51
|
Transcriptional drug repositioning and cheminformatics approach for differentiation therapy of leukaemia cells. Sci Rep 2021; 11:12537. [PMID: 34131166 PMCID: PMC8206077 DOI: 10.1038/s41598-021-91629-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023] Open
Abstract
Differentiation therapy is attracting increasing interest in cancer as it can be more specific than conventional chemotherapy approaches, and it has offered new treatment options for some cancer types, such as treating acute promyelocytic leukaemia (APL) by retinoic acid. However, there is a pressing need to identify additional molecules which act in this way, both in leukaemia and other cancer types. In this work, we hence developed a novel transcriptional drug repositioning approach, based on both bioinformatics and cheminformatics components, that enables selecting such compounds in a more informed manner. We have validated the approach for leukaemia cells, and retrospectively retinoic acid was successfully identified using our method. Prospectively, the anti-parasitic compound fenbendazole was tested in leukaemia cells, and we were able to show that it can induce the differentiation of leukaemia cells to granulocytes in low concentrations of 0.1 μM and within as short a time period as 3 days. This work hence provides a systematic and validated approach for identifying small molecules for differentiation therapy in cancer.
Collapse
|
52
|
Hudson ML, Samudrala R. Multiscale Virtual Screening Optimization for Shotgun Drug Repurposing Using the CANDO Platform. Molecules 2021; 26:2581. [PMID: 33925237 PMCID: PMC8125683 DOI: 10.3390/molecules26092581] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/02/2022] Open
Abstract
Drug repurposing, the practice of utilizing existing drugs for novel clinical indications, has tremendous potential for improving human health outcomes and increasing therapeutic development efficiency. The goal of multi-disease multitarget drug repurposing, also known as shotgun drug repurposing, is to develop platforms that assess the therapeutic potential of each existing drug for every clinical indication. Our Computational Analysis of Novel Drug Opportunities (CANDO) platform for shotgun multitarget repurposing implements several pipelines for the large-scale modeling and simulation of interactions between comprehensive libraries of drugs/compounds and protein structures. In these pipelines, each drug is described by an interaction signature that is compared to all other signatures that are subsequently sorted and ranked based on similarity. Pipelines within the platform are benchmarked based on their ability to recover known drugs for all indications in our library, and predictions are generated based on the hypothesis that (novel) drugs with similar signatures may be repurposed for the same indication(s). The drug-protein interactions used to create the drug-proteome signatures may be determined by any screening or docking method, but the primary approach used thus far has been BANDOCK, our in-house bioanalytical or similarity docking protocol. In this study, we calculated drug-proteome interaction signatures using the publicly available molecular docking method Autodock Vina and created hybrid decision tree pipelines that combined our original bio- and chem-informatic approach with the goal of assessing and benchmarking their drug repurposing capabilities and performance. The hybrid decision tree pipeline outperformed the two docking-based pipelines from which it was synthesized, yielding an average indication accuracy of 13.3% at the top10 cutoff (the most stringent), relative to 10.9% and 7.1% for its constituent pipelines, and a random control accuracy of 2.2%. We demonstrate that docking-based virtual screening pipelines have unique performance characteristics and that the CANDO shotgun repurposing paradigm is not dependent on a specific docking method. Our results also provide further evidence that multiple CANDO pipelines can be synthesized to enhance drug repurposing predictive capability relative to their constituent pipelines. Overall, this study indicates that pipelines consisting of varied docking-based signature generation methods can capture unique and useful signals for accurate comparison of drug-proteome interaction signatures, leading to improvements in the benchmarking and predictive performance of the CANDO shotgun drug repurposing platform.
Collapse
Affiliation(s)
| | - Ram Samudrala
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA;
| |
Collapse
|
53
|
Xuan P, Zhang Y, Cui H, Zhang T, Guo M, Nakaguchi T. Integrating multi-scale neighbouring topologies and cross-modal similarities for drug-protein interaction prediction. Brief Bioinform 2021; 22:6220173. [PMID: 33839743 DOI: 10.1093/bib/bbab119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/15/2021] [Accepted: 03/12/2021] [Indexed: 01/02/2023] Open
Abstract
MOTIVATION Identifying the proteins that interact with drugs can reduce the cost and time of drug development. Existing computerized methods focus on integrating drug-related and protein-related data from multiple sources to predict candidate drug-target interactions (DTIs). However, multi-scale neighboring node sequences and various kinds of drug and protein similarities are neither fully explored nor considered in decision making. RESULTS We propose a drug-target interaction prediction method, DTIP, to encode and integrate multi-scale neighbouring topologies, multiple kinds of similarities, associations, interactions related to drugs and proteins. We firstly construct a three-layer heterogeneous network to represent interactions and associations across drug, protein, and disease nodes. Then a learning framework based on fully-connected autoencoder is proposed to learn the nodes' low-dimensional feature representations within the heterogeneous network. Secondly, multi-scale neighbouring sequences of drug and protein nodes are formulated by random walks. A module based on bidirectional gated recurrent unit is designed to learn the neighbouring sequential information and integrate the low-dimensional features of nodes. Finally, we propose attention mechanisms at feature level, neighbouring topological level and similarity level to learn more informative features, topologies and similarities. The prediction results are obtained by integrating neighbouring topologies, similarities and feature attributes using a multiple layer CNN. Comprehensive experimental results over public dataset demonstrated the effectiveness of our innovative features and modules. Comparison with other state-of-the-art methods and case studies of five drugs further validated DTIP's ability in discovering the potential candidate drug-related proteins.
