiple GWA loci. Using associations from ∼100 GWA studies covering ten cancer types, our approach outperformed the common alternative strategy in ranking known cancer genes. As more GWA loci are discovered, the strategy will have increased power to elucidate the causes of human disease.
Collapse
Affiliation(s)
- Murat Taşan
- 1] Donnelly Centre, University of Toronto, Toronto, Ontario, Canada. [2] Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada. [3] Department of Computer Science, University of Toronto, Toronto, Ontario, Canada. [4] Center for Cancer Systems Biology (CCSB), Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA. [5] Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Gabriel Musso
- 1] Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA. [2] Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tong Hao
- 1] Center for Cancer Systems Biology (CCSB), Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Marc Vidal
- 1] Center for Cancer Systems Biology (CCSB), Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Calum A MacRae
- 1] Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA. [2] Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Frederick P Roth
- 1] Donnelly Centre, University of Toronto, Toronto, Ontario, Canada. [2] Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada. [3] Department of Computer Science, University of Toronto, Toronto, Ontario, Canada. [4] Center for Cancer Systems Biology (CCSB), Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA. [5] Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada. [6] Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| |
Collapse
|
386
|
Bargsten JW, Nap JP, Sanchez-Perez GF, van Dijk ADJ. Prioritization of candidate genes in QTL regions based on associations between traits and biological processes. BMC PLANT BIOLOGY 2014; 14:330. [PMID: 25492368 PMCID: PMC4274756 DOI: 10.1186/s12870-014-0330-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/10/2014] [Indexed: 05/18/2023]
Abstract
BACKGROUND Elucidation of genotype-to-phenotype relationships is a major challenge in biology. In plants, it is the basis for molecular breeding. Quantitative Trait Locus (QTL) mapping enables to link variation at the trait level to variation at the genomic level. However, QTL regions typically contain tens to hundreds of genes. In order to prioritize such candidate genes, we show that we can identify potentially causal genes for a trait based on overrepresentation of biological processes (gene functions) for the candidate genes in the QTL regions of that trait. RESULTS The prioritization method was applied to rice QTL data, using gene functions predicted on the basis of sequence- and expression-information. The average reduction of the number of genes was over ten-fold. Comparison with various types of experimental datasets (including QTL fine-mapping and Genome Wide Association Study results) indicated both statistical significance and biological relevance of the obtained connections between genes and traits. A detailed analysis of flowering time QTLs illustrates that genes with completely unknown function are likely to play a role in this important trait. CONCLUSIONS Our approach can guide further experimentation and validation of causal genes for quantitative traits. This way it capitalizes on QTL data to uncover how individual genes influence trait variation.
Collapse
Affiliation(s)
- Joachim W Bargsten
- />Applied Bioinformatics, Bioscience, Plant Sciences Group, Wageningen University and Research Centre, Wageningen, The Netherlands
- />Netherlands Bioinformatics Centre (NBIC), Nijmegen, The Netherlands
- />Laboratory for Plant Breeding, Plant Sciences Group, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Jan-Peter Nap
- />Applied Bioinformatics, Bioscience, Plant Sciences Group, Wageningen University and Research Centre, Wageningen, The Netherlands
- />Netherlands Bioinformatics Centre (NBIC), Nijmegen, The Netherlands
| | - Gabino F Sanchez-Perez
- />Applied Bioinformatics, Bioscience, Plant Sciences Group, Wageningen University and Research Centre, Wageningen, The Netherlands
- />Laboratory of Bioinformatics, Plant Sciences Group, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Aalt DJ van Dijk
- />Applied Bioinformatics, Bioscience, Plant Sciences Group, Wageningen University and Research Centre, Wageningen, The Netherlands
- />Biometris, Wageningen University and Research Centre, Wageningen, The Netherlands
| |
Collapse
|
387
|
Deo RC, Musso G, Tasan M, Tang P, Poon A, Yuan C, Felix JF, Vasan RS, Beroukhim R, De Marco T, Kwok PY, MacRae CA, Roth FP. Prioritizing causal disease genes using unbiased genomic features. Genome Biol 2014; 15:534. [PMID: 25633252 PMCID: PMC4279789 DOI: 10.1186/s13059-014-0534-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 11/06/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is the leading cause of death in the developed world. Human genetic studies, including genome-wide sequencing and SNP-array approaches, promise to reveal disease genes and mechanisms representing new therapeutic targets. In practice, however, identification of the actual genes contributing to disease pathogenesis has lagged behind identification of associated loci, thus limiting the clinical benefits. RESULTS To aid in localizing causal genes, we develop a machine learning approach, Objective Prioritization for Enhanced Novelty (OPEN), which quantitatively prioritizes gene-disease associations based on a diverse group of genomic features. This approach uses only unbiased predictive features and thus is not hampered by a preference towards previously well-characterized genes. We demonstrate success in identifying genetic determinants for CVD-related traits, including cholesterol levels, blood pressure, and conduction system and cardiomyopathy phenotypes. Using OPEN, we prioritize genes, including FLNC, for association with increased left ventricular diameter, which is a defining feature of a prevalent cardiovascular disorder, dilated cardiomyopathy or DCM. Using a zebrafish model, we experimentally validate FLNC and identify a novel FLNC splice-site mutation in a patient with severe DCM. CONCLUSION Our approach stands to assist interpretation of large-scale genetic studies without compromising their fundamentally unbiased nature.
