51
|
Dozmorov MG, Cresswell KG, Bacanu SA, Craver C, Reimers M, Kendler KS. A method for estimating coherence of molecular mechanisms in major human disease and traits. BMC Bioinformatics 2020; 21:473. [PMID: 33087046 PMCID: PMC7579960 DOI: 10.1186/s12859-020-03821-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/15/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Phenotypes such as height and intelligence, are thought to be a product of the collective effects of multiple phenotype-associated genes and interactions among their protein products. High/low degree of interactions is suggestive of coherent/random molecular mechanisms, respectively. Comparing the degree of interactions may help to better understand the coherence of phenotype-specific molecular mechanisms and the potential for therapeutic intervention. However, direct comparison of the degree of interactions is difficult due to different sizes and configurations of phenotype-associated gene networks. METHODS We introduce a metric for measuring coherence of molecular-interaction networks as a slope of internal versus external distributions of the degree of interactions. The internal degree distribution is defined by interaction counts within a phenotype-specific gene network, while the external degree distribution counts interactions with other genes in the whole protein-protein interaction (PPI) network. We present a novel method for normalizing the coherence estimates, making them directly comparable. RESULTS Using STRING and BioGrid PPI databases, we compared the coherence of 116 phenotype-associated gene sets from GWAScatalog against size-matched KEGG pathways (the reference for high coherence) and random networks (the lower limit of coherence). We observed a range of coherence estimates for each category of phenotypes. Metabolic traits and diseases were the most coherent, while psychiatric disorders and intelligence-related traits were the least coherent. We demonstrate that coherence and modularity measures capture distinct network properties. CONCLUSIONS We present a general-purpose method for estimating and comparing the coherence of molecular-interaction gene networks that accounts for the network size and shape differences. Our results highlight gaps in our current knowledge of genetics and molecular mechanisms of complex phenotypes and suggest priorities for future GWASs.
Collapse
Affiliation(s)
- Mikhail G. Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA USA
- Department of Pathology, Virginia Commonwealth University, Richmond, VA USA
| | - Kellen G. Cresswell
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA USA
| | - Silviu-Alin Bacanu
- Virginia Institute for Psychiatric and Behavior Genetics and the Department of Psychiatry, Virginia Commonwealth University, Richmond, VA USA
| | - Carl Craver
- Philosophy-Neuroscience-Psychology Program, Washington University in St. Louis, St. Louis, MO USA
| | - Mark Reimers
- Department Physiology, Michigan State University, East Lansing, MI USA
- Department Biomedical Engineering, Michigan State University, East Lansing, MI USA
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavior Genetics and the Department of Psychiatry, Virginia Commonwealth University, Richmond, VA USA
| |
Collapse
|
52
|
Pap ÉM, Farkas K, Széll M, Németh G, Rajan N, Nagy N. Identification of putative phenotype-modifying genetic factors associated with phenotypic diversity in Brooke-Spiegler syndrome. Exp Dermatol 2020; 29:1017-1020. [PMID: 32744342 DOI: 10.1111/exd.14161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/29/2020] [Accepted: 07/27/2020] [Indexed: 01/02/2023]
Abstract
Brooke-Spiegler syndrome (BSS, OMIM 605041) is a rare monogenic skin disease characterized by the development of skin appendage tumors caused by mutations in the cylindromatosis gene. We recently investigated a Hungarian and an Anglo-Saxon pedigrees affected by Brooke-Spiegler syndrome. Despite carrying the same disease-causing mutation (c.2806C>T, p.Arg936X) of the cylindromatosis (CYLD) gene, the affected family members of the two pedigrees exhibit striking differences in their phenotypes. To identify phenotype-modifying genetic factors, whole exome sequencing was performed and the data from the Hungarian and Anglo-Saxon BSS patients were compared. Three putative phenotype-modifying genetic variants were identified: the rs1053023 SNP of the signal transducer and activator of transcription 3 (STAT3) gene, the rs1131877 SNP of the tumor necrosis factor receptor-associated factor 3 (TRAF3) gene and the rs202122812 SNP of the neighbour of BRCA1 gene 1 (NBR1) gene. Our study contributes to the accumulating evidence for the clinical importance of phenotype-modifying genetic factors, which are potentially important for the elucidation of disease prognosis.
Collapse
Affiliation(s)
- Éva Melinda Pap
- Department of Obstetrics and Gynecology, University of Szeged, Szeged, Hungary
| | - Katalin Farkas
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - Márta Széll
- Department of Medical Genetics, University of Szeged, Szeged, Hungary.,Dermatological Research Group of the Hungarian Academy of Sciences, University of Szeged, Szeged, Hungary
| | - Gábor Németh
- Department of Obstetrics and Gynecology, University of Szeged, Szeged, Hungary
| | - Neil Rajan
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Nikoletta Nagy
- Department of Medical Genetics, University of Szeged, Szeged, Hungary.,Dermatological Research Group of the Hungarian Academy of Sciences, University of Szeged, Szeged, Hungary
| |
Collapse
|
53
|
Peng J, Guan J, Hui W, Shang X. A novel subnetwork representation learning method for uncovering disease-disease relationships. Methods 2020; 192:77-84. [PMID: 32946974 DOI: 10.1016/j.ymeth.2020.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/20/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Analyzing disease-disease relationships plays an important role for understanding disease mechanisms and finding alternative uses for a drug. A disease is usually the result of abnormal state of multiple molecular process. Since biological networks can model the interplay of multiple molecular processes, network-based methods have been proposed to uncover the disease-disease relationships recently. Given a disease and a network, the disease could be represented as a subnetwork constructed by the disease genes involved in the given network, named disease subnetwork. Because it is difficult to learn the feature representation of disease subnetworks, most existing methods are unsupervised ones without using labeled information. To fill this gap, we propose a novel method named SubNet2vec to learn the feature vectors of diseases from their corresponding subnetwork in the biological network. By utilizing the feature representation of disease subnetwork, we can analyze disease-disease relationships in a supervised fashion. The evaluation results show that the proposed framework outperforms some state-of-the-art approaches in a large margin on disease-disease/disease-drug association prediction. The source code and data are available athttps://github.com/MedicineBiology-AI/SubNet2vec.git.
Collapse
Affiliation(s)
- Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China.
| | - Jiaojiao Guan
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China.
| | - Weiwei Hui
- Vivo mobile communications (Hang Zhou) co. LTD, China.
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China.
| |
Collapse
|
54
|
Lei X, Tie J, Fujita H. Relational completion based non-negative matrix factorization for predicting metabolite-disease associations. Knowl Based Syst 2020. [DOI: 10.1016/j.knosys.2020.106238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
55
|
Systematic identification of genetic systems associated with phenotypes in patients with rare genomic copy number variations. Hum Genet 2020; 140:457-475. [PMID: 32778951 DOI: 10.1007/s00439-020-02214-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/30/2020] [Indexed: 01/02/2023]
Abstract
Copy number variation (CNV) related disorders tend to show complex phenotypic profiles that do not match known diseases. This makes it difficult to ascertain their underlying molecular basis. A potential solution is to compare the affected genomic regions for multiple patients that share a pathological phenotype, looking for commonalities. Here, we present a novel approach to associate phenotypes with functional systems, in terms of GO categories and KEGG and Reactome pathways, based on patient data. The approach uses genomic and phenomic data from the same patients, finding shared genomic regions between patients with similar phenotypes. These regions are mapped to genes to find associated functional systems. We applied the approach to analyse patients in the DECIPHER database with de novo CNVs, finding functional systems associated with most phenotypes, often due to mutations affecting related genes in the same genomic region. Manual inspection of the ten top-scoring phenotypes found multiple FunSys connections supported by the previous studies for seven of them. The workflow also produces reports focussed on the genes and FunSys connected to the different phenotypes, alongside patient-specific reports, which give details of the associated genes and FunSys for each individual in the cohort. These can be run in "confidential" mode, preserving patient confidentiality. The workflow presented here can be used to associate phenotypes with functional systems using data at the level of a whole cohort of patients, identifying important connections that could not be found when considering them individually. The full workflow is available for download, enabling it to be run on any patient cohort for which phenotypic and CNV data are available.
Collapse
|
56
|
Greene E, Cauble R, Dhamad AE, Kidd MT, Kong B, Howard SM, Castro HF, Campagna SR, Bedford M, Dridi S. Muscle Metabolome Profiles in Woody Breast-(un)Affected Broilers: Effects of Quantum Blue Phytase-Enriched Diet. Front Vet Sci 2020; 7:458. [PMID: 32851035 PMCID: PMC7417653 DOI: 10.3389/fvets.2020.00458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Woody breast (WB) myopathy is significantly impacting modern broilers and is imposing a huge economic burden on the poultry industry worldwide. Yet, its etiology is not fully defined. In a previous study, we have shown that hypoxia and the activation of its upstream mediators (AKT/PI3K/mTOR) played a key role in WB myopathy, and supplementation of quantum blue (QB) can help to reduce WB severity via modulation of hypoxia-related pathways. To gain further insights, we undertook here a metabolomics approach to identify key metabolite signatures and outline their most enriched biological functions. Ultra performance liquid chromatography coupled with high resolution mass spectrometry (UPLC-HRMS) identified a total of 108 known metabolites. Of these, mean intensity differences at P < 0.05 were found in 60 metabolites with 42 higher and 18 lower in WB-affected compared to unaffected muscles. Multivariate analysis and Partial Least Squares Discriminant analysis (PLS-DA) scores plot displayed different clusters when comparing metabolites profile from affected and unaffected tissues and from moderate (MOD) and severe (SEV) WB muscles indicating that unique metabolite profiles are present for the WB-affected and unaffected muscles. To gain biologically related molecule networks, a stringent pathway analyses was conducted using IPA knowledge-base. The top 10 canonical pathways generated, using a fold-change -1.5 and 1.5 cutoff, with the 50 differentially abundant-metabolites were purine nucleotide degradation and de novo biosynthesis, sirtuin signaling pathway, citrulline-nitric oxide cycle, salvage pathways of pyrimidine DNA, IL-1 signaling, iNOS, Angiogenesis, PI3K/AKT signaling, and oxidative phosphorylation. The top altered bio-functions in term of molecular and cellular functions in WB-affected tissues included cellular development, cellular growth and proliferation, cellular death and survival, small molecular biochemistry, inflammatory response, free radical scavenging, cell signaling and cell-to-cell interaction, cell cycles, and lipid, carbohydrate, amino acid, and nucleic acid metabolisms. The top disorder functions identified were organismal injury and abnormalities, cancer, skeletal and muscular disorders, connective tissue disorders, and inflammatory diseases. Breast tissues from birds fed with high dose (2,000 FTU) of QB phytase exhibited 22 metabolites with significantly different levels compared to the control group with a clear cluster using PLS-DA analysis. Of these 22 metabolites, 9 were differentially abundant between WB-affected and unaffected muscles. Taken together, this study determined many metabolic signatures and disordered pathways, which could be regarded as new routes for discovering potential mechanisms of WB myopathy.
Collapse
Affiliation(s)
- Elizabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Reagan Cauble
- Department of Animal Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Ahmed E Dhamad
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Michael T Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Byungwhi Kong
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sara M Howard
- Biological and Small Molecule Mass Spectrometry Core, Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Hector F Castro
- Biological and Small Molecule Mass Spectrometry Core, Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Shawn R Campagna
- Biological and Small Molecule Mass Spectrometry Core, Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | | | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
57
|
Polak D, Sanui T, Nishimura F, Shapira L. Diabetes as a risk factor for periodontal disease-plausible mechanisms. Periodontol 2000 2020; 83:46-58. [PMID: 32385872 DOI: 10.1111/prd.12298] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The present narrative review examines the scientific evidence of the biological mechanisms that may link periodontitis and diabetes, as a source of comorbidity. Publications regarding periodontitis and diabetes, in human, animals, and in vitro were screened for their relevance. Periodontal microbiome studies indicate a possible association between altered glucose metabolism in prediabetes and diabetes and changes in the periodontal microbiome. Coinciding with this, hyperglycemia enhances expression of pathogen receptors, which enhance host response to the dysbiotic microbiome. Hyperglycemia also promotes pro-inflammatory response independently or via the advanced glycation end product/receptor for advanced glycation end product pathway. These processes excite cellular tissue destruction functions, which further enhance pro-inflammatory cytokines expression and alteration in the RANKL/osteoprotegerin ratio, promoting formation and activation of osteoclasts. The evidence supports the role of several pathogenic mechanisms in the path of true causal comorbidity between poorly controlled diabetes and periodontitis. However, further research is needed to better understand these mechanisms and to explore other mechanisms.