Collapse
Affiliation(s)
- Ping Xuan
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
| | - Yu Zhang
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
| | - Hui Cui
- Department of Computer Science and Information Technology, La Trobe University, Melbourne 3083, Australia
| | - Tiangang Zhang
- School of Mathematical Science, Heilongjiang University, Harbin 150080, China
| | - Maozu Guo
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Toshiya Nakaguchi
- Center for Frontier Medical Engineering, Chiba University, Chiba 2638522, Japan
| |
Collapse
|
54
|
Liu Z, Chen Q, Lan W, Pan H, Hao X, Pan S. GADTI: Graph Autoencoder Approach for DTI Prediction From Heterogeneous Network. Front Genet 2021; 12:650821. [PMID: 33912218 PMCID: PMC8072283 DOI: 10.3389/fgene.2021.650821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/12/2021] [Indexed: 12/26/2022] Open
Abstract
Identifying drug–target interaction (DTI) is the basis for drug development. However, the method of using biochemical experiments to discover drug-target interactions has low coverage and high costs. Many computational methods have been developed to predict potential drug-target interactions based on known drug-target interactions, but the accuracy of these methods still needs to be improved. In this article, a graph autoencoder approach for DTI prediction (GADTI) was proposed to discover potential interactions between drugs and targets using a heterogeneous network, which integrates diverse drug-related and target-related datasets. Its encoder consists of two components: a graph convolutional network (GCN) and a random walk with restart (RWR). And the decoder is DistMult, a matrix factorization model, using embedding vectors from encoder to discover potential DTIs. The combination of GCN and RWR can provide nodes with more information through a larger neighborhood, and it can also avoid over-smoothing and computational complexity caused by multi-layer message passing. Based on the 10-fold cross-validation, we conduct three experiments in different scenarios. The results show that GADTI is superior to the baseline methods in both the area under the receiver operator characteristic curve and the area under the precision–recall curve. In addition, based on the latest Drugbank dataset (V5.1.8), the case study shows that 54.8% of new approved DTIs are predicted by GADTI.
Collapse
Affiliation(s)
- Zhixian Liu
- School of Medical, Guangxi University, Nanning, China.,School of Electronics and Information Engineering, Beibu Gulf University, Qinzhou, China
| | - Qingfeng Chen
- School of Computer, Electronic and Information, Guangxi University, Nanning, China
| | - Wei Lan
- School of Computer, Electronic and Information, Guangxi University, Nanning, China
| | - Haiming Pan
- School of Computer, Electronic and Information, Guangxi University, Nanning, China
| | - Xinkun Hao
- School of Computer, Electronic and Information, Guangxi University, Nanning, China
| | - Shirui Pan
- Department of Data Science and AI, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
55
|
Shang Y, Gao L, Zou Q, Yu L. Prediction of drug-target interactions based on multi-layer network representation learning. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2020.12.068] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
56
|
Zhou W, Chen Z, Lu A, Liu Z. Systems Pharmacology-Based Strategy to Explore the Pharmacological Mechanisms of Citrus Peel (Chenpi) for Treating Complicated Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:391-411. [PMID: 33622210 DOI: 10.1142/s0192415x2150018x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Citri Reticulatae Pericarpium (CRP), also known as Chenpi in Chinese, is the dry mature peel of Citrus reticulata Blanco or its cultivated varieties. CRP as the health-care food and dietary supplement has been widely used in various diseases. However, the potential pharmacological mechanisms of CRP to predict and treat various diseases have not yet been fully elucidated. A systems pharmacology-based approach is developed by integrating absorption, distribution, metabolism, and excretion screening, multiple target fishing, network pharmacology, as well as pathway analysis to comprehensively dissect the potential mechanism of CRP for therapy of various diseases. The results showed that 39 bioactive components and 121 potential protein targets were identified from CRP. The 121 targets are closely related to various diseases of the cardiovascular system, respiratory system, gastrointestinal system, etc. These targets are further mapped to compound-target, target-disease, and target-pathway networks to clarify the therapeutic mechanism of CRP at the system level. The current study sheds light on a promising way for promoting the discovery of new botanical drugs.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen University, Shenzhen, P. R. China.,State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen University, Shenzhen, P. R. China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, P. R. China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong, P. R. China