Collapse
Affiliation(s)
- Rahul C Deo
- />Cardiovascular Research Institute, University of California, San Francisco, CA 94158 USA
- />Department of Medicine, University of California, San Francisco, CA 94143 USA
- />Institute for Human Genetics, University of California, San Francisco, CA 94158 USA
- />California Institute for Quantitative Biosciences, San Francisco, CA 94143 USA
- />Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
| | - Gabriel Musso
- />Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
- />Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Murat Tasan
- />Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
- />Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto and Lunenfeld Research Institute, Mt Sinai Hospital, Toronto, Ontario M5G 1X5 Canada
| | - Paul Tang
- />Institute for Human Genetics, University of California, San Francisco, CA 94158 USA
| | - Annie Poon
- />Institute for Human Genetics, University of California, San Francisco, CA 94158 USA
| | - Christiana Yuan
- />Cardiovascular Research Institute, University of California, San Francisco, CA 94158 USA
| | - Janine F Felix
- />Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Ramachandran S Vasan
- />Preventive Medicine and Cardiology Sections, and Department of Medicine, Boston University School of Medicine, Boston, MA 02118 USA
- />Framingham Heart Study, Boston University School of Medicine, Framingham, MA 01702 USA
| | - Rameen Beroukhim
- />Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
- />Center for Cancer Genome Discovery and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - Teresa De Marco
- />Department of Medicine, University of California, San Francisco, CA 94143 USA
| | - Pui-Yan Kwok
- />Cardiovascular Research Institute, University of California, San Francisco, CA 94158 USA
- />Institute for Human Genetics, University of California, San Francisco, CA 94158 USA
| | - Calum A MacRae
- />Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
| | - Frederick P Roth
- />Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 USA
- />Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto and Lunenfeld Research Institute, Mt Sinai Hospital, Toronto, Ontario M5G 1X5 Canada
- />Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
- />The Canadian Institute for Advanced Research, Toronto, ON M5G 1Z8 Canada
| |
Collapse
|
388
|
Nelson RM, Pettersson ME. Degrees of separation as a statistical tool for evaluating candidate genes. Comput Biol Med 2014; 55:49-52. [PMID: 25450218 DOI: 10.1016/j.compbiomed.2014.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/22/2014] [Accepted: 10/01/2014] [Indexed: 11/29/2022]
Abstract
Selection of candidate genes is an important step in the exploration of complex genetic architecture. The number of gene networks available is increasing and these can provide information to help with candidate gene selection. It is currently common to use the degree of connectedness in gene networks as validation in Genome Wide Association (GWA) and Quantitative Trait Locus (QTL) mapping studies. However, it can cause misleading results if not validated properly. Here we present a method and tool for validating the gene pairs from GWA studies given the context of the network they co-occur in. It ensures that proposed interactions and gene associations are not statistical artefacts inherent to the specific gene network architecture. The CandidateBacon package provides an easy and efficient method to calculate the average degree of separation (DoS) between pairs of genes to currently available gene networks. We show how these empirical estimates of average connectedness are used to validate candidate gene pairs. Validation of interacting genes by comparing their connectedness with the average connectedness in the gene network will provide support for said interactions by utilising the growing amount of gene network information available.