Collapse
Affiliation(s)
- David Polak
- Department of Periodontology, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Terukazu Sanui
- Section of Periodontology, Division of Oral Rehabilitation, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| | - Fusanori Nishimura
- Section of Periodontology, Division of Oral Rehabilitation, Kyushu University Faculty of Dental Science, Fukuoka, Japan
| | - Lior Shapira
- Department of Periodontology, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| |
Collapse
|
58
|
Leier HC, Weinstein JB, Kyle JE, Lee JY, Bramer LM, Stratton KG, Kempthorne D, Navratil AR, Tafesse EG, Hornemann T, Messer WB, Dennis EA, Metz TO, Barklis E, Tafesse FG. A global lipid map defines a network essential for Zika virus replication. Nat Commun 2020; 11:3652. [PMID: 32694525 PMCID: PMC7374707 DOI: 10.1038/s41467-020-17433-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV), an arbovirus of global concern, remodels intracellular membranes to form replication sites. How ZIKV dysregulates lipid networks to allow this, and consequences for disease, is poorly understood. Here, we perform comprehensive lipidomics to create a lipid network map during ZIKV infection. We find that ZIKV significantly alters host lipid composition, with the most striking changes seen within subclasses of sphingolipids. Ectopic expression of ZIKV NS4B protein results in similar changes, demonstrating a role for NS4B in modulating sphingolipid pathways. Disruption of sphingolipid biosynthesis in various cell types, including human neural progenitor cells, blocks ZIKV infection. Additionally, the sphingolipid ceramide redistributes to ZIKV replication sites, and increasing ceramide levels by multiple pathways sensitizes cells to ZIKV infection. Thus, we identify a sphingolipid metabolic network with a critical role in ZIKV replication and show that ceramide flux is a key mediator of ZIKV infection.
Collapse
Affiliation(s)
- Hans C Leier
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA
| | - Jules B Weinstein
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Joon-Yong Lee
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Lisa M Bramer
- Computing and Analytics Division, National Security Directorate, PNNL, Richland, WA, 99352, USA
| | - Kelly G Stratton
- Computing and Analytics Division, National Security Directorate, PNNL, Richland, WA, 99352, USA
| | - Douglas Kempthorne
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA
- Center for Diversity and Inclusion, OHSU, Portland, OR, 97239, USA
| | - Aaron R Navratil
- Departments of Chemistry & Biochemistry and Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Endale G Tafesse
- Department of Plant Sciences, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK, S7N 5A8, Canada
| | - Thorsten Hornemann
- University Zurich and University Hospital Zurich, University of Zurich, Zurich, 8091, Switzerland
| | - William B Messer
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA
- Department of Medicine, Division of Infectious Diseases, OHSU, Portland, Oregon, 97239, USA
| | - Edward A Dennis
- Departments of Chemistry & Biochemistry and Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Thomas O Metz
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Eric Barklis
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA
| | - Fikadu G Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA.
| |
Collapse
|
59
|
Hwang S, Lee T, Yoon Y. Exploring disease comorbidity in a module-module interaction network. J Bioinform Comput Biol 2020; 18:2050010. [PMID: 32404015 DOI: 10.1142/s0219720020500109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Understanding disease comorbidity contributes to improved quality of life in patients who are suffering from multiple diseases. Therefore, to better explore comorbid diseases, the clarification of associations between diseases based on biological functions is essential. In our study, we propose a method for identifying disease comorbidity in a module-based network, named the module-module interaction (MMI) network, which represents how biological functions influence each other. To construct the MMI network, we detected gene modules - sets of genes that have a higher probability of taking part in specific functions - and established a link between these modules. Subsequently, we constructed disease-related networks in the MMI network to understand inherent disease mechanisms and calculated comorbidity scores of disease pairs using Gene Ontology (GO) terms. Our results show that we can obtain further information on disease mechanisms by considering interactions between functional modules instead of between genes. In addition, we verified that predicted comorbid relationships of disease pairs based on the MMI network are more significant than those based on the protein-protein interaction (PPI) network. This study can be useful to elucidate the mechanisms underlying comorbidities for further study, which will provide a broader insight into the pathogenesis of diseases.
Collapse
Affiliation(s)
- Soyoun Hwang
- Department of IT Convergence Engineering, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Taekeon Lee
- Department of Computer Engineering, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Youngmi Yoon
- Department of Computer Engineering, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do, Korea
| |
Collapse
|
60
|
Gaudelet T, Malod-Dognin N, Sánchez-Valle J, Pancaldi V, Valencia A, Pržulj N. Unveiling new disease, pathway, and gene associations via multi-scale neural network. PLoS One 2020; 15:e0231059. [PMID: 32251458 PMCID: PMC7135208 DOI: 10.1371/journal.pone.0231059] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/14/2020] [Indexed: 12/16/2022] Open
Abstract
Diseases involve complex modifications to the cellular machinery. The gene expression profile of the affected cells contains characteristic patterns linked to a disease. Hence, new biological knowledge about a disease can be extracted from these profiles, improving our ability to diagnose and assess disease risks. This knowledge can be used for drug re-purposing, or by physicians to evaluate a patient’s condition and co-morbidity risk. Here, we consider differential gene expressions obtained by microarray technology for patients diagnosed with various diseases. Based on these data and cellular multi-scale organization, we aim at uncovering disease–disease, disease–gene and disease–pathway associations. We propose a neural network with structure based on the multi-scale organization of proteins in a cell into biological pathways. We show that this model is able to correctly predict the diagnosis for the majority of patients. Through the analysis of the trained model, we predict disease–disease, disease–pathway, and disease–gene associations and validate the predictions by comparisons to known interactions and literature search, proposing putative explanations for the predictions.
Collapse
Affiliation(s)
- Thomas Gaudelet
- Department of Computer Science, University College London, London, United Kingdom
| | | | | | - Vera Pancaldi
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 Inserm, ERL5294 CNRS, 31037, Toulouse, France
- University Paul Sabatier III, Toulouse, France
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- ICREA, Pg. Lluis Companys, Barcelona, Spain
| | - Nataša Pržulj
- Department of Computer Science, University College London, London, United Kingdom
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- ICREA, Pg. Lluis Companys, Barcelona, Spain
- * E-mail:
| |
Collapse
|
61
|
Pap ÉM, Farkas K, Tóth L, Fábos B, Széll M, Németh G, Nagy N. Identification of putative genetic modifying factors that influence the development of Papillon-Lefévre or Haim-Munk syndrome phenotypes. Clin Exp Dermatol 2020; 45:555-559. [PMID: 31925812 DOI: 10.1111/ced.14171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Papillon-Lefévre syndrome (PLS; OMIM 245000) and Haim-Munk syndrome (HMS; OMIM 245010), which are both characterized by palmoplantar hyperkeratosis and periodontitis, are phenotypic variants of the same disease caused by mutations of the cathepsin C (CTSC) gene. AIM To identify putative genetic modifying factors responsible for the differential development of the PLS or HMS phenotypes, we investigated two Hungarian patients with different phenotypic variants (PLS and HMS) but carrying the same homozygous nonsense CTSC mutation (c.748C/T; p.Arg250X). METHODS To gain insights into phenotype-modifying associations, whole exome sequencing (WES) was performed for both patients, and the results were compared to identify potentially relevant genetic modifying factors. RESULTS WES revealed two putative phenotype-modifying variants: (i) a missense mutation (rs34608771) of the SH2 domain containing 4A (SH2D4A) gene encoding an adaptor protein involved in intracellular signalling of cystatin F, a known inhibitor of the cathepsin protein, and (ii) a missense variant (rs55695858) of the odorant binding protein 2A (OBP2A) gene, influencing the function of the cathepsin protein through the glycosyltransferase 6 domain containing 1 (GLT6D1) protein. CONCLUSION Our study contributes to the accumulating evidence supporting the clinical importance of phenotype-modifying genetic factors, which have high potential to aid the elucidation of genotype-phenotype correlations and disease prognosis.
Collapse
Affiliation(s)
- É M Pap
- Department of Obstetrics and Gynecology Szeged, University of Szeged, Szeged, Hungary
| | - K Farkas
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - L Tóth
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - B Fábos
- Mór Kaposi Teaching Hospital, Kaposvár, Hungary
| | - M Széll
- Department of Medical Genetics, University of Szeged, Szeged, Hungary.,Dermatological Research Group of the Hungarian Academy of Sciences, University of Szeged, Szeged, Hungary
| | - G Németh
- Department of Obstetrics and Gynecology Szeged, University of Szeged, Szeged, Hungary
| | - N Nagy
- Department of Medical Genetics, University of Szeged, Szeged, Hungary.,Dermatological Research Group of the Hungarian Academy of Sciences, University of Szeged, Szeged, Hungary
| |
Collapse
|
62
|
Pham M, Wilson S, Govindarajan H, Lin CH, Lichtarge O. Discovery of disease- and drug-specific pathways through community structures of a literature network. Bioinformatics 2020; 36:1881-1888. [PMID: 31738408 PMCID: PMC7103064 DOI: 10.1093/bioinformatics/btz857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/29/2019] [Accepted: 11/15/2019] [Indexed: 01/18/2023] Open
Abstract
Motivation In light of the massive growth of the scientific literature, text mining is increasingly used to extract biological pathways. Though multiple tools explore individual connections between genes, diseases and drugs, few extensively synthesize pathways for specific diseases and drugs. Results Through community detection of a literature network, we extracted 3444 functional gene groups that represented biological pathways for specific diseases and drugs. The network linked Medical Subject Headings (MeSH) terms of genes, diseases and drugs that co-occurred in publications. The resulting communities detected highly associated genes, diseases and drugs. These significantly matched current knowledge of biological pathways and predicted future ones in time-stamped experiments. Likewise, disease- and drug-specific communities also recapitulated known pathways for those given diseases and drugs. Moreover, diseases sharing communities had high comorbidity with each other and drugs sharing communities had many common side effects, consistent with related mechanisms. Indeed, the communities robustly recovered mutual targets for drugs [area under Receiver Operating Characteristic curve (AUROC)=0.75] and shared pathogenic genes for diseases (AUROC=0.82). These data show that literature communities inform not only just known biological processes but also suggest novel disease- and drug-specific mechanisms that may guide disease gene discovery and drug repurposing. Availability and implementation Application tools are available at http://meteor.lichtargelab.org. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Minh Pham
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen Wilson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Harikumar Govindarajan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chih-Hsu Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
63
|
Akram P, Liao L. Prediction of comorbid diseases using weighted geometric embedding of human interactome. BMC Med Genomics 2019; 12:161. [PMID: 31888634 PMCID: PMC6936100 DOI: 10.1186/s12920-019-0605-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/16/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Comorbidity is the phenomenon of two or more diseases occurring simultaneously not by random chance and presents great challenges to accurate diagnosis and treatment. As an effort toward better understanding the genetic causes of comorbidity, in this work, we have developed a computational method to predict comorbid diseases. Two diseases sharing common genes tend to increase their comorbidity. Previous work shows that after mapping the associated genes onto the human interactome the distance between the two disease modules (subgraphs) is correlated with comorbidity. METHODS To fully incorporate structural characteristics of interactome as features into prediction of comorbidity, our method embeds the human interactome into a high dimensional geometric space with weights assigned to the network edges and uses the projection onto different dimension to "fingerprint" disease modules. A supervised machine learning classifier is then trained to discriminate comorbid diseases versus non-comorbid diseases. RESULTS In cross-validation using a benchmark dataset of more than 10,000 disease pairs, we report that our model achieves remarkable performance of ROC score = 0.90 for comorbidity threshold at relative risk RR = 0 and 0.76 for comorbidity threshold at RR = 1, and significantly outperforms the previous method and the interactome generated by annotated data. To further incorporate prior knowledge pathways association with diseases, we weight the protein-protein interaction network edges according to their frequency of occurring in those pathways in such a way that edges with higher frequency will more likely be selected in the minimum spanning tree for geometric embedding. Such weighted embedding is shown to lead to further improvement of comorbid disease prediction. CONCLUSION The work demonstrates that embedding the two-dimension planar graph of human interactome into a high dimensional geometric space allows for characterizing and capturing disease modules (subgraphs formed by the disease associated genes) from multiple perspectives, and hence provides enriched features for a supervised classifier to discriminate comorbid disease pairs from non-comorbid disease pairs more accurately than based on simply the module separation.