| | - Zhigang Liu
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen University, Shenzhen, P. R. China.,State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen University, Shenzhen, P. R. China
| |
Collapse
|
57
|
Peng J, Wang Y, Guan J, Li J, Han R, Hao J, Wei Z, Shang X. An end-to-end heterogeneous graph representation learning-based framework for drug-target interaction prediction. Brief Bioinform 2021; 22:6124914. [PMID: 33517357 DOI: 10.1093/bib/bbaa430] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/01/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022] Open
Abstract
Accurately identifying potential drug-target interactions (DTIs) is a key step in drug discovery. Although many related experimental studies have been carried out for identifying DTIs in the past few decades, the biological experiment-based DTI identification is still timeconsuming and expensive. Therefore, it is of great significance to develop effective computational methods for identifying DTIs. In this paper, we develop a novel 'end-to-end' learning-based framework based on heterogeneous 'graph' convolutional networks for 'DTI' prediction called end-to-end graph (EEG)-DTI. Given a heterogeneous network containing multiple types of biological entities (i.e. drug, protein, disease, side-effect), EEG-DTI learns the low-dimensional feature representation of drugs and targets using a graph convolutional networks-based model and predicts DTIs based on the learned features. During the training process, EEG-DTI learns the feature representation of nodes in an end-to-end mode. The evaluation test shows that EEG-DTI performs better than existing state-of-art methods. The data and source code are available at: https://github.com/MedicineBiology-AI/EEG-DTI.
Collapse
Affiliation(s)
- Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yuxian Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an 710072, China
| | - Jiaojiao Guan
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an 710072, China
| | - Jingyi Li
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an 710072, China
| | - Ruijiang Han
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jianye Hao
- College of Intelligence and Computing, Tianjin University, Tianjin 300072, China
| | - Zhongyu Wei
- School of Data Science, Fudan University, Shanghai 200433, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an 710072, China
| |
Collapse
|
58
|
Zhao J, Yang L, Huang L, Li Z. Screening of disease-related biomarkers related to neuropathic pain (NP) after spinal cord injury (SCI). Hum Genomics 2021; 15:5. [PMID: 33494823 PMCID: PMC7831171 DOI: 10.1186/s40246-021-00303-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 01/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Based on the molecular expression level, this paper compares lncRNA and mRNA expressions respectively in peripheral blood samples of the patients after SCI with NP and without NP, and screens disease-related biomarkers related to NP after SCI in peripheral blood samples of patients. METHOD The expression spectrum of 25 human peripheral blood samples (12 samples of refractory NP patients after SCI) was downloaded and data were normalized. Screening of GO annotations significantly associated with significant differentially expressed mRNAs and significant involvement of the KEGG pathway. The WGCNA algorithm was used to screen for modules and RNAs that were significantly associated with disease characterization. A co-expression network was constructed to extract the genes involved in the disease pathway from the co-expression network, construct a network of SCI pain-related pathways, and screen important disease-related biomarkers. Quantitative real-time PCR was used to detect the mRNA expression of hub genes. RESULTS Data were normalized and re-annotated by detection of platform information, resulting in a total of 289 lncRNA and 18197 mRNAs. Screening resulted in 338 significant differentially expressed RNAs that met the threshold requirements. Differentially expressed RNAs were significantly enriched with the brown and magenta modules. Six KEGG signaling pathways were screened in the co-expression network, and three KEGG pathways with direct neuropathic pain were identified. The expression levels of E2F1, MAX, MITF, CTNNA1, and ADORA2B in the disease group were all significantly upregulated (p < 0.01). Compared with the normal group, the expression of OXTR was upregulated. CONCLUSION We speculate that there are 7 genes and 2 lncRNAs directly involved in the pain pathway: E2F1, MAX, MITF, CTNNA1, ADORA2B, GRIK3, OXTR, LINC01119, and LINC02447. These molecules may be important for NP after SCI.