Collapse
Affiliation(s)
- Ronald M Nelson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Mats E Pettersson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
389
|
Chan KHK, Huang YT, Meng Q, Wu C, Reiner A, Sobel EM, Tinker L, Lusis AJ, Yang X, Liu S. Shared molecular pathways and gene networks for cardiovascular disease and type 2 diabetes mellitus in women across diverse ethnicities. CIRCULATION. CARDIOVASCULAR GENETICS 2014; 7:911-9. [PMID: 25371518 DOI: 10.1161/circgenetics.114.000676] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although cardiovascular disease (CVD) and type 2 diabetes mellitus (T2D) share many common risk factors, potential molecular mechanisms that may also be shared for these 2 disorders remain unknown. METHODS AND RESULTS Using an integrative pathway and network analysis, we performed genome-wide association studies in 8155 blacks, 3494 Hispanic American, and 3697 Caucasian American women who participated in the national Women's Health Initiative single-nucleotide polymorphism (SNP) Health Association Resource and the Genomics and Randomized Trials Network. Eight top pathways and gene networks related to cardiomyopathy, calcium signaling, axon guidance, cell adhesion, and extracellular matrix seemed to be commonly shared between CVD and T2D across all 3 ethnic groups. We also identified ethnicity-specific pathways, such as cell cycle (specific for Hispanic American and Caucasian American) and tight junction (CVD and combined CVD and T2D in Hispanic American). In network analysis of gene-gene or protein-protein interactions, we identified key drivers that included COL1A1, COL3A1, and ELN in the shared pathways for both CVD and T2D. These key driver genes were cross-validated in multiple mouse models of diabetes mellitus and atherosclerosis. CONCLUSIONS Our integrative analysis of American women of 3 ethnicities identified multiple shared biological pathways and key regulatory genes for the development of CVD and T2D. These prospective findings also support the notion that ethnicity-specific susceptibility genes and process are involved in the pathogenesis of CVD and T2D.
Collapse
Affiliation(s)
- Kei Hang K Chan
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Yen-Tsung Huang
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Qingying Meng
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Chunyuan Wu
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Alexander Reiner
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Eric M Sobel
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Lesley Tinker
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Aldons J Lusis
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.)
| | - Xia Yang
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.).
| | - Simin Liu
- From the Department of Epidemiology (K.H.K.C., Y.-T.H., S.L.) and Division of Endocrinology, Department of Medicine (S.L.), Warren Alpert Medical School of Brown University, Providence, RI; Department of Integrative Biology and Physiology (K.H.K.C., Q.M., X.Y.), Department of Human Genetics (E.M.S.), Department of Medicine/Division of Cardiology, David Geffen School of Medicine (A.J.L.), and Departments of Medicine and Obstetrics and Gynecology, David Geffen School of Medicine (S.L.), University of California Los Angeles; Biostatistics Division (C.W.), Public Health Sciences Division (L.T.), Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Epidemiology, University of Washington, Seattle (A.R.).
| |
Collapse
|
390
|
Kwon T, Chung MI, Gupta R, Baker JC, Wallingford JB, Marcotte EM. Identifying direct targets of transcription factor Rfx2 that coordinate ciliogenesis and cell movement. GENOMICS DATA 2014; 2:192-194. [PMID: 25419512 PMCID: PMC4236849 DOI: 10.1016/j.gdata.2014.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recently, using the frog Xenopus laevis as a model system, we showed that transcription factor Rfx2 coordinates many genes involved in ciliogenesis and cell movement in multiciliated cells (Chung et al., 2014). To our knowledge, it was the first paper to utilize the genomic resources, including genome sequences and interim gene annotations, from the ongoing Xenopus laevis genome project. For researchers who are interested in the application of genomics and systems biology approaches in Xenopus studies, here we provide additional details about our dataset (NCBI GEO accession number GSE50593) and describe how we analyzed RNA-seq and ChIP-seq data to identify direct targets of Rfx2.