Collapse
Affiliation(s)
- Pakeeza Akram
- School of Electrical Engineering and Computer Science (SEECS), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
- Department of Computer Science, University of Delaware, Newark, USA
| | - Li Liao
- Department of Computer Science, University of Delaware, Newark, USA
| |
Collapse
|
64
|
Abstract
BACKGROUND A collection of disease-associated data contributes to study the association between diseases. Discovering closely related diseases plays a crucial role in revealing their common pathogenic mechanisms. This might further imply treatment that can be appropriated from one disease to another. During the past decades, a number of approaches for calculating disease similarity have been developed. However, most of them are designed to take advantage of single or few data sources, which results in their low accuracy. METHODS In this paper, we propose a novel method, called MultiSourcDSim, to calculate disease similarity by integrating multiple data sources, namely, gene-disease associations, GO biological process-disease associations and symptom-disease associations. Firstly, we establish three disease similarity networks according to the three disease-related data sources respectively. Secondly, the representation of each node is obtained by integrating the three small disease similarity networks. In the end, the learned representations are applied to calculate the similarity between diseases. RESULTS Our approach shows the best performance compared to the other three popular methods. Besides, the similarity network built by MultiSourcDSim suggests that our method can also uncover the latent relationships between diseases. CONCLUSIONS MultiSourcDSim is an efficient approach to predict similarity between diseases.
Collapse
Affiliation(s)
- Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha, 410075 China
| | - Danyi Ye
- School of Computer Science and Engineering, Central South University, Changsha, 410075 China
| | - Junmin Zhao
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan, 467000 China
| | - Jingpu Zhang
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan, 467000 China
| |
Collapse
|
65
|
Wang Y, Juan L, Peng J, Zang T, Wang Y. Prioritizing candidate diseases-related metabolites based on literature and functional similarity. BMC Bioinformatics 2019; 20:574. [PMID: 31760947 PMCID: PMC6876110 DOI: 10.1186/s12859-019-3127-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background As the terminal products of cellular regulatory process, functional related metabolites have a close relationship with complex diseases, and are often associated with the same or similar diseases. Therefore, identification of disease related metabolites play a critical role in understanding comprehensively pathogenesis of disease, aiming at improving the clinical medicine. Considering that a large number of metabolic markers of diseases need to be explored, we propose a computational model to identify potential disease-related metabolites based on functional relationships and scores of referred literatures between metabolites. First, obtaining associations between metabolites and diseases from the Human Metabolome database, we calculate the similarities of metabolites based on modified recommendation strategy of collaborative filtering utilizing the similarities between diseases. Next, a disease-associated metabolite network (DMN) is built with similarities between metabolites as weight. To improve the ability of identifying disease-related metabolites, we introduce scores of text mining from the existing database of chemicals and proteins into DMN and build a new disease-associated metabolite network (FLDMN) by fusing functional associations and scores of literatures. Finally, we utilize random walking with restart (RWR) in this network to predict candidate metabolites related to diseases. Results We construct the disease-associated metabolite network and its improved network (FLDMN) with 245 diseases, 587 metabolites and 28,715 disease-metabolite associations. Subsequently, we extract training sets and testing sets from two different versions of the Human Metabolome database and assess the performance of DMN and FLDMN on 19 diseases, respectively. As a result, the average AUC (area under the receiver operating characteristic curve) of DMN is 64.35%. As a further improved network, FLDMN is proven to be successful in predicting potential metabolic signatures for 19 diseases with an average AUC value of 76.03%. Conclusion In this paper, a computational model is proposed for exploring metabolite-disease pairs and has good performance in predicting potential metabolites related to diseases through adequate validation. This result suggests that integrating literature and functional associations can be an effective way to construct disease associated metabolite network for prioritizing candidate diseases-related metabolites.
Collapse
Affiliation(s)
- Yongtian Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, People's Republic of China
| | - Liran Juan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, People's Republic of China
| | - Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, People's Republic of China
| | - Tianyi Zang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, People's Republic of China.
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, People's Republic of China.
| |
Collapse
|
66
|
Chen X, Shi W, Deng L. Prediction of Disease Comorbidity Using HeteSim Scores based on Multiple Heterogeneous Networks. Curr Gene Ther 2019; 19:232-241. [DOI: 10.2174/1566523219666190917155959] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/14/2019] [Accepted: 06/16/2019] [Indexed: 12/25/2022]
Abstract
Background:
Accumulating experimental studies have indicated that disease comorbidity
causes additional pain to patients and leads to the failure of standard treatments compared to patients
who have a single disease. Therefore, accurate prediction of potential comorbidity is essential to design
more efficient treatment strategies. However, only a few disease comorbidities have been discovered
in the clinic.
Objective:
In this work, we propose PCHS, an effective computational method for predicting disease
comorbidity.
Materials and Methods:
We utilized the HeteSim measure to calculate the relatedness score for different
disease pairs in the global heterogeneous network, which integrates six networks based on biological
information, including disease-disease associations, drug-drug interactions, protein-protein interactions
and associations among them. We built the prediction model using the Support Vector Machine
(SVM) based on the HeteSim scores.
Results and Conclusion:
The results showed that PCHS performed significantly better than previous
state-of-the-art approaches and achieved an AUC score of 0.90 in 10-fold cross-validation. Furthermore,
some of our predictions have been verified in literatures, indicating the effectiveness of our method.
Collapse
Affiliation(s)
- Xuegong Chen
- School of Computer Science and Engineering, Central South University, Changsha, 410075, China
| | - Wanwan Shi
- School of Computer Science and Engineering, Central South University, Changsha, 410075, China
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha, 410075, China
| |
Collapse
|
67
|
Lei X, Tie J. Prediction of disease-related metabolites using bi-random walks. PLoS One 2019; 14:e0225380. [PMID: 31730648 PMCID: PMC6857945 DOI: 10.1371/journal.pone.0225380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 11/04/2019] [Indexed: 12/25/2022] Open
Abstract
Metabolites play a significant role in various complex human disease. The exploration of the relationship between metabolites and diseases can help us to better understand the underlying pathogenesis. Several network-based methods have been used to predict the association between metabolite and disease. However, some methods ignored hierarchical differences in disease network and failed to work in the absence of known metabolite-disease associations. This paper presents a bi-random walks based method for disease-related metabolites prediction, called MDBIRW. First of all, we reconstruct the disease similarity network and metabolite functional similarity network by integrating Gaussian Interaction Profile (GIP) kernel similarity of diseases and GIP kernel similarity of metabolites, respectively. Then, the bi-random walks algorithm is executed on the reconstructed disease similarity network and metabolite functional similarity network to predict potential disease-metabolite associations. At last, MDBIRW achieves reliable performance in leave-one-out cross validation (AUC of 0.910) and 5-fold cross validation (AUC of 0.924). The experimental results show that our method outperforms other existing methods for predicting disease-related metabolites.
Collapse
Affiliation(s)
- Xiujuan Lei
- School of Computer Science, Shaanxi Normal University, Xi’an China
| | - Jiaojiao Tie
- School of Computer Science, Shaanxi Normal University, Xi’an China
| |
Collapse
|
68
|
Nam Y, Lee DG, Bang S, Kim JH, Kim JH, Shin H. The translational network for metabolic disease - from protein interaction to disease co-occurrence. BMC Bioinformatics 2019; 20:576. [PMID: 31722666 PMCID: PMC6854734 DOI: 10.1186/s12859-019-3106-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/20/2019] [Indexed: 02/08/2023] Open
Abstract
Background The recent advances in human disease network have provided insights into establishing the relationships between the genotypes and phenotypes of diseases. In spite of the great progress, it yet remains as only a map of topologies between diseases, but not being able to be a pragmatic diagnostic/prognostic tool in medicine. It can further evolve from a map to a translational tool if it equips with a function of scoring that measures the likelihoods of the association between diseases. Then, a physician, when practicing on a patient, can suggest several diseases that are highly likely to co-occur with a primary disease according to the scores. In this study, we propose a method of implementing ‘n-of-1 utility’ (n potential diseases of one patient) to human disease network—the translational disease network. Results We first construct a disease network by introducing the notion of walk in graph theory to protein-protein interaction network, and then provide a scoring algorithm quantifying the likelihoods of disease co-occurrence given a primary disease. Metabolic diseases, that are highly prevalent but have found only a few associations in previous studies, are chosen as entries of the network. Conclusions The proposed method substantially increased connectivity between metabolic diseases and provided scores of co-occurring diseases. The increase in connectivity turned the disease network info-richer. The result lifted the AUC of random guessing up to 0.72 and appeared to be concordant with the existing literatures on disease comorbidity.
Collapse
Affiliation(s)
- Yonghyun Nam
- Department of Industrial Engineering, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Dong-Gi Lee
- Department of Industrial Engineering, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Sunjoo Bang
- Department of Industrial Engineering, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Ju Han Kim
- Seoul National University Biomedical Informatics (SNUBI), Division of Biomedical Informatics, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jae-Hoon Kim
- Department of Industrial Engineering, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea.
| | - Hyunjung Shin
- Department of Industrial Engineering, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea.
| |
Collapse
|
69
|
Zolotareva O, Saik OV, Königs C, Bragina EY, Goncharova IA, Freidin MB, Dosenko VE, Ivanisenko VA, Hofestädt R. Comorbidity of asthma and hypertension may be mediated by shared genetic dysregulation and drug side effects. Sci Rep 2019; 9:16302. [PMID: 31705029 PMCID: PMC6841742 DOI: 10.1038/s41598-019-52762-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Asthma and hypertension are complex diseases coinciding more frequently than expected by chance. Unraveling the mechanisms of comorbidity of asthma and hypertension is necessary for choosing the most appropriate treatment plan for patients with this comorbidity. Since both diseases have a strong genetic component in this article we aimed to find and study genes simultaneously associated with asthma and hypertension. We identified 330 shared genes and found that they form six modules on the interaction network. A strong overlap between genes associated with asthma and hypertension was found on the level of eQTL regulated genes and between targets of drugs relevant for asthma and hypertension. This suggests that the phenomenon of comorbidity of asthma and hypertension may be explained by altered genetic regulation or result from drug side effects. In this work we also demonstrate that not only drug indications but also contraindications provide an important source of molecular evidence helpful to uncover disease mechanisms. These findings give a clue to the possible mechanisms of comorbidity and highlight the direction for future research.