Collapse
Affiliation(s)
- Jia Zhao
- Department of Internal Neurology, The Third Hospital of Jilin University, 126 Xiantai Street, Changchun, 130000 Jilin, People’s Republic of China
| | - Li Yang
- Department of Internal Neurology, The Third Hospital of Jilin University, 126 Xiantai Street, Changchun, 130000 Jilin, People’s Republic of China
| | - Limin Huang
- Department of Internal Neurology , The Third Hospital of Jilin University , 126 Xiantai Street, Changchun, 130000 Jilin People’s Republic of China
| | - Zinan Li
- Department of Internal Neurology, The Third Hospital of Jilin University, 126 Xiantai Street, Changchun, 130000 Jilin, People’s Republic of China
| |
Collapse
|
59
|
Kamdar MR, Musen MA. An empirical meta-analysis of the life sciences linked open data on the web. Sci Data 2021; 8:24. [PMID: 33479214 PMCID: PMC7819992 DOI: 10.1038/s41597-021-00797-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/04/2020] [Indexed: 01/29/2023] Open
Abstract
While the biomedical community has published several "open data" sources in the last decade, most researchers still endure severe logistical and technical challenges to discover, query, and integrate heterogeneous data and knowledge from multiple sources. To tackle these challenges, the community has experimented with Semantic Web and linked data technologies to create the Life Sciences Linked Open Data (LSLOD) cloud. In this paper, we extract schemas from more than 80 biomedical linked open data sources into an LSLOD schema graph and conduct an empirical meta-analysis to evaluate the extent of semantic heterogeneity across the LSLOD cloud. We observe that several LSLOD sources exist as stand-alone data sources that are not inter-linked with other sources, use unpublished schemas with minimal reuse or mappings, and have elements that are not useful for data integration from a biomedical perspective. We envision that the LSLOD schema graph and the findings from this research will aid researchers who wish to query and integrate data and knowledge from multiple biomedical sources simultaneously on the Web.
Collapse
Affiliation(s)
- Maulik R Kamdar
- Center for Biomedical Informatics Research, Stanford University, Stanford, CA, USA.
- Elsevier Health Markets, Philadelphia, PA, USA.
| | - Mark A Musen
- Center for Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| |
Collapse
|
60
|
Davis AP, Grondin CJ, Johnson RJ, Sciaky D, Wiegers J, Wiegers TC, Mattingly CJ. Comparative Toxicogenomics Database (CTD): update 2021. Nucleic Acids Res 2021; 49:D1138-D1143. [PMID: 33068428 PMCID: PMC7779006 DOI: 10.1093/nar/gkaa891] [Citation(s) in RCA: 571] [Impact Index Per Article: 190.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
The public Comparative Toxicogenomics Database (CTD; http://ctdbase.org/) is an innovative digital ecosystem that relates toxicological information for chemicals, genes, phenotypes, diseases, and exposures to advance understanding about human health. Literature-based, manually curated interactions are integrated to create a knowledgebase that harmonizes cross-species heterogeneous data for chemical exposures and their biological repercussions. In this biennial update, we report a 20% increase in CTD curated content and now provide 45 million toxicogenomic relationships for over 16 300 chemicals, 51 300 genes, 5500 phenotypes, 7200 diseases and 163 000 exposure events, from 600 comparative species. Furthermore, we increase the functionality of chemical-phenotype content with new data-tabs on CTD Disease pages (to help fill in knowledge gaps for environmental health) and new phenotype search parameters (for Batch Query and Venn analysis tools). As well, we introduce new CTD Anatomy pages that allow users to uniquely explore and analyze chemical-phenotype interactions from an anatomical perspective. Finally, we have enhanced CTD Chemical pages with new literature-based chemical synonyms (to improve querying) and added 1600 amino acid-based compounds (to increase chemical landscape). Together, these updates continue to augment CTD as a powerful resource for generating testable hypotheses about the etiologies and molecular mechanisms underlying environmentally influenced diseases.
Collapse
Affiliation(s)
- Allan Peter Davis
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Cynthia J Grondin
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Robin J Johnson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Daniela Sciaky
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Jolene Wiegers
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Thomas C Wiegers
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Carolyn J Mattingly
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
61
|
Yang M, Huang L, Xu Y, Lu C, Wang J. Heterogeneous graph inference with matrix completion for computational drug repositioning. Bioinformatics 2020; 36:5456-5464. [PMID: 33331887 DOI: 10.1093/bioinformatics/btaa1024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Emerging evidence presents that traditional drug discovery experiment is time-consuming and high costs. Computational drug repositioning plays a critical role in saving time and resources for drug research and discovery. Therefore, developing more accurate and efficient approaches is imperative. Heterogeneous graph inference is a classical method in computational drug repositioning, which not only has high convergence precision, but also has fast convergence speed. However, the method has not fully considered the sparsity of heterogeneous association network. In addition, rough similarity measure can reduce the performance in identifying drug-associated indications. RESULTS In this article, we propose a heterogeneous graph inference with matrix completion (HGIMC) method to predict potential indications for approved and novel drugs. First, we use a bounded matrix completion (BMC) model to prefill a part of the missing entries in original drug-disease association matrix. This step can add more positive and formative drug-disease edges between drug network and disease network. Second, Gaussian radial basis function (GRB) is employed to improve the drug and disease similarities since the performance of heterogeneous graph inference more relies on similarity measures. Next, based on the updated drug-disease associations and new similarity measures of drug and disease, we construct a novel heterogeneous drug-disease network. Finally, HGIMC utilizes the heterogeneous network to infer the scores of unknown association pairs, and then recommend the promising indications for drugs. To evaluate the performance of our method, HGIMC is compared with five state-of-the-art approaches of drug repositioning in the 10-fold cross-validation and de novo tests. As the numerical results shown, HGIMC not only achieves a better prediction performance, but also has an excellent computation efficiency. In addition, cases studies also confirm the effectiveness of our method in practical application. AVAILABILITY The HGIMC software is freely available at https://github.com/BioinformaticsCSU/HGIMC, https://hub.docker.com/repository/docker/yangmy84/hgimc, and http://doi.org/10.5281/zenodo.4285640. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mengyun Yang
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China.,School of Science, Shaoyang University, Shaoyang, P.R.China
| | - Lan Huang
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| | - Yunpei Xu
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| | - Chengqian Lu
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| | - Jianxin Wang
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| |
Collapse
|
62
|
Abstract
Background:
At present, using computer methods to predict drug-target interactions
(DTIs) is a very important step in the discovery of new drugs and drug relocation
processes. The potential DTIs identified by machine learning methods can provide guidance
in biochemical or clinical experiments.