Collapse
Affiliation(s)
- Taejoon Kwon
- Department of Molecular Biosciences, University of Texas at Austin
| | - Mei-I Chung
- Department of Molecular Biosciences, University of Texas at Austin
| | | | | | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin ; Center for Systems & Synthetic Biology and Institute for Cellular & Molecular Biology, University of Texas ; Howard Hughes Medical Institute
| | - Edward M Marcotte
- Department of Molecular Biosciences, University of Texas at Austin ; Center for Systems & Synthetic Biology and Institute for Cellular & Molecular Biology, University of Texas
| |
Collapse
|
391
|
Xu Y, Guo M, Zou Q, Liu X, Wang C, Liu Y. System-level insights into the cellular interactome of a non-model organism: inferring, modelling and analysing functional gene network of soybean (Glycine max). PLoS One 2014; 9:e113907. [PMID: 25423109 PMCID: PMC4244207 DOI: 10.1371/journal.pone.0113907] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/24/2014] [Indexed: 01/30/2023] Open
Abstract
Cellular interactome, in which genes and/or their products interact on several levels, forming transcriptional regulatory-, protein interaction-, metabolic-, signal transduction networks, etc., has attracted decades of research focuses. However, such a specific type of network alone can hardly explain the various interactive activities among genes. These networks characterize different interaction relationships, implying their unique intrinsic properties and defects, and covering different slices of biological information. Functional gene network (FGN), a consolidated interaction network that models fuzzy and more generalized notion of gene-gene relations, have been proposed to combine heterogeneous networks with the goal of identifying functional modules supported by multiple interaction types. There are yet no successful precedents of FGNs on sparsely studied non-model organisms, such as soybean (Glycine max), due to the absence of sufficient heterogeneous interaction data. We present an alternative solution for inferring the FGNs of soybean (SoyFGNs), in a pioneering study on the soybean interactome, which is also applicable to other organisms. SoyFGNs exhibit the typical characteristics of biological networks: scale-free, small-world architecture and modularization. Verified by co-expression and KEGG pathways, SoyFGNs are more extensive and accurate than an orthology network derived from Arabidopsis. As a case study, network-guided disease-resistance gene discovery indicates that SoyFGNs can provide system-level studies on gene functions and interactions. This work suggests that inferring and modelling the interactome of a non-model plant are feasible. It will speed up the discovery and definition of the functions and interactions of other genes that control important functions, such as nitrogen fixation and protein or lipid synthesis. The efforts of the study are the basis of our further comprehensive studies on the soybean functional interactome at the genome and microRNome levels. Additionally, a web tool for information retrieval and analysis of SoyFGNs can be accessed at SoyFN: http://nclab.hit.edu.cn/SoyFN.
Collapse
Affiliation(s)
- Yungang Xu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Maozu Guo
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Quan Zou
- School of Information Science and Technology, Xiamen University, Xiamen, China
| | - Xiaoyan Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Chunyu Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yang Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
392
|
Correia C, Oliveira G, Vicente AM. Protein interaction networks reveal novel autism risk genes within GWAS statistical noise. PLoS One 2014; 9:e112399. [PMID: 25409314 PMCID: PMC4237351 DOI: 10.1371/journal.pone.0112399] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 10/15/2014] [Indexed: 11/19/2022] Open
Abstract
Genome-wide association studies (GWAS) for Autism Spectrum Disorder (ASD) thus far met limited success in the identification of common risk variants, consistent with the notion that variants with small individual effects cannot be detected individually in single SNP analysis. To further capture disease risk gene information from ASD association studies, we applied a network-based strategy to the Autism Genome Project (AGP) and the Autism Genetics Resource Exchange GWAS datasets, combining family-based association data with Human Protein-Protein interaction (PPI) data. Our analysis showed that autism-associated proteins at higher than conventional levels of significance (P<0.1) directly interact more than random expectation and are involved in a limited number of interconnected biological processes, indicating that they are functionally related. The functionally coherent networks generated by this approach contain ASD-relevant disease biology, as demonstrated by an improved positive predictive value and sensitivity in retrieving known ASD candidate genes relative to the top associated genes from either GWAS, as well as a higher gene overlap between the two ASD datasets. Analysis of the intersection between the networks obtained from the two ASD GWAS and six unrelated disease datasets identified fourteen genes exclusively present in the ASD networks. These are mostly novel genes involved in abnormal nervous system phenotypes in animal models, and in fundamental biological processes previously implicated in ASD, such as axon guidance, cell adhesion or cytoskeleton organization. Overall, our results highlighted novel susceptibility genes previously hidden within GWAS statistical "noise" that warrant further analysis for causal variants.