Collapse
Affiliation(s)
- Olga Zolotareva
- Bielefeld University, International Research Training Group "Computational Methods for the Analysis of the Diversity and Dynamics of Genomes" and Genome Informatics, Faculty of Technology and Center for Biotechnology, Bielefeld, Germany.
| | - Olga V Saik
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Cassandra Königs
- Bielefeld University, Bioinformatics and Medical Informatics Department, Bielefeld, Germany
| | - Elena Yu Bragina
- Research Institute of Medical Genetics, Tomsk NRMC, Tomsk, Russia
| | | | - Maxim B Freidin
- Research Institute of Medical Genetics, Tomsk NRMC, Tomsk, Russia
| | | | - Vladimir A Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Ralf Hofestädt
- Bielefeld University, Bioinformatics and Medical Informatics Department, Bielefeld, Germany
| |
Collapse
|
70
|
Aguilar D, Lemonnier N, Koppelman GH, Melén E, Oliva B, Pinart M, Guerra S, Bousquet J, Anto JM. Understanding allergic multimorbidity within the non-eosinophilic interactome. PLoS One 2019; 14:e0224448. [PMID: 31693680 PMCID: PMC6834334 DOI: 10.1371/journal.pone.0224448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The mechanisms explaining multimorbidity between asthma, dermatitis and rhinitis (allergic multimorbidity) are not well known. We investigated these mechanisms and their specificity in distinct cell types by means of an interactome-based analysis of expression data. METHODS Genes associated to the diseases were identified using data mining approaches, and their multimorbidity mechanisms in distinct cell types were characterized by means of an in silico analysis of the topology of the human interactome. RESULTS We characterized specific pathomechanisms for multimorbidities between asthma, dermatitis and rhinitis for distinct emergent non-eosinophilic cell types. We observed differential roles for cytokine signaling, TLR-mediated signaling and metabolic pathways for multimorbidities across distinct cell types. Furthermore, we also identified individual genes potentially associated to multimorbidity mechanisms. CONCLUSIONS Our results support the existence of differentiated multimorbidity mechanisms between asthma, dermatitis and rhinitis at cell type level, as well as mechanisms common to distinct cell types. These results will help understanding the biology underlying allergic multimorbidity, assisting in the design of new clinical studies.
Collapse
MESH Headings
- Asthma/epidemiology
- Asthma/genetics
- Asthma/immunology
- Blood Cells/immunology
- Blood Cells/metabolism
- Cytokines/immunology
- Cytokines/metabolism
- Datasets as Topic
- Dermatitis, Allergic Contact/epidemiology
- Dermatitis, Allergic Contact/genetics
- Dermatitis, Allergic Contact/immunology
- Dermatitis, Atopic/epidemiology
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/immunology
- Gene Expression Profiling
- Humans
- Immunity, Cellular/genetics
- Multimorbidity
- Protein Interaction Maps/genetics
- Protein Interaction Maps/immunology
- Rhinitis, Allergic/epidemiology
- Rhinitis, Allergic/genetics
- Rhinitis, Allergic/immunology
Collapse
Affiliation(s)
- Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Barcelona, Spain
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- 6AM Data Mining, Barcelona, Spain
| | - Nathanael Lemonnier
- Institute for Advanced Biosciences, Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes, Site Santé, Allée des Alpes, La Tronche, France
| | - Gerard H. Koppelman
- University of Groningen, University Medical Center Groningen, Beatrix Children’s Hospital, Department of Pediatric Pulmonology and Pediatric Allergology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Baldo Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mariona Pinart
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Stefano Guerra
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, United States of America
| | - Jean Bousquet
- Hopital Arnaud de Villeneuve University Hospital, Montpellier, France
- Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Comprehensive Allergy Center, Department of Dermatology and Allergy, Berlin, Germany
| | - Josep M. Anto
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| |
Collapse
|
71
|
Ahmed Z, Zeeshan S, Xiong R, Liang BT. Debutant iOS app and gene-disease complexities in clinical genomics and precision medicine. Clin Transl Med 2019; 8:26. [PMID: 31586224 PMCID: PMC6778157 DOI: 10.1186/s40169-019-0243-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background The last decade has seen a dramatic increase in the availability of scientific data, where human-related biological databases have grown not only in count but also in volume, posing unprecedented challenges in data storage, processing, analysis, exchange, and curation. Next generation sequencing (NGS) advancements have facilitated and accelerated the process of identifying genetic variations. Adopting NGS with Whole-Genome and RNA sequencing in a diagnostic context has the potential to improve disease-risk detection in support of precision medicine and drug discovery. Several bioinformatics pipelines have been developed to strengthen variant interpretation by efficiently processing and analyzing sequence data, whereas many published results show how genomics data can be proactively incorporated into medical practices and improve utilization of clinical information. To utilize the wealth of genomics and health, there is a crucial need to generate appropriate gene-disease annotation repositories accessed through modern technology. Results Our focus here is to create a comprehensive database with mobile access to actionable genes and classified diseases, considered the foundation for clinical genomics and precision medicine. We present a publicly available iOS app, PAS-Gen, which invites global users to freely download it on iPhone and iPad devices, quickly adopt its easy to use interface, and search for genes and related diseases. PAS-Gen was developed using Swift, XCODE, and PHP scripting that uses Web and MySQL database servers, which includes over 59,000 protein-coding and non-coding genes, and over 90,000 classified gene-disease associations. PAS-Gen is founded on the clinical and scientific premise that easier healthcare and genomics data sharing will accelerate future medical discoveries. Conclusions We present a cutting-edge gene-disease database with a smart phone application, integrating information on classified diseases and related genes. The PAS-Gen app will assist researchers, medical practitioners, and pharmacists by providing a broad and view of genes that may be implicated in the likelihood of developing certain diseases. This tool with accelerate users’ abilities to understand the genetic basis of human complex diseases and by assimilating genomic and phenotypic data will support future work to identify gene-specific designer drugs, target precise molecular fingerprints for tumors, suggest appropriate drug therapies, predict individual susceptibility to disease, and diagnose and treat rare illnesses.
Collapse
Affiliation(s)
- Zeeshan Ahmed
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center (UConn Health), 263 Farmington Ave, Farmington, CT, 06032, USA. .,Institute for Systems Genomics, University of Connecticut, 263 Farmington Ave, Farmington, CT, 06032, USA.
| | - Saman Zeeshan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Ruoyun Xiong
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center (UConn Health), 263 Farmington Ave, Farmington, CT, 06032, USA.,The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Bruce T Liang
- Pat and Jim Calhoun Cardiology Center, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06032, USA
| |
Collapse
|
72
|
Cheng L, Zhao H, Wang P, Zhou W, Luo M, Li T, Han J, Liu S, Jiang Q. Computational Methods for Identifying Similar Diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:590-604. [PMID: 31678735 PMCID: PMC6838934 DOI: 10.1016/j.omtn.2019.09.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/01/2023]
Abstract
Although our knowledge of human diseases has increased dramatically, the molecular basis, phenotypic traits, and therapeutic targets of most diseases still remain unclear. An increasing number of studies have observed that similar diseases often are caused by similar molecules, can be diagnosed by similar markers or phenotypes, or can be cured by similar drugs. Thus, the identification of diseases similar to known ones has attracted considerable attention worldwide. To this end, the associations between diseases at the molecular, phenotypic, and taxonomic levels were used to measure the pairwise similarity in diseases. The corresponding performance assessment strategies for these methods involving the terms “category-based,” “simulated-patient-based,” and “benchmark-data-based” were thus further emphasized. Then, frequently used methods were evaluated using a benchmark-data-based strategy. To facilitate the assessment of disease similarity scores, researchers have designed dozens of tools that implement these methods for calculating disease similarity. Currently, disease similarity has been advantageous in predicting noncoding RNA (ncRNA) function and therapeutic drugs for diseases. In this article, we review disease similarity methods, evaluation strategies, tools, and their applications in the biomedical community. We further evaluate the performance of these methods and discuss the current limitations and future trends for calculating disease similarity.
Collapse
Affiliation(s)
- Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Hengqiang Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Pingping Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Wenyang Zhou
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Meng Luo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Tianxin Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China.
| | - Shulin Liu
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China.
| |
Collapse
|
73
|
Chagoyen M, Ranea JAG, Pazos F. Applications of molecular networks in biomedicine. Biol Methods Protoc 2019; 4:bpz012. [PMID: 32395629 PMCID: PMC7200821 DOI: 10.1093/biomethods/bpz012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/20/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Due to the large interdependence between the molecular components of living systems, many phenomena, including those related to pathologies, cannot be explained in terms of a single gene or a small number of genes. Molecular networks, representing different types of relationships between molecular entities, embody these large sets of interdependences in a framework that allow their mining from a systemic point of view to obtain information. These networks, often generated from high-throughput omics datasets, are used to study the complex phenomena of human pathologies from a systemic point of view. Complementing the reductionist approach of molecular biology, based on the detailed study of a small number of genes, systemic approaches to human diseases consider that these are better reflected in large and intricate networks of relationships between genes. These networks, and not the single genes, provide both better markers for diagnosing diseases and targets for treating them. Network approaches are being used to gain insight into the molecular basis of complex diseases and interpret the large datasets associated with them, such as genomic variants. Network formalism is also suitable for integrating large, heterogeneous and multilevel datasets associated with diseases from the molecular level to organismal and epidemiological scales. Many of these approaches are available to nonexpert users through standard software packages.
Collapse
Affiliation(s)
- Monica Chagoyen
- Computational Systems Biology Group, Systems Biology Program, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Juan A G Ranea
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Florencio Pazos
- Computational Systems Biology Group, Systems Biology Program, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| |
Collapse
|
74
|
Suls J, Green PA, Boyd CM. Multimorbidity: Implications and directions for health psychology and behavioral medicine. Health Psychol 2019; 38:772-782. [PMID: 31436463 PMCID: PMC6750244 DOI: 10.1037/hea0000762] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The increasing prevalence of multimorbidity in the United States and the rest of the world poses problems for patients and for health care providers, care systems, and policy. After clarifying the difference between comorbidity and multimorbidity, this article describes the challenges that the prevalence of multimorbidity presents for well-being, prevention, and medical treatment. We submit that health psychology and behavioral medicine have an important role to play in meeting these challenges because of the holistic vision of health afforded by the foundational biopsychosocial model. Furthermore, opportunities abound for health psychology/behavioral medicine to study how biological, social and psychological factors influence multimorbidity. This article describes three major areas in which health psychologists can contribute to understanding and treatment of multimorbidity: (a) etiology; (b) prevention and self-management; and (c) clinical care. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
- Jerry Suls
- Behavioral Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute
| | - Paige A Green
- Basic Biobehavioral and Psychological Sciences Branch, Behavioral Research Program, National Cancer Institute
| | - Cynthia M Boyd
- Cynthia M. Boyd, School of Medicine, Johns Hopkins University
| |
Collapse
|
75
|
Abstract
OBJECTIVE Multimorbidity is a robust predictor of disability in aging adults, but the mechanisms by which multimorbidity is disabling are not clear. Most existing research focuses on disease-specific phenomena, such as diminished lung capacity in chronic obstructive pulmonary disease, which can result in functional limitations. This review takes a different approach by highlighting the potential role of a biological process-inflammation-that is common to many chronic medical conditions and thus, from a medical perspective, relatively disease nonspecific. METHOD Beginning with a description of inflammation and its measurement, this paper will provide an overview of research on inflammation as a predictor of disease risk in healthy adults and of adverse outcomes (e.g., disability) in those with multimorbidity. RESULTS The discussion of inflammation is then situated in the context of biopsychosocial influences on health, as inflammation has been shown to be sensitive to a wide range of social and psychological processes that are thought to contribute to healthy aging, including successful adaptation to multimorbidity and reduced risk of disability. CONCLUSIONS Finally, implications of this broader perspective for interventions to improve outcomes in aging adults with multimorbidity are briefly considered. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
|
76
|
McCabe ERB. Metabolite flux: A dynamic concept for inherited metabolic disorders as complex traits. Mol Genet Metab 2019; 128:14-18. [PMID: 31331738 DOI: 10.1016/j.ymgme.2019.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/06/2019] [Accepted: 07/16/2019] [Indexed: 11/20/2022]
Abstract
In 2000 and 2001, we described factors that lead to the phenotypes of individuals with "simple," "single" gene disorders, like inherited metabolic disorders, being complex, multi-genic traits. These factors include functional thresholds, genetic and environmental modifiers, and systems dynamics, encompassing metabolic control analysis and scale-free, hub-and-spoke networks. This mini-review will consider topics influencing complexity developed in the ensuing nearly two decades, such as "synergistic heterozygosity" and "moonlighting proteins." There will be a focus on the value of the measurement of flux in evaluating the metabolome to ascertain phenotypic variability and the potential role of the gut microbiome in metabolomic flux. A point-of-care metabolomics tool, under development to improve the real time, inter-operative ascertainment of tumor margins and similar devices, will provide opportunities to improve acute care and ongoing management of individuals with inherited metabolic disorders.