Objective:
The goal of this article is to combine the latest network representation learning
methods for drug-target prediction research, improve model prediction capabilities, and
promote new drug development.
Methods:
We use large-scale information network embedding (LINE) method to extract
network topology features of drugs, targets, diseases, etc., integrate features obtained
from heterogeneous networks, construct binary classification samples, and use random
forest (RF) method to predict DTIs.
Results:
The experiments in this paper compare the common classifiers of RF, LR, and
SVM, as well as the typical network representation learning methods of LINE,
Node2Vec, and DeepWalk. It can be seen that the combined method LINE-RF achieves
the best results, reaching an AUC of 0.9349 and an AUPR of 0.9016.
Conclusion:
The learning method based on LINE network can effectively learn drugs,
targets, diseases and other hidden features from the network topology. The combination
of features learned through multiple networks can enhance the expression ability. RF is an
effective method of supervised learning. Therefore, the Line-RF combination method is a
widely applicable method.
Collapse
Affiliation(s)
- Jihong Wang
- School of Data and Computer Science, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yue Shi
- School of Data and Computer Science, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaodan Wang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, Guangdong, China
| | - Huiyou Chang
- School of Data and Computer Science, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
63
|
Aliashrafi M, Nasehi M, Zarrindast MR, Joghataei MT, Zali H, Siadat SD. Association of microbiota-derived propionic acid and Alzheimer's disease; bioinformatics analysis. J Diabetes Metab Disord 2020; 19:783-804. [PMID: 33553012 PMCID: PMC7843825 DOI: 10.1007/s40200-020-00564-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/06/2020] [Accepted: 06/02/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE Microbiota-derived metabolites could alter the brain tissue toward the neurodegeneration disease. This study aims to select the genes associated with Propionic acid (PPA) and compromise Alzheimer's disease (AD) to find the possible roles of PPA in AD pathogenesis. METHODS Microbiota-derived metabolites could alter the brain tissue toward the neurodegeneration disease. This study aims to select the genes associated with Propionic acid (PPA) and compromise Alzheimer's disease (AD) to find the possible roles of PPA in AD pathogenesis. RESULTS Amongst all genes associated with PPA and AD, 284 genes to be shared by searching databases and were subjected to further analysis. AD-PPA genes mainly involved in cancer, bacterial and virus infection, and neurological and non-neurological diseases. Gene Ontology and pathway analysis covered the most AD hallmark, such as amyloid formation, apoptosis, proliferation, inflammation, and immune system. Network analysis revealed hub and bottleneck genes. MCODE analysis also indicated the seed genes represented in the significant subnetworks. ICAM1 and CCND1 were the hub, bottleneck, and seed genes. CONCLUSIONS PPA interacted genes implicated in AD act through pathways initiate neuronal cell death. In sum up, AD-PPA shared genes exhibited evidence that supports the idea PPA secreted from bacteria could alter brain physiology toward the emerging AD signs. This idea needs to confirm by more future investigation in animal models.