Collapse
Affiliation(s)
- Catarina Correia
- Departamento de Promoção da Saúde e Doenças não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, 1649-016 Lisboa, Portugal
- Center for Biodiversity, Functional & Integrative Genomics, Faculty of Sciences, University of Lisbon, 1749-016 Lisboa, Portugal
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Guiomar Oliveira
- Unidade Neurodesenvolvimento e Autismo, Centro de Desenvolvimento, Hospital Pediátrico (HP) do Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-602 Coimbra, Portugal
- Centro de Investigação e Formação Clinica do HP-CHUC, 3000-602 Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, 3000-548 Coimbra, Portugal
| | - Astrid M. Vicente
- Departamento de Promoção da Saúde e Doenças não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, 1649-016 Lisboa, Portugal
- Center for Biodiversity, Functional & Integrative Genomics, Faculty of Sciences, University of Lisbon, 1749-016 Lisboa, Portugal
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
- * E-mail:
| |
Collapse
|
393
|
Bag S, Anbarasu A. Revealing the Strong Functional Association of adipor2 and cdh13 with adipoq: A Gene Network Study. Cell Biochem Biophys 2014; 71:1445-56. [PMID: 25388841 DOI: 10.1007/s12013-014-0367-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
394
|
A derived network-based interferon-related signature of human macrophages responding to Mycobacterium tuberculosis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:713071. [PMID: 25371902 PMCID: PMC4209755 DOI: 10.1155/2014/713071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 12/11/2022]
Abstract
Network analysis of transcriptional signature typically relies on direct interaction between two highly expressed genes. However, this approach misses indirect and biological relevant interactions through a third factor (hub). Here we determine whether a hub-based network analysis can select an improved signature subset that correlates with a biological change in a stronger manner than the original signature. We have previously reported an interferon-related transcriptional signature (THP1r2Mtb-induced) from Mycobacterium tuberculosis (M. tb)-infected THP-1 human macrophage. We selected hub-connected THP1r2Mtb-induced genes into the refined network signature TMtb-iNet and grouped the excluded genes into the excluded signature TMtb-iEx. TMtb-iNet retained the enrichment of binding sites of interferon-related transcription factors and contained relatively more interferon-related interacting genes when compared to THP1r2Mtb-induced signature. TMtb-iNet correlated as strongly as THP1r2Mtb-induced signature on a public transcriptional dataset of patients with pulmonary tuberculosis (PTB). TMtb-iNet correlated more strongly in CD4(+) and CD8(+) T cells from PTB patients than THP1r2Mtb-induced signature and TMtb-iEx. When TMtb-iNet was applied to data during clinical therapy of tuberculosis, it resulted in the most pronounced response and the weakest correlation. Correlation on dataset from patients with AIDS or malaria was stronger for TMtb-iNet, indicating an involvement of TMtb-iNet in these chronic human infections. Collectively, the significance of this work is twofold: (1) we disseminate a hub-based approach in generating a biologically meaningful and clinically useful signature; (2) using this approach we introduce a new network-based signature and demonstrate its promising applications in understanding host responses to infections.