Collapse
Affiliation(s)
- Edward R B McCabe
- Department of Pediatrics, UCLA School of Medicine, Mattel Children's Hospital UCLA, USA.
| |
Collapse
|
77
|
Zitnik M, Agrawal M, Leskovec J. Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics 2019; 34:i457-i466. [PMID: 29949996 PMCID: PMC6022705 DOI: 10.1093/bioinformatics/bty294] [Citation(s) in RCA: 422] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Motivation The use of drug combinations, termed polypharmacy, is common to treat patients with complex diseases or co-existing conditions. However, a major consequence of polypharmacy is a much higher risk of adverse side effects for the patient. Polypharmacy side effects emerge because of drug-drug interactions, in which activity of one drug may change, favorably or unfavorably, if taken with another drug. The knowledge of drug interactions is often limited because these complex relationships are rare, and are usually not observed in relatively small clinical testing. Discovering polypharmacy side effects thus remains an important challenge with significant implications for patient mortality and morbidity. Results Here, we present Decagon, an approach for modeling polypharmacy side effects. The approach constructs a multimodal graph of protein-protein interactions, drug-protein target interactions and the polypharmacy side effects, which are represented as drug-drug interactions, where each side effect is an edge of a different type. Decagon is developed specifically to handle such multimodal graphs with a large number of edge types. Our approach develops a new graph convolutional neural network for multirelational link prediction in multimodal networks. Unlike approaches limited to predicting simple drug-drug interaction values, Decagon can predict the exact side effect, if any, through which a given drug combination manifests clinically. Decagon accurately predicts polypharmacy side effects, outperforming baselines by up to 69%. We find that it automatically learns representations of side effects indicative of co-occurrence of polypharmacy in patients. Furthermore, Decagon models particularly well polypharmacy side effects that have a strong molecular basis, while on predominantly non-molecular side effects, it achieves good performance because of effective sharing of model parameters across edge types. Decagon opens up opportunities to use large pharmacogenomic and patient population data to flag and prioritize polypharmacy side effects for follow-up analysis via formal pharmacological studies. Availability and implementation Source code and preprocessed datasets are at: http://snap.stanford.edu/decagon.
Collapse
Affiliation(s)
- Marinka Zitnik
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Monica Agrawal
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
78
|
A computational approach to identify blood cell-expressed Parkinson's disease biomarkers that are coordinately expressed in brain tissue. Comput Biol Med 2019; 113:103385. [PMID: 31437626 DOI: 10.1016/j.compbiomed.2019.103385] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/09/2023]
Abstract
Identification of genes whose regulation of expression is functionally similar in both brain tissue and blood cells could in principle enable monitoring of significant neurological traits and disorders by analysis of blood samples. We thus employed transcriptional analysis of pathologically affected tissues, using agnostic approaches to identify overlapping gene functions and integrating this transcriptomic information with expression quantitative trait loci (eQTL) data. Here, we estimate the correlation of gene expression in the top-associated cis-eQTLs of brain tissue and blood cells in Parkinson's Disease (PD). We introduced quantitative frameworks to reveal the complex relationship of various biasing genetic factors in PD, a neurodegenerative disease. We examined gene expression microarray and RNA-Seq datasets from human brain and blood tissues from PD-affected and control individuals. Differentially expressed genes (DEG) were identified for both brain and blood cells to determine common DEG overlaps. Based on neighborhood-based benchmarking and multilayer network topology approaches we then developed genetic associations of factors with PD. Overlapping DEG sets underwent gene enrichment using pathway analysis and gene ontology methods, which identified candidate common genes and pathways. We identified 12 significantly dysregulated genes shared by brain and blood cells, which were validated using dbGaP (gene SNP-disease linkage) database for gold-standard benchmarking of their significance in disease processes. Ontological and pathway analyses identified significant gene ontology and molecular pathways that indicate PD progression. In sum, we found possible novel links between pathological processes in brain tissue and blood cells by examining cell pathway commonalities, corroborating these associations using well validated datasets. This demonstrates that for brain-related pathologies combining gene expression analysis and blood cell cis-eQTL is a potentially powerful analytical approach. Thus, our methodologies facilitate data-driven approaches that can advance knowledge of disease mechanisms and may, with clinical validation, enable prediction of neurological dysfunction using blood cell transcript profiling.
Collapse
|
79
|
Cai C, Bian X, Xue M, Liu X, Hu H, Wang J, Zheng SG, Sun B, Wu JL. Eicosanoids metabolized through LOX distinguish asthma-COPD overlap from COPD by metabolomics study. Int J Chron Obstruct Pulmon Dis 2019; 14:1769-1778. [PMID: 31496676 PMCID: PMC6689553 DOI: 10.2147/copd.s207023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022] Open
Abstract
Background and objective The prevalence of asthma is greater than 20% in patients previously diagnosed with COPD. Patients with asthma–COPD overlap (ACO) are at risk of rapid progression of disease and severe exacerbations. However, in some patients with ACO, a clear distinction from COPD is very difficult by using physiological testing techniques. This study aimed to apply a novel metabolomic approach to identify the metabolites in sera in order to distinguish ACO from COPD. Methods In the study, blood samples were collected from patients with COPD, ACO, and healthy controls. Cholamine derivatization-ultrahigh performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF/MS) was used to investigate serum metabolites of eicosanoids. Results A clear intergroup separation existed between the patients with ACO and those with COPD, while ACO tends to have higher serum metabolic levels of eicosanoids. A robust Orthogonal Projections to Latent Structures-Discriminant Analysis (OPLS-DA) model was found for discriminating between ACO and COPD (R2Y =0.81, Q2=0.79). In addition, there is a significant correlation between some metabolites and clinical indicators, such as hydroxyeicosatetraenoic acids (HETEs), hydroperoxyeicosatetraenoic acids (HPETEs) and FEV1/FVC. The higher values of area under the receiver operating characteristic curves (ROC) of HETEs, which were metabolized from HPETEs through lipoxygenase (LOX), indicated that they should be the potential biomarkers to distinguish ACO from COPD. Conclusion Eicosanoids can clearly discriminate different biochemical metabolic profiles between ACO and COPD. The results possibly provide a new perspective to identify potential biomarkers of ACO and may be helpful for personalized treatment.
Collapse
Affiliation(s)
- Chuanxu Cai
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China.,Department of Laboratory Medicine, Shenzhen People's Hospital, Shenzhen, People's Republic of China
| | - Xiqing Bian
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macao, People's Republic of China
| | - Mingshan Xue
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoqing Liu
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Haisheng Hu
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jingxian Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Song Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jian-Lin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macao, People's Republic of China
| |
Collapse
|
80
|
Liu HC, Peng YS, Lee HC. miRDRN-miRNA disease regulatory network: a tool for exploring disease and tissue-specific microRNA regulatory networks. PeerJ 2019; 7:e7309. [PMID: 31404401 PMCID: PMC6688598 DOI: 10.7717/peerj.7309] [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: 04/30/2019] [Accepted: 06/17/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND MicroRNA (miRNA) regulates cellular processes by acting on specific target genes, and cellular processes proceed through multiple interactions often organized into pathways among genes and gene products. Hundreds of miRNAs and their target genes have been identified, as are many miRNA-disease associations. These, together with huge amounts of data on gene annotation, biological pathways, and protein-protein interactions are available in public databases. Here, using such data we built a database and web service platform, miRNA disease regulatory network (miRDRN), for users to construct disease and tissue-specific miRNA-protein regulatory networks, with which they may explore disease related molecular and pathway associations, or find new ones, and possibly discover new modes of drug action. METHODS Data on disease-miRNA association, miRNA-target association and validation, gene-tissue association, gene-tumor association, biological pathways, human protein interaction, gene ID, gene ontology, gene annotation, and product were collected from publicly available databases and integrated. A large set of miRNA target-specific regulatory sub-pathways (RSPs) having the form (T, G 1, G 2) was built from the integrated data and stored, where T is a miRNA-associated target gene, G 1 (G 2) is a gene/protein interacting with T (G 1). Each sequence (T, G 1, G 2) was assigned a p-value weighted by the participation of the three genes in molecular interactions and reaction pathways. RESULTS A web service platform, miRDRN (http://mirdrn.ncu.edu.tw/mirdrn/), was built. The database part of miRDRN currently stores 6,973,875 p-valued RSPs associated with 116 diseases in 78 tissue types built from 207 diseases-associated miRNA regulating 389 genes. miRDRN also provides facilities for the user to construct disease and tissue-specific miRNA regulatory networks from RSPs it stores, and to download and/or visualize parts or all of the product. User may use miRDRN to explore a single disease, or a disease-pair to gain insights on comorbidity. As demonstrations, miRDRN was applied: to explore the single disease colorectal cancer (CRC), in which 26 novel potential CRC target genes were identified; to study the comorbidity of the disease-pair Alzheimer's disease-Type 2 diabetes, in which 18 novel potential comorbid genes were identified; and, to explore possible causes that may shed light on recent failures of late-phase trials of anti-AD, BACE1 inhibitor drugs, in which genes downstream to BACE1 whose suppression may affect signal transduction were identified.
Collapse
Affiliation(s)
- Hsueh-Chuan Liu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Yi-Shian Peng
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Hoong-Chien Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
- Department of Physics, Chung Yuan Christian University, Zhongli District, Taoyuan City, Taiwan
| |
Collapse
|
81
|
Cheng L, Yang H, Zhao H, Pei X, Shi H, Sun J, Zhang Y, Wang Z, Zhou M. MetSigDis: a manually curated resource for the metabolic signatures of diseases. Brief Bioinform 2019; 20:203-209. [PMID: 28968812 DOI: 10.1093/bib/bbx103] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Indexed: 12/18/2022] Open
Abstract
Complex diseases cannot be understood only on the basis of single gene, single mRNA transcript or single protein but the effect of their collaborations. The combination consequence in molecular level can be captured by the alterations of metabolites. With the rapidly developing of biomedical instruments and analytical platforms, a large number of metabolite signatures of complex diseases were identified and documented in the literature. Biologists' hardship in the face of this large amount of papers recorded metabolic signatures of experiments' results calls for an automated data repository. Therefore, we developed MetSigDis aiming to provide a comprehensive resource of metabolite alterations in various diseases. MetSigDis is freely available at http://www.bio-annotation.cn/MetSigDis/. By reviewing hundreds of publications, we collected 6849 curated relationships between 2420 metabolites and 129 diseases across eight species involving Homo sapiens and model organisms. All of these relationships were used in constructing a metabolite disease network (MDN). This network displayed scale-free characteristics according to the degree distribution (power-law distribution with R2 = 0.909), and the subnetwork of MDN for interesting diseases and their related metabolites can be visualized in the Web. The common alterations of metabolites reflect the metabolic similarity of diseases, which is measured using Jaccard index. We observed that metabolite-based similar diseases are inclined to share semantic associations of Disease Ontology. A human disease network was then built, where a node represents a disease, and an edge indicates similarity of pair-wise diseases. The network validated the observation that linked diseases based on metabolites should have more overlapped genes.