Collapse
Affiliation(s)
- Morteza Aliashrafi
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran
- Shahid Beheshti University, Tehran, Iran
| | - Mohammad Nasehi
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran
- Cognitive and Neuroscience Research Center, Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neuroendocrinology, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Molecular and Cellular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Mycobacteriology & Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
64
|
Mohammadi E, Benfeitas R, Turkez H, Boren J, Nielsen J, Uhlen M, Mardinoglu A. Applications of Genome-Wide Screening and Systems Biology Approaches in Drug Repositioning. Cancers (Basel) 2020; 12:E2694. [PMID: 32967266 PMCID: PMC7563533 DOI: 10.3390/cancers12092694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
Modern drug discovery through de novo drug discovery entails high financial costs, low success rates, and lengthy trial periods. Drug repositioning presents a suitable approach for overcoming these issues by re-evaluating biological targets and modes of action of approved drugs. Coupling high-throughput technologies with genome-wide essentiality screens, network analysis, genome-scale metabolic modeling, and machine learning techniques enables the proposal of new drug-target signatures and uncovers unanticipated modes of action for available drugs. Here, we discuss the current issues associated with drug repositioning in light of curated high-throughput multi-omic databases, genome-wide screening technologies, and their application in systems biology/medicine approaches.
Collapse
Affiliation(s)
- Elyas Mohammadi
- Science for Life Laboratory, KTH–Royal Institute of Technology, SE-17121 Stockholm, Sweden; (E.M.); (M.U.)
- Department of Animal Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, SE-10691 Stockholm, Sweden;
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey;
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, SE-41345 Gothenburg, Sweden;
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-41296 Gothenburg, Sweden;
- BioInnovation Institute, DK-2200 Copenhagen N, Denmark
| | - Mathias Uhlen
- Science for Life Laboratory, KTH–Royal Institute of Technology, SE-17121 Stockholm, Sweden; (E.M.); (M.U.)
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH–Royal Institute of Technology, SE-17121 Stockholm, Sweden; (E.M.); (M.U.)
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
65
|
Peng J, Li J, Shang X. A learning-based method for drug-target interaction prediction based on feature representation learning and deep neural network. BMC Bioinformatics 2020; 21:394. [PMID: 32938374 PMCID: PMC7495825 DOI: 10.1186/s12859-020-03677-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Drug-target interaction prediction is of great significance for narrowing down the scope of candidate medications, and thus is a vital step in drug discovery. Because of the particularity of biochemical experiments, the development of new drugs is not only costly, but also time-consuming. Therefore, the computational prediction of drug target interactions has become an essential way in the process of drug discovery, aiming to greatly reducing the experimental cost and time. RESULTS We propose a learning-based method based on feature representation learning and deep neural network named DTI-CNN to predict the drug-target interactions. We first extract the relevant features of drugs and proteins from heterogeneous networks by using the Jaccard similarity coefficient and restart random walk model. Then, we adopt a denoising autoencoder model to reduce the dimension and identify the essential features. Third, based on the features obtained from last step, we constructed a convolutional neural network model to predict the interaction between drugs and proteins. The evaluation results show that the average AUROC score and AUPR score of DTI-CNN were 0.9416 and 0.9499, which obtains better performance than the other three existing state-of-the-art methods. CONCLUSIONS All the experimental results show that the performance of DTI-CNN is better than that of the three existing methods and the proposed method is appropriately designed.
Collapse
Affiliation(s)
- Jiajie Peng
- The School of Computer Science, Northwestern Polytechnical University, Xian, 710072, China.,The Key Laboratory of Big Data Storage an Management, Northwestern Polytechnical Universitythe, Ministry of Industry and Information Technology, Xian, 710072, China
| | - Jingyi Li
- The School of Computer Science, Northwestern Polytechnical University, Xian, 710072, China.,The Key Laboratory of Big Data Storage an Management, Northwestern Polytechnical Universitythe, Ministry of Industry and Information Technology, Xian, 710072, China
| | - Xuequn Shang
- The School of Computer Science, Northwestern Polytechnical University, Xian, 710072, China. .,The Key Laboratory of Big Data Storage an Management, Northwestern Polytechnical Universitythe, Ministry of Industry and Information Technology, Xian, 710072, China.
| |
Collapse
|
66
|
Bubier JA, He H, Philip VM, Roy T, Hernandez CM, Bernat R, Donohue KD, O'Hara BF, Chesler EJ. Genetic variation regulates opioid-induced respiratory depression in mice. Sci Rep 2020; 10:14970. [PMID: 32917924 PMCID: PMC7486296 DOI: 10.1038/s41598-020-71804-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
In the U.S., opioid prescription for treatment of pain nearly quadrupled from 1999 to 2014. The diversion and misuse of prescription opioids along with increased use of drugs like heroin and fentanyl, has led to an epidemic in addiction and overdose deaths. The most common cause of opioid overdose and death is opioid-induced respiratory depression (OIRD), a life-threatening depression in respiratory rate thought to be caused by stimulation of opioid receptors in the inspiratory-generating regions of the brain. Studies in mice have revealed that variation in opiate lethality is associated with strain differences, suggesting that sensitivity to OIRD is genetically determined. We first tested the hypothesis that genetic variation in inbred strains of mice influences the innate variability in opioid-induced responses in respiratory depression, recovery time and survival time. Using the founders of the advanced, high-diversity mouse population, the Diversity Outbred (DO), we found substantial sex and genetic effects on respiratory sensitivity and opiate lethality. We used DO mice treated with morphine to map quantitative trait loci for respiratory depression, recovery time and survival time. Trait mapping and integrative functional genomic analysis in GeneWeaver has allowed us to implicate Galnt11, an N-acetylgalactosaminyltransferase, as a gene that regulates OIRD.