Collapse
|
395
|
He H, Zhang L, Li J, Wang YP, Zhang JG, Shen J, Guo YF, Deng HW. Integrative analysis of GWASs, human protein interaction, and gene expression identified gene modules associated with BMDs. J Clin Endocrinol Metab 2014; 99:E2392-E2399. [PMID: 25119315 PMCID: PMC4223444 DOI: 10.1210/jc.2014-2563] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/04/2014] [Indexed: 01/23/2023]
Abstract
CONTEXT To date, few systems genetics studies in the bone field have been performed. We designed our study from a systems-level perspective by integrating genome-wide association studies (GWASs), human protein-protein interaction (PPI) network, and gene expression to identify gene modules contributing to osteoporosis risk. METHODS First we searched for modules significantly enriched with bone mineral density (BMD)-associated genes in human PPI network by using 2 large meta-analysis GWAS datasets through a dense module search algorithm. One included 7 individual GWAS samples (Meta7). The other was from the Genetic Factors for Osteoporosis Consortium (GEFOS2). One was assigned as a discovery dataset and the other as an evaluation dataset, and vice versa. RESULTS In total, 42 modules and 129 modules were identified significantly in both Meta7 and GEFOS2 datasets for femoral neck and spine BMD, respectively. There were 3340 modules identified for hip BMD only in Meta7. As candidate modules, they were assessed for the biological relevance to BMD by gene set enrichment analysis in 2 expression profiles generated from circulating monocytes in subjects with low versus high BMD values. Interestingly, there were 2 modules significantly enriched in monocytes from the low BMD group in both gene expression datasets (nominal P value <.05). Two modules had 16 nonredundant genes. Functional enrichment analysis revealed that both modules were enriched for genes involved in Wnt receptor signaling and osteoblast differentiation. CONCLUSION We highlighted 2 modules and novel genes playing important roles in the regulation of bone mass, providing important clues for therapeutic approaches for osteoporosis.
Collapse
Affiliation(s)
- Hao He
- Center of Genomics and Bioinformatics and Department of Biostatistics and Bioinformatics (H.H., L.Z., J.L., Y.-P.W., J.-G.Z., H.-W.D.), Tulane University, New Orleans, Louisiana 70112; Biomedical Engineering Department (Y.-P.W.), Tulane University, New Orleans, Louisiana 70118; and Third Affiliated Hospital (J.S., Y.-F.G., H.-W.D.), China Southern Medical University, Guang Zhou 510000, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
396
|
Wang X, Zhang D, Tzeng JY. Pathway-guided identification of gene-gene interactions. Ann Hum Genet 2014; 78:478-91. [PMID: 25227508 PMCID: PMC4363308 DOI: 10.1111/ahg.12080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/03/2014] [Indexed: 12/26/2022]
Abstract
Assessing gene-gene interactions (GxG) at the gene level can permit examination of epistasis at biologically functional units with amplified interaction signals from marker-marker pairs. While current gene-based GxG methods tend to be designed for two or a few genes, for complex traits, it is often common to have a list of many candidate genes to explore GxG. We propose a regression model with pathway-guided regularization for detecting interactions among genes. Specifically, we use the principal components to summarize the SNP-SNP interactions between a gene pair, and use an L1 penalty that incorporates adaptive weights based on biological guidance and trait supervision to identify important main and interaction effects. Our approach aims to combine biological guidance and data adaptiveness, and yields credible findings that may be likely to shed insights in order to formulate biological hypotheses for further molecular studies. The proposed approach can be used to explore the GxG with a list of many candidate genes and is applicable even when sample size is smaller than the number of predictors studied. We evaluate the utility of the proposed method using simulation and real data analysis. The results suggest improved performance over methods not utilizing pathway and trait guidance.
Collapse
Affiliation(s)
- Xin Wang
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Daowen Zhang
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Jung-Ying Tzeng
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
- Department of Statistics, National Cheng-Kung University, Tainan, Taiwan
| |
Collapse
|
397
|
Emmert-Streib F, Tripathi S, Simoes RDM, Hawwa AF, Dehmer M. The human disease network. ACTA ACUST UNITED AC 2014. [DOI: 10.4161/sysb.22816] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
398
|
Luo Y, Riedlinger G, Szolovits P. Text mining in cancer gene and pathway prioritization. Cancer Inform 2014; 13:69-79. [PMID: 25392685 PMCID: PMC4216063 DOI: 10.4137/cin.s13874] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/18/2014] [Accepted: 05/18/2014] [Indexed: 12/18/2022] Open
Abstract
Prioritization of cancer implicated genes has received growing attention as an effective way to reduce wet lab cost by computational analysis that ranks candidate genes according to the likelihood that experimental verifications will succeed. A multitude of gene prioritization tools have been developed, each integrating different data sources covering gene sequences, differential expressions, function annotations, gene regulations, protein domains, protein interactions, and pathways. This review places existing gene prioritization tools against the backdrop of an integrative Omic hierarchy view toward cancer and focuses on the analysis of their text mining components. We explain the relatively slow progress of text mining in gene prioritization, identify several challenges to current text mining methods, and highlight a few directions where more effective text mining algorithms may improve the overall prioritization task and where prioritizing the pathways may be more desirable than prioritizing only genes.