Collapse
Affiliation(s)
- Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Haixiu Yang
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Hengqiang Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Xiaoya Pei
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Hongbo Shi
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Jie Sun
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Zhenzhen Wang
- College of Bioinformatics Science and Technology, Harbin Medical University
| | - Meng Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University
| |
Collapse
|
82
|
Yu L, Gao L. Human Pathway-Based Disease Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:1240-1249. [PMID: 29990107 DOI: 10.1109/tcbb.2017.2774802] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Constructing disease-disease similarity network is important in elucidating the associations between the origin and molecular mechanism of diseases, and in researching disease function and medical research. In this paper, we use a high-quality protein interaction network and a collection of pathway databases to construct a Human Pathway-based Disease Network (HPDN) to explore the relationship between diseases and their intrinsic interactions. We find that the similarity of two diseases has a strong correlation with the number of their shared functional pathways and the interaction between their related gene sets. Comparing HPDN with disease networks based on genes and symptoms respectively, we find the three networks have high overlap rates. Additionally, HPDN can predict new disease-disease correlations, which are supported by Comparative Toxicogenomics Database (CTD) benchmark and large-scale biomedical literature database. The comprehensive, high-quality relations between diseases based on pathways can further be applied to study important matters in systems medicine, for instance, drug repurposing. Based on a dense subgraph in our network, we find two drugs, prednisone and folic acid, may have new indications, which will provide potential directions for the treatments of complex diseases.
Collapse
|
83
|
Mi Z, Guo B, Yin Z, Li J, Zheng Z. Disease classification via gene network integrating modules and pathways. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190214. [PMID: 31417727 PMCID: PMC6689581 DOI: 10.1098/rsos.190214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/04/2019] [Indexed: 06/10/2023]
Abstract
Disease classification based on gene information has been of significance as the foundation for achieving precision medicine. Previous works focus on classifying diseases according to the gene expression data of patient samples, and constructing disease network based on the overlap of disease genes, as many genes have been confirmed to be associated with diseases. In this work, the effects of diseases on human biological functions are assessed from the perspective of gene network modules and pathways, and the distances between diseases are defined to carry out the classification models. In total, 1728 diseases are divided into 12 and 14 categories by the intensity and scope of effects on pathways, respectively. Each category is a mix of several types of diseases identified based on congenital and acquired factors as well as diseased tissues and organs. The disease classification models on the basis of gene network are parallel with traditional pathology classification based on anatomic and clinical manifestations, and enable us to look at diseases in the viewpoint of commonalities in etiology and pathology. Our models provide a foundation for exploring combination therapy of diseases, which in turn may inform strategies for future gene-targeted therapy.
Collapse
Affiliation(s)
- Zhilong Mi
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing 100191, People’s Republic of China
- LMIB and School of Mathematics and Systems Science, Beihang University, Beijing 100191, People’s Republic of China
- Peng Cheng Laboratory, Shenzhen, Guangdong Province 518055, People’s Republic of China
| | - Binghui Guo
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing 100191, People’s Republic of China
- LMIB and School of Mathematics and Systems Science, Beihang University, Beijing 100191, People’s Republic of China
- Peng Cheng Laboratory, Shenzhen, Guangdong Province 518055, People’s Republic of China
| | - Ziqiao Yin
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing 100191, People’s Republic of China
- LMIB and School of Mathematics and Systems Science, Beihang University, Beijing 100191, People’s Republic of China
- Shenyuan Honors College, Beihang University, Beijing 100191, People’s Republic of China
- Peng Cheng Laboratory, Shenzhen, Guangdong Province 518055, People’s Republic of China
| | - Jiahui Li
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing 100191, People’s Republic of China
- LMIB and School of Mathematics and Systems Science, Beihang University, Beijing 100191, People’s Republic of China
- Peng Cheng Laboratory, Shenzhen, Guangdong Province 518055, People’s Republic of China
| | - Zhiming Zheng
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing 100191, People’s Republic of China
- LMIB and School of Mathematics and Systems Science, Beihang University, Beijing 100191, People’s Republic of China
- Peng Cheng Laboratory, Shenzhen, Guangdong Province 518055, People’s Republic of China
| |
Collapse
|
84
|
Lee DG, Kim M, Shin H. Inference on chains of disease progression based on disease networks. PLoS One 2019; 14:e0218871. [PMID: 31251766 PMCID: PMC6599221 DOI: 10.1371/journal.pone.0218871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/11/2019] [Indexed: 01/16/2023] Open
Abstract
MOTIVATION Disease progression originates from the concept that an individual disease may go through different changes as it evolves, and such changes can cause new diseases. It is important to find a progression between diseases since knowing the prior-posterior relationship beforehand can prevent further complications or evolutions to other diseases. Furthermore, the series of progressions can be represented in the form of a chain, which enables us to readily infer successive influences from one disease to another after many passages through other diseases. METHODS In this paper, we propose a systematic approach for finding a disease progression chain from a source disease to a target one via exploring a disease network. The network is constructed based on various sets of biomedical data. To find the most influential progression chains, the k-shortest path search algorithm is employed. The most representative algorithms such as A*, Dijkstra, and Yen's are incorporated into the proposed method. RESULTS A disease network consisting of 3,302 diseases was constructed based on four sources of biomedical data: disease-protein relations, biological pathways, clinical history, and biomedical literature information. The last three sets of data contain prior-posterior information, and they endow directionality on the edges of the network. The results were interesting and informative: for example, when colitis and respiratory insufficiency were set as a source disease and a target one, respectively, five progression chains were found within several seconds (when k = 5). Each chain was provided with a progression score, which indicates the strength of plausibility relative to others. Similarly, the proposed method can be expanded to any pair of source-target diseases in the network. This can be utilized as a preliminary tool for inferring complications or progressions between diseases.
Collapse
Affiliation(s)
- Dong-gi Lee
- Department of Industrial Engineering, Ajou University, Yeongtong-gu, Suwon, South Korea
| | - Myungjun Kim
- Department of Industrial Engineering, Ajou University, Yeongtong-gu, Suwon, South Korea
| | - Hyunjung Shin
- Department of Industrial Engineering, Ajou University, Yeongtong-gu, Suwon, South Korea
| |
Collapse
|
85
|
Ren Y, Ay A, Dobra A, Kahveci T. Characterizing building blocks of resource constrained biological networks. BMC Bioinformatics 2019; 20:318. [PMID: 31216986 PMCID: PMC6584510 DOI: 10.1186/s12859-019-2838-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background Identification of motifs–recurrent and statistically significant patterns–in biological networks is the key to understand the design principles, and to infer governing mechanisms of biological systems. This, however, is a computationally challenging task. This task is further complicated as biological interactions depend on limited resources, i.e., a reaction takes place if the reactant molecule concentrations are above a certain threshold level. This biochemical property implies that network edges can participate in a limited number of motifs simultaneously. Existing motif counting methods ignore this problem. This simplification often leads to inaccurate motif counts (over- or under-estimates), and thus, wrong biological interpretations. Results In this paper, we develop a novel motif counting algorithm, Partially Overlapping MOtif Counting (POMOC), that considers capacity levels for all interactions in counting motifs. Conclusions Our experiments on real and synthetic networks demonstrate that motif count using the POMOC method significantly differs from the existing motif counting approaches, and our method extends to large-scale biological networks in practical time. Our results also show that our method makes it possible to characterize the impact of different stress factors on cell’s organization of network. In this regard, analysis of a S. cerevisiae transcriptional regulatory network using our method shows that oxidative stress is more disruptive to organization and abundance of motifs in this network than mutations of individual genes. Our analysis also suggests that by focusing on the edges that lead to variation in motif counts, our method can be used to find important genes, and to reveal subtle topological and functional differences of the biological networks under different cell states.
Collapse
Affiliation(s)
- Yuanfang Ren
- Computer and Information Science and Engineering, University of Florida, Gainesville, 32611, FL, USA
| | - Ahmet Ay
- Departments of Biology and Mathematics, Colgate University, Hamilton, 13346, NY, USA
| | - Alin Dobra
- Computer and Information Science and Engineering, University of Florida, Gainesville, 32611, FL, USA
| | - Tamer Kahveci
- Computer and Information Science and Engineering, University of Florida, Gainesville, 32611, FL, USA.
| |
Collapse
|
86
|
Dozmorov MG. Disease classification: from phenotypic similarity to integrative genomics and beyond. Brief Bioinform 2019; 20:1769-1780. [DOI: 10.1093/bib/bby049] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
Abstract
A fundamental challenge of modern biomedical research is understanding how diseases that are similar on the phenotypic level are similar on the molecular level. Integration of various genomic data sets with the traditionally used phenotypic disease similarity revealed novel genetic and molecular mechanisms and blurred the distinction between monogenic (Mendelian) and complex diseases. Network-based medicine has emerged as a complementary approach for identifying disease-causing genes, genetic mediators, disruptions in the underlying cellular functions and for drug repositioning. The recent development of machine and deep learning methods allow for leveraging real-life information about diseases to refine genetic and phenotypic disease relationships. This review describes the historical development and recent methodological advancements for studying disease classification (nosology).
Collapse
Affiliation(s)
- Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, 830 East Main Street, Richmond, VA, USA
| |
Collapse
|
87
|
Bland JS. What is Evidence-Based Functional Medicine in the 21st Century? Integr Med (Encinitas) 2019; 18:14-18. [PMID: 32549804 PMCID: PMC7217393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The 21st century has already demonstrated itself to be an era of change for medicine and science. There is a new openness-to ideas, to a shift in perspectives, to a redefinition of evidence and the many ways it can be gathered. New interest in real-world data, patient-experience information has also become an increasingly important contributor to the evaluation of treatment effectiveness. It is a fertile time on many fronts, including an expanded reach for a systems biology formalism and the Functional Medicine movement.
Collapse
|
88
|
García del Valle EP, Lagunes García G, Prieto Santamaría L, Zanin M, Menasalvas Ruiz E, Rodríguez-González A. Disease networks and their contribution to disease understanding: A review of their evolution, techniques and data sources. J Biomed Inform 2019; 94:103206. [DOI: 10.1016/j.jbi.2019.103206] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/14/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
|
89
|
Lammers M, Kraaijeveld K, Mariën J, Ellers J. Gene expression changes associated with the evolutionary loss of a metabolic trait: lack of lipogenesis in parasitoids. BMC Genomics 2019; 20:309. [PMID: 31014246 PMCID: PMC6480896 DOI: 10.1186/s12864-019-5673-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 04/08/2019] [Indexed: 12/24/2022] Open
Abstract
Background Trait loss is a pervasive phenomenon in evolution, yet the underlying molecular causes have been identified in only a handful of cases. Most of these cases involve loss-of-function mutations in one or more trait-specific genes. However, in parasitoid insects the evolutionary loss of a metabolic trait is not associated with gene decay. Parasitoids have lost the ability to convert dietary sugars into fatty acids. Earlier research suggests that lack of lipogenesis in the parasitoid wasp Nasonia vitripennis is caused by changes in gene regulation. Results We compared transcriptomic responses to sugar-feeding in the non-lipogenic parasitoid species Nasonia vitripennis and the lipogenic Drosophila melanogaster. Both species adjusted their metabolism within 4 hours after sugar-feeding, but there were sharp differences between the expression profiles of the two species, especially in the carbohydrate and lipid metabolic pathways. Several genes coding for key enzymes in acetyl-CoA metabolism, such as malonyl-CoA decarboxylase (mcd) and HMG-CoA synthase (hmgs) differed in expression between the two species. Their combined action likely blocks lipogenesis in the parasitoid species. Network-based analysis showed connectivity of genes to be negatively correlated to the fold change of gene expression. Furthermore, genes involved in the fatty acid metabolic pathway were more connected than the set of genes of all metabolic pathways combined. Conclusion High connectivity of lipogenesis genes is indicative of pleiotropic effects and could explain the absence of gene degradation. We conclude that modification of expression levels of only a few little-connected genes, such as mcd, is sufficient to enable complete loss of lipogenesis in N. vitripennis. Electronic supplementary material The online version of this article (10.1186/s12864-019-5673-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mark Lammers
- Department of Ecological Sciences, Section Animal Ecology, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands.
| | - Ken Kraaijeveld
- Department of Ecological Sciences, Section Animal Ecology, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Janine Mariën
- Department of Ecological Sciences, Section Animal Ecology, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Jacintha Ellers
- Department of Ecological Sciences, Section Animal Ecology, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
90
|
Almasi SM, Hu T. Measuring the importance of vertices in the weighted human disease network. PLoS One 2019; 14:e0205936. [PMID: 30901770 PMCID: PMC6430629 DOI: 10.1371/journal.pone.0205936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/26/2019] [Indexed: 12/11/2022] Open
Abstract
Many human genetic disorders and diseases are known to be related to each other through frequently observed co-occurrences. Studying the correlations among multiple diseases provides an important avenue to better understand the common genetic background of diseases and to help develop new drugs that can treat multiple diseases. Meanwhile, network science has seen increasing applications on modeling complex biological systems, and can be a powerful tool to elucidate the correlations of multiple human diseases. In this article, known disease-gene associations were represented using a weighted bipartite network. We extracted a weighted human diseases network from such a bipartite network to show the correlations of diseases. Subsequently, we proposed a new centrality measurement for the weighted human disease network (WHDN) in order to quantify the importance of diseases. Using our centrality measurement to quantify the importance of vertices in WHDN, we were able to find a set of most central diseases. By investigating the 30 top diseases and their most correlated neighbors in the network, we identified disease linkages including known disease pairs and novel findings. Our research helps better understand the common genetic origin of human diseases and suggests top diseases that likely induce other related diseases.