Collapse
Affiliation(s)
| | - Hao He
- The Jackson Laboratory, Bar Harbor, ME, 04605, USA
| | | | - Tyler Roy
- The Jackson Laboratory, Bar Harbor, ME, 04605, USA
| | | | | | - Kevin D Donohue
- Signal Solutions, LLC, Lexington, KY, USA
- Electrical and Computer Engineering Department, University of Kentucky, Lexington, KY, USA
| | - Bruce F O'Hara
- Signal Solutions, LLC, Lexington, KY, USA
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
67
|
Perera N, Dehmer M, Emmert-Streib F. Named Entity Recognition and Relation Detection for Biomedical Information Extraction. Front Cell Dev Biol 2020; 8:673. [PMID: 32984300 PMCID: PMC7485218 DOI: 10.3389/fcell.2020.00673] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/02/2020] [Indexed: 12/29/2022] Open
Abstract
The number of scientific publications in the literature is steadily growing, containing our knowledge in the biomedical, health, and clinical sciences. Since there is currently no automatic archiving of the obtained results, much of this information remains buried in textual details not readily available for further usage or analysis. For this reason, natural language processing (NLP) and text mining methods are used for information extraction from such publications. In this paper, we review practices for Named Entity Recognition (NER) and Relation Detection (RD), allowing, e.g., to identify interactions between proteins and drugs or genes and diseases. This information can be integrated into networks to summarize large-scale details on a particular biomedical or clinical problem, which is then amenable for easy data management and further analysis. Furthermore, we survey novel deep learning methods that have recently been introduced for such tasks.
Collapse
Affiliation(s)
- Nadeesha Perera
- Predictive Society and Data Analytics Lab, Faculty of Information Technology and Communication Sciences, Tampere University, Tampere, Finland
| | - Matthias Dehmer
- Department of Mechatronics and Biomedical Computer Science, University for Health Sciences, Medical Informatics and Technology (UMIT), Hall in Tirol, Austria
- College of Artificial Intelligence, Nankai University, Tianjin, China
| | - Frank Emmert-Streib
- Predictive Society and Data Analytics Lab, Faculty of Information Technology and Communication Sciences, Tampere University, Tampere, Finland
- Faculty of Medicine and Health Technology, Institute of Biosciences and Medical Technology, Tampere University, Tampere, Finland
| |
Collapse
|
68
|
Drug repositioning based on the target microRNAs using bilateral-inductive matrix completion. Mol Genet Genomics 2020; 295:1305-1314. [DOI: 10.1007/s00438-020-01702-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
|
69
|
Deng S, Sun Y, Zhao T, Hu Y, Zang T. A Review of Drug Side Effect Identification Methods. Curr Pharm Des 2020; 26:3096-3104. [PMID: 32532187 DOI: 10.2174/1381612826666200612163819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/18/2020] [Indexed: 11/22/2022]
Abstract
Drug side effects have become an important indicator for evaluating the safety of drugs. There are two main factors in the frequent occurrence of drug safety problems; on the one hand, the clinical understanding of drug side effects is insufficient, leading to frequent adverse drug reactions, while on the other hand, due to the long-term period and complexity of clinical trials, side effects of approved drugs on the market cannot be reported in a timely manner. Therefore, many researchers have focused on developing methods to identify drug side effects. In this review, we summarize the methods of identifying drug side effects and common databases in this field. We classified methods of identifying side effects into four categories: biological experimental, machine learning, text mining and network methods. We point out the key points of each kind of method. In addition, we also explain the advantages and disadvantages of each method. Finally, we propose future research directions.