Collapse
Affiliation(s)
- Yuan Luo
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gregory Riedlinger
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Peter Szolovits
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
399
|
Abstract
MOTIVATION Most existing methods for predicting causal disease genes rely on specific type of evidence, and are therefore limited in terms of applicability. More often than not, the type of evidence available for diseases varies-for example, we may know linked genes, keywords associated with the disease obtained by mining text, or co-occurrence of disease symptoms in patients. Similarly, the type of evidence available for genes varies-for example, specific microarray probes convey information only for certain sets of genes. In this article, we apply a novel matrix-completion method called Inductive Matrix Completion to the problem of predicting gene-disease associations; it combines multiple types of evidence (features) for diseases and genes to learn latent factors that explain the observed gene-disease associations. We construct features from different biological sources such as microarray expression data and disease-related textual data. A crucial advantage of the method is that it is inductive; it can be applied to diseases not seen at training time, unlike traditional matrix-completion approaches and network-based inference methods that are transductive. RESULTS Comparison with state-of-the-art methods on diseases from the Online Mendelian Inheritance in Man (OMIM) database shows that the proposed approach is substantially better-it has close to one-in-four chance of recovering a true association in the top 100 predictions, compared to the recently proposed Catapult method (second best) that has <15% chance. We demonstrate that the inductive method is particularly effective for a query disease with no previously known gene associations, and for predicting novel genes, i.e. genes that are previously not linked to diseases. Thus the method is capable of predicting novel genes even for well-characterized diseases. We also validate the novelty of predictions by evaluating the method on recently reported OMIM associations and on associations recently reported in the literature. AVAILABILITY Source code and datasets can be downloaded from http://bigdata.ices.utexas.edu/project/gene-disease.
Collapse
Affiliation(s)
- Nagarajan Natarajan
- Department of Computer Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Inderjit S Dhillon
- Department of Computer Science, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
400
|
Sokolowski M, Wasserman J, Wasserman D. Genome-wide association studies of suicidal behaviors: a review. Eur Neuropsychopharmacol 2014; 24:1567-77. [PMID: 25219938 DOI: 10.1016/j.euroneuro.2014.08.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/24/2014] [Accepted: 08/10/2014] [Indexed: 11/17/2022]
Abstract
Suicidal behaviors represent a fatal dimension of mental ill-health, involving both environmental and heritable (genetic) influences. The putative genetic components of suicidal behaviors have until recent years been mainly investigated by hypothesis-driven research (of "candidate genes"). But technological progress in genotyping has opened the possibilities towards (hypothesis-generating) genomic screens and novel opportunities to explore polygenetic perspectives, now spanning a wide array of possible analyses falling under the term Genome-Wide Association Study (GWAS). Here we introduce and discuss broadly some apparent limitations but also certain developing opportunities of GWAS. We summarize the results from all the eight GWAS conducted up to date focused on suicidality outcomes; treatment emergent suicidal ideation (3 studies), suicide attempts (4 studies) and completed suicides (1 study). Clearly, there are few (if any) genome-wide significant and reproducible findings yet to be demonstrated. We then discuss and pinpoint certain future considerations in relation to sample sizes, the units of genetic associations used, study designs and outcome definitions, psychiatric diagnoses or biological measures, as well as the use of genomic sequencing. We conclude that GWAS should have a lot more potential to show in the case of suicidal outcomes, than what has yet been realized.
Collapse
Affiliation(s)
- Marcus Sokolowski
- National Centre for Suicide Research and Prevention of Mental Ill-Health (NASP), Karolinska Institute (KI), S-171 77 Stockholm, Sweden.
| | - Jerzy Wasserman
- National Centre for Suicide Research and Prevention of Mental Ill-Health (NASP), Karolinska Institute (KI), S-171 77 Stockholm, Sweden
| | - Danuta Wasserman
- National Centre for Suicide Research and Prevention of Mental Ill-Health (NASP), Karolinska Institute (KI), S-171 77 Stockholm, Sweden
| |
Collapse
|