Collapse
Affiliation(s)
| | - Ting Hu
- Department of Computer Science, Memorial University, St. John’s, NL, Canada
| |
Collapse
|
91
|
Chen SJ, Liao DL, Chen CH, Wang TY, Chen KC. Construction and Analysis of Protein-Protein Interaction Network of Heroin Use Disorder. Sci Rep 2019; 9:4980. [PMID: 30899073 PMCID: PMC6428805 DOI: 10.1038/s41598-019-41552-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Heroin use disorder (HUD) is a complex disease resulting from interactions among genetic and other factors (e.g., environmental factors). The mechanism of HUD development remains unknown. Newly developed network medicine tools provide a platform for exploring complex diseases at the system level. This study proposes that protein–protein interactions (PPIs), particularly those among proteins encoded by casual or susceptibility genes, are extremely crucial for HUD development. The giant component of our constructed PPI network comprised 111 nodes with 553 edges, including 16 proteins with large degree (k) or high betweenness centrality (BC), which were further identified as the backbone of the network. JUN with the largest degree was suggested to be central to the PPI network associated with HUD. Moreover, PCK1 with the highest BC and MAPK14 with the secondary largest degree and 9th highest BC might be involved in the development HUD and other substance diseases.
Collapse
Affiliation(s)
- Shaw-Ji Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Psychiatry, Mackay Memorial Hospital, Taitung Branch, Taiwan
| | - Ding-Lieh Liao
- Bali Psychiatric Center, Department of Health, Executive Yuan, New Taipei, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou and Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Tse-Yi Wang
- Department of Medical Informatics, Tzu Chi University, Hualien, Taiwan
| | - Kuang-Chi Chen
- Department of Medical Informatics, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
92
|
Cao JQ, Li CX, Wang RY, Chen JJ, Ma SM, Wang WY, Meng LJ. Identification of atherosclerosis-related prioritizing metabolites based on a multi-omics composite network. Exp Ther Med 2019; 17:3391-3398. [PMID: 30988716 PMCID: PMC6447794 DOI: 10.3892/etm.2019.7351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/31/2019] [Indexed: 12/23/2022] Open
Abstract
Metabolites are the final products of cellular regulation processes, their level is the ultimate response of biological systems to environmental and genetic changes. Therefore, the identification of key metabolites is required for the diagnosis and therapy of diseases. In this study, atherosclerosis-related gene expression profile information was extracted from ArrayExpress database (GEOD-57691), and analyzed with limma package. Furthermore, we constructed an intricate multi-omics network involved in genes, phenotypes, metabolites and their associations. To identify the prioritization of atherosclerosis-related metabolites, the relation score of each metabolite in the composite network was computed with the random walk with restart (RWR) method. The top 50 metabolites and top 100 genes were chosen based on the score in the weighted composite network. Consequently, several key metabolites that were ranked in the top 5 of relation score or degree greater than 70 were confirmed. Particularly, metabolites Tretinoin and Estraderm not only have high relation scores, but also contain more degrees. Moreover, we obtained 24 co-expression genes that may be regarded as the targets of atherosclerosis therapy. Therefore, identification of metabolite prioritizations by the composite network integrated the information of genes, phenotypes and metabolites may be available to diagnose atherosclerosis, and can provide the potential therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Jun-Qiang Cao
- Department of Cardiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| | - Cai-Xia Li
- Department of Cardiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| | - Ru-Yi Wang
- Department of Cardiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| | - Jin-Jin Chen
- Department of Anesthesiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| | - Shu-Mei Ma
- Department of Cardiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| | - Wen-Ying Wang
- Department of Cardiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| | - Li-Jun Meng
- Department of Cardiology, Binzhou City Center Hospital, Binzhou, Shandong 251700, P.R. China
| |
Collapse
|
93
|
Ma J, Wang J, Ghoraie LS, Men X, Haibe-Kains B, Dai P. A Comparative Study of Cluster Detection Algorithms in Protein-Protein Interaction for Drug Target Discovery and Drug Repurposing. Front Pharmacol 2019; 10:109. [PMID: 30837876 PMCID: PMC6389713 DOI: 10.3389/fphar.2019.00109] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/28/2019] [Indexed: 12/29/2022] Open
Abstract
The interactions between drugs and their target proteins induce altered expression of genes involved in complex intracellular networks. The properties of these functional network modules are critical for the identification of drug targets, for drug repurposing, and for understanding the underlying mode of action of the drug. The topological modules generated by a computational approach are defined as functional clusters. However, the functions inferred for these topological modules extracted from a large-scale molecular interaction network, such as a protein–protein interaction (PPI) network, could differ depending on different cluster detection algorithms. Moreover, the dynamic gene expression profiles among tissues or cell types causes differential functional interaction patterns between the molecular components. Thus, the connections in the PPI network should be modified by the transcriptomic landscape of specific cell lines before producing topological clusters. Here, we systematically investigated the clusters of a cell-based PPI network by using four cluster detection algorithms. We subsequently compared the performance of these algorithms for target gene prediction, which integrates gene perturbation data with the cell-based PPI network using two drug target prioritization methods, shortest path and diffusion correlation. In addition, we validated the proportion of perturbed genes in clusters by finding candidate anti-breast cancer drugs and confirming our predictions using literature evidence and cases in the ClinicalTrials.gov. Our results indicate that the Walktrap (CW) clustering algorithm achieved the best performance overall in our comparative study.
Collapse
Affiliation(s)
- Jun Ma
- National Engineering Research Center for Miniaturized Detection Systems, Northwest University, Xi'an, China.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jenny Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Xin Men
- Shaanxi Microbiology Institute, Xi'an, China
| | | | - Penggao Dai
- National Engineering Research Center for Miniaturized Detection Systems, Northwest University, Xi'an, China
| |
Collapse
|
94
|
Xi Y, Wang F. Extreme pathway analysis reveals the organizing rules of metabolic regulation. PLoS One 2019; 14:e0210539. [PMID: 30721240 PMCID: PMC6363282 DOI: 10.1371/journal.pone.0210539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/27/2018] [Indexed: 11/18/2022] Open
Abstract
Cellular systems shift metabolic states by adjusting gene expression and enzyme activities to adapt to physiological and environmental changes. Biochemical and genetic studies are identifying how metabolic regulation affects the selection of metabolic phenotypes. However, how metabolism influences its regulatory architecture still remains unexplored. We present a new method of extreme pathway analysis (the minimal set of conically independent metabolic pathways) to deduce regulatory structures from pure pathway information. Applying our method to metabolic networks of human red blood cells and Escherichia coli, we shed light on how metabolic regulation are organized by showing which reactions within metabolic networks are more prone to transcriptional or allosteric regulation. Applied to a human genome-scale metabolic system, our method detects disease-associated reactions. Thus, our study deepens the understanding of the organizing principle of cellular metabolic regulation and may contribute to metabolic engineering, synthetic biology, and disease treatment.
Collapse
Affiliation(s)
- Yanping Xi
- Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China
- School of Computer Science and Technology, Fudan University, Shanghai, China
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Fei Wang
- Shanghai Key Lab of Intelligent Information Processing, Fudan University, Shanghai, China
- School of Computer Science and Technology, Fudan University, Shanghai, China
| |
Collapse
|
95
|
Li H, Fan J, Vitali F, Berghout J, Aberasturi D, Li J, Wilson L, Chiu W, Pumarejo M, Han J, Kenost C, Koripella PC, Pouladi N, Billheimer D, Bedrick EJ, Lussier YA. Novel disease syndromes unveiled by integrative multiscale network analysis of diseases sharing molecular effectors and comorbidities. BMC Med Genomics 2018; 11:112. [PMID: 30598089 PMCID: PMC6311938 DOI: 10.1186/s12920-018-0428-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background Forty-two percent of patients experience disease comorbidity, contributing substantially to mortality rates and increased healthcare costs. Yet, the possibility of underlying shared mechanisms for diseases remains not well established, and few studies have confirmed their molecular predictions with clinical datasets. Methods In this work, we integrated genome-wide association study (GWAS) associating diseases and single nucleotide polymorphisms (SNPs) with transcript regulatory activity from expression quantitative trait loci (eQTL). This allowed novel mechanistic insights for noncoding and intergenic regions. We then analyzed pairs of SNPs across diseases to identify shared molecular effectors robust to multiple test correction (False Discovery Rate FDReRNA < 0.05). We hypothesized that disease pairs found to be molecularly convergent would also be significantly overrepresented among comorbidities in clinical datasets. To assess our hypothesis, we used clinical claims datasets from the Healthcare Cost and Utilization Project (HCUP) and calculated significant disease comorbidities (FDRcomorbidity < 0.05). We finally verified if disease pairs resulting molecularly convergent were also statistically comorbid more than by chance using the Fisher’s Exact Test. Results Our approach integrates: (i) 6175 SNPs associated with 238 diseases from ~ 1000 GWAS, (ii) eQTL associations from 19 tissues, and (iii) claims data for 35 million patients from HCUP. Logistic regression (controlled for age, gender, and race) identified comorbidities in HCUP, while enrichment analyses identified cis- and trans-eQTL downstream effectors of GWAS-identified variants. Among ~ 16,000 combinations of diseases, 398 disease-pairs were prioritized by both convergent eQTL-genetics (RNA overlap enrichment, FDReRNA < 0.05) and clinical comorbidities (OR > 1.5, FDRcomorbidity < 0.05). Case studies of comorbidities illustrate specific convergent noncoding regulatory elements. An intergenic architecture of disease comorbidity was unveiled due to GWAS and eQTL-derived convergent mechanisms between distinct diseases being overrepresented among observed comorbidities in clinical datasets (OR = 8.6, p-value = 6.4 × 10− 5 FET). Conclusions These comorbid diseases with convergent eQTL genetic mechanisms suggest clinical syndromes. While it took over a decade to confirm the genetic underpinning of the metabolic syndrome, this study is likely highlighting hundreds of new ones. Further, this knowledge may improve the clinical management of comorbidities with precision and shed light on novel approaches of drug repositioning or SNP-guided precision molecular therapy inclusive of intergenic risks. Electronic supplementary material The online version of this article (10.1186/s12920-018-0428-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haiquan Li
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA. .,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA. .,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA. .,Department of Biosystems Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
| | - Jungwei Fan
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Francesca Vitali
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Joanne Berghout
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,The Center for Applied Genetics and Genomics Medicine, The University of Arizona, Tucson, AZ, 85721, USA.,The Center for Innovation in Brain Science, The University of Arizona, Tucson, AZ, 85721, USA
| | - Dillon Aberasturi
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianrong Li
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Liam Wilson
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Wesley Chiu
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Minsu Pumarejo
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jiali Han
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Systems & Industrial Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Colleen Kenost
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Pradeep C Koripella
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Nima Pouladi
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Dean Billheimer
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA.,Epidemiology and Biostatistics Department, College of Public Health, The University of Arizona, Tucson, AZ, 85721, USA
| | - Edward J Bedrick
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA.,Epidemiology and Biostatistics Department, College of Public Health, The University of Arizona, Tucson, AZ, 85721, USA
| | - Yves A Lussier
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA. .,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA. .,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA. .,The Center for Applied Genetics and Genomics Medicine, The University of Arizona, Tucson, AZ, 85721, USA. .,The Center for Innovation in Brain Science, The University of Arizona, Tucson, AZ, 85721, USA. .,UA Cancer Center, The University of Arizona, Tucson, AZ, 85721, USA. .,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
96
|
Roca J, Tenyi A, Cano I. Paradigm changes for diagnosis: using big data for prediction. ACTA ACUST UNITED AC 2018; 57:317-327. [DOI: 10.1515/cclm-2018-0971] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/21/2018] [Indexed: 11/15/2022]
Abstract
Abstract
Due to profound changes occurring in biomedical knowledge and in health systems worldwide, an entirely new health and social care scenario is emerging. Moreover, the enormous technological potential developed over the last years is increasingly influencing life sciences and driving changes toward personalized medicine and value-based healthcare. However, the current slow progression of adoption, limiting the generation of healthcare efficiencies through technological innovation, can be realistically overcome by fostering convergence between a systems medicine approach and the principles governing Integrated Care. Implicit with this strategy is the multidisciplinary active collaboration of all stakeholders involved in the change, namely: citizens, professionals with different profiles, academia, policy makers, industry and payers. The article describes the key building blocks of an open and collaborative hub currently being developed in Catalonia (Spain) aiming at generation, deployment and evaluation of a personalized medicine program addressing highly prevalent chronic conditions that often show co-occurrence, namely: cardiovascular disorders, chronic obstructive pulmonary disease, type 2 diabetes mellitus; metabolic syndrome and associated mental disturbances (anxiety-depression and altered behavioral patterns leading to unhealthy life styles).
Collapse
Affiliation(s)
- Josep Roca
- Hospital Clínic, IDIBAPS, Facultat de Medicina , Universitat de Barcelona , Barcelona, Catalunya , Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES) , Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 , 28029, Madrid, Catalunya , Spain , Phone: +34-932275747, Fax: +34-932275455
| | - Akos Tenyi
- Hospital Clínic, IDIBAPS, Facultat de Medicina , Universitat de Barcelona , Barcelona, Catalunya , Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES) , Madrid, Catalunya , Spain
| | - Isaac Cano
- Hospital Clínic, IDIBAPS, Facultat de Medicina , Universitat de Barcelona , Barcelona, Catalunya , Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES) , Madrid, Catalunya , Spain
| |
Collapse
|
97
|
Abstract
BACKGROUND Inflammation is a core element of many different, systemic and chronic diseases that usually involve an important autoimmune component. The clinical phase of inflammatory diseases is often the culmination of a long series of pathologic events that started years before. The systemic characteristics and related mechanisms could be investigated through the multi-omic comparative analysis of many inflammatory diseases. Therefore, it is important to use molecular data to study the genesis of the diseases. Here we propose a new methodology to study the relationships between inflammatory diseases and signalling molecules whose dysregulation at molecular levels could lead to systemic pathological events observed in inflammatory diseases. RESULTS We first perform an exploratory analysis of gene expression data of a number of diseases that involve a strong inflammatory component. The comparison of gene expression between disease and healthy samples reveals the importance of members of gene families coding for signalling factors. Next, we focus on interested signalling gene families and a subset of inflammation related diseases with multi-omic features including both gene expression and DNA methylation. We introduce a phylogenetic-based multi-omic method to study the relationships between multi-omic features of inflammation related diseases by integrating gene expression, DNA methylation through sequence based phylogeny of the signalling gene families. The models of adaptations between gene expression and DNA methylation can be inferred from pre-estimated evolutionary relationship of a gene family. Members of the gene family whose expression or methylation levels significantly deviate from the model are considered as the potential disease associated genes. CONCLUSIONS Applying the methodology to four gene families (the chemokine receptor family, the TNF receptor family, the TGF- β gene family, the IL-17 gene family) in nine inflammation related diseases, we identify disease associated genes which exhibit significant dysregulation in gene expression or DNA methylation in the inflammation related diseases, which provides clues for functional associations between the diseases.
Collapse
Affiliation(s)
- Hui Xiao
- Computer Laboratory, University of Cambridge, Cambridge, UK
| | - Krzysztof Bartoszek
- Department of Computer and Information Science, Linköping University, Linköping, Sweden
| | - Pietro Lio’
- Computer Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
98
|
Fotouhi B, Momeni N, Riolo MA, Buckeridge DL. Statistical methods for constructing disease comorbidity networks from longitudinal inpatient data. APPLIED NETWORK SCIENCE 2018; 3:46. [PMID: 30465022 PMCID: PMC6223974 DOI: 10.1007/s41109-018-0101-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/12/2018] [Indexed: 06/09/2023]
Abstract
Tools from network science can be utilized to study relations between diseases. Different studies focus on different types of inter-disease linkages. One of them is the comorbidity patterns derived from large-scale longitudinal data of hospital discharge records. Researchers seek to describe comorbidity relations as a network to characterize pathways of disease progressions and to predict future risks. The first step in such studies is the construction of the network itself, which subsequent analyses rest upon. There are different ways to build such a network. In this paper, we provide an overview of several existing statistical approaches in network science applicable to weighted directed networks. We discuss the differences between the null models that these models assume and their applications. We apply these methods to the inpatient data of approximately one million people, spanning approximately 17 years, pertaining to the Montreal Census Metropolitan Area. We discuss the differences in the structure of the networks built by different methods, and different features of the comorbidity relations that they extract. We also present several example applications of these methods.
Collapse
Affiliation(s)
- Babak Fotouhi
- Program for Evolutionary Dynamics, Harvard University, Cambridge, USA
| | - Naghmeh Momeni
- Sloan School of Management, Massachusetts Institute of Technology, Cambridge, USA
| | - Maria A. Riolo
- Center for the Study of Complex Systems, University of Michigan, Ann Arbor, Michigan USA
| | - David L. Buckeridge
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada
| |
Collapse
|
99
|
Chami GF, Kabatereine NB, Tukahebwa EM, Dunne DW. Precision global health and comorbidity: a population-based study of 16 357 people in rural Uganda. J R Soc Interface 2018; 15:20180248. [PMID: 30381343 PMCID: PMC6228477 DOI: 10.1098/rsif.2018.0248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022] Open
Abstract
In low-income countries, complex comorbidities and weak health systems confound disease diagnosis and treatment. Yet, data-driven approaches have not been applied to develop better diagnostic strategies or to tailor treatment delivery for individuals within rural poor communities. We observed symptoms/diseases reported within three months by 16 357 individuals aged 1+ years in 17 villages of Mayuge District, Uganda. Symptoms were mapped to the Human Phenotype Ontology. Comorbidity networks were constructed. An edge between two symptoms/diseases was generated if the relative risk greater than 1, ϕ correlation greater than 0, and local false discovery rate less than 0.05. We studied how network structure and flagship symptom profiles varied against biosocial factors. 88.05% of individuals (14 402/16 357) reported at least one symptom/disease. Young children and individuals in worse-off households-low socioeconomic status, poor water, sanitation, and hygiene, and poor medical care-had dense network structures with the highest comorbidity burden and/or were conducive to the onset of new comorbidities from existing flagship symptoms, such as fever. Flagship symptom profiles for fever revealed self-misdiagnoses of fever as malaria and sexually transmitted infections as a potentially missed cause of fever in individuals of reproductive age. Network analysis may inform the development of new diagnostic and treatment strategies for flagship symptoms used to characterize syndromes/diseases of global concern.
Collapse
Affiliation(s)
- Goylette F Chami
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Narcis B Kabatereine
- Schistosomiasis Control Initiative, Imperial College London, Norfolk Place, London W2 1PG, UK
- Bilharzia and Worm Control Programme, Vector Control Division, Ministry of Health, 15 Bombo Road, Kampala, Uganda
| | - Edridah M Tukahebwa
- Bilharzia and Worm Control Programme, Vector Control Division, Ministry of Health, 15 Bombo Road, Kampala, Uganda
| | - David W Dunne
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
100
|
Amell A, Roso-Llorach A, Palomero L, Cuadras D, Galván-Femenía I, Serra-Musach J, Comellas F, de Cid R, Pujana MA, Violán C. Disease networks identify specific conditions and pleiotropy influencing multimorbidity in the general population. Sci Rep 2018; 8:15970. [PMID: 30374096 PMCID: PMC6206057 DOI: 10.1038/s41598-018-34361-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/15/2018] [Indexed: 01/16/2023] Open
Abstract
Multimorbidity is an emerging topic in public health policy because of its increasing prevalence and socio-economic impact. However, the age- and gender-dependent trends of disease associations at fine resolution, and the underlying genetic factors, remain incompletely understood. Here, by analyzing disease networks from electronic medical records of primary health care, we identify key conditions and shared genetic factors influencing multimorbidity. Three types of diseases are outlined: "central", which include chronic and non-chronic conditions, have higher cumulative risks of disease associations; "community roots" have lower cumulative risks, but inform on continuing clustered disease associations with age; and "seeds of bursts", which most are chronic, reveal outbreaks of disease associations leading to multimorbidity. The diseases with a major impact on multimorbidity are caused by genes that occupy central positions in the network of human disease genes. Alteration of lipid metabolism connects breast cancer, diabetic neuropathy and nutritional anemia. Evaluation of key disease associations by a genome-wide association study identifies shared genetic factors and further supports causal commonalities between nervous system diseases and nutritional anemias. This study also reveals many shared genetic signals with other diseases. Collectively, our results depict novel population-based multimorbidity patterns, identify key diseases within them, and highlight pleiotropy influencing multimorbidity.
Collapse
Affiliation(s)
- A Amell
- Department of Mathematics, Technical University of Catalonia, Castelldefels, Barcelona, 08860, Catalonia, Spain
| | - A Roso-Llorach
- Jordi Gol University Institute for Research Primary Healthcare (IDIAP Jordi Gol), Barcelona, 08007, Catalonia, Spain
- Autonomous University of Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - L Palomero
- ProCURE, Catalan Institute of Oncology (ICO), Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - D Cuadras
- Statistics Department, Foundation Sant Joan de Déu, Esplugues, 08950, Catalonia, Spain
| | - I Galván-Femenía
- GCAT-Genomes for Life, Germans Trias i Pujol Health Sciences Research Institute (IGTP), Program for Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, 08916, Catalonia, Spain
| | - J Serra-Musach
- ProCURE, Catalan Institute of Oncology (ICO), Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - F Comellas
- Department of Mathematics, Technical University of Catalonia, Castelldefels, Barcelona, 08860, Catalonia, Spain
| | - R de Cid
- GCAT-Genomes for Life, Germans Trias i Pujol Health Sciences Research Institute (IGTP), Program for Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, 08916, Catalonia, Spain.
| | - M A Pujana
- ProCURE, Catalan Institute of Oncology (ICO), Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.
| | - C Violán
- Jordi Gol University Institute for Research Primary Healthcare (IDIAP Jordi Gol), Barcelona, 08007, Catalonia, Spain.
- Autonomous University of Barcelona, Bellaterra, 08193, Catalonia, Spain.
| |
Collapse
|