Collapse
Affiliation(s)
- Shuai Deng
- College of Science, Beijing Forestry University, Beijing, China
| | - Yige Sun
- Microbiology Department, Harbin Medical University, Harbin, 150081, China
| | - Tianyi Zhao
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yang Hu
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Tianyi Zang
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
70
|
Abstract
Motivation Computational drug repositioning is a cost-effective strategy to identify novel indications for existing drugs. Drug repositioning is often modeled as a recommendation system problem. Taking advantage of the known drug–disease associations, the objective of the recommendation system is to identify new treatments by filling out the unknown entries in the drug–disease association matrix, which is known as matrix completion. Underpinned by the fact that common molecular pathways contribute to many different diseases, the recommendation system assumes that the underlying latent factors determining drug–disease associations are highly correlated. In other words, the drug–disease matrix to be completed is low-rank. Accordingly, matrix completion algorithms efficiently constructing low-rank drug–disease matrix approximations consistent with known associations can be of immense help in discovering the novel drug–disease associations. Results In this article, we propose to use a bounded nuclear norm regularization (BNNR) method to complete the drug–disease matrix under the low-rank assumption. Instead of strictly fitting the known elements, BNNR is designed to tolerate the noisy drug–drug and disease–disease similarities by incorporating a regularization term to balance the approximation error and the rank properties. Moreover, additional constraints are incorporated into BNNR to ensure that all predicted matrix entry values are within the specific interval. BNNR is carried out on an adjacency matrix of a heterogeneous drug–disease network, which integrates the drug–drug, drug–disease and disease–disease networks. It not only makes full use of available drugs, diseases and their association information, but also is capable of dealing with cold start naturally. Our computational results show that BNNR yields higher drug–disease association prediction accuracy than the current state-of-the-art methods. The most significant gain is in prediction precision measured as the fraction of the positive predictions that are truly positive, which is particularly useful in drug design practice. Cases studies also confirm the accuracy and reliability of BNNR. Availability and implementation The code of BNNR is freely available at https://github.com/BioinformaticsCSU/BNNR. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mengyun Yang
- School of Computer Science and Engineering, Central South University, Changsha.,Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang, China
| | - Huimin Luo
- School of Computer Science and Engineering, Central South University, Changsha
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha
| |
Collapse
|
71
|
Bioinformatic Analysis Reveals Phosphodiesterase 4D-Interacting Protein as a Key Frontal Cortex Dementia Switch Gene. Int J Mol Sci 2020; 21:ijms21113787. [PMID: 32471155 PMCID: PMC7313474 DOI: 10.3390/ijms21113787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanisms that initiate dementia are poorly understood and there are currently no treatments that can slow their progression. The identification of key genes and molecular pathways that may trigger dementia should help reveal potential therapeutic reagents. In this study, SWItch Miner software was used to identify phosphodiesterase 4D-interacting protein as a key factor that may lead to the development of Alzheimer’s disease, vascular dementia, and frontotemporal dementia. Inflammation, PI3K-AKT, and ubiquitin-mediated proteolysis were identified as the main pathways that are dysregulated in these dementias. All of these dementias are regulated by 12 shared transcription factors. Protein–chemical interaction network analysis of dementia switch genes revealed that valproic acid may be neuroprotective for these dementias. Collectively, we identified shared and unique dysregulated gene expression, pathways and regulatory factors among dementias. New key mechanisms that lead to the development of dementia were revealed and it is expected that these data will advance personalized medicine for patients.
Collapse
|
72
|
Gomez-Cadena A, Barreto A, Fioretino S, Jandus C. Immune system activation by natural products and complex fractions: a network pharmacology approach in cancer treatment. Cell Stress 2020; 4:154-166. [PMID: 32656498 PMCID: PMC7328673 DOI: 10.15698/cst2020.07.224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Natural products and traditional herbal medicine are an important source of alternative bioactive compounds but very few plant-based preparations have been scientifically evaluated and validated for their potential as medical treatments. However, a promising field in the current therapies based on plant-derived compounds is the study of their immunomodulation properties and their capacity to activate the immune system to fight against multifactorial diseases like cancer. In this review we discuss how network pharmacology could help to characterize and validate natural single molecules or more complex preparations as promising cancer therapies based on their multitarget capacities.
Collapse
Affiliation(s)
- Alejandra Gomez-Cadena
- Department of Pathology and Immunology, Targeting of Cytokine Secreting Lymphocyte group, Geneva University, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Switzerland.,Departamento de Microbiología, Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Departamento de Microbiología, Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Susana Fioretino
- Departamento de Microbiología, Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Camilla Jandus
- Department of Pathology and Immunology, Targeting of Cytokine Secreting Lymphocyte group, Geneva University, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Switzerland
| |
Collapse
|
73
|
Davis AP, Grondin CJ, Johnson RJ, Sciaky D, McMorran R, Wiegers J, Wiegers TC, Mattingly CJ. The Comparative Toxicogenomics Database: update 2019. Nucleic Acids Res 2020; 47:D948-D954. [PMID: 30247620 PMCID: PMC6323936 DOI: 10.1093/nar/gky868] [Citation(s) in RCA: 589] [Impact Index Per Article: 147.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/14/2018] [Indexed: 11/27/2022] Open
Abstract
The Comparative Toxicogenomics Database (CTD; http://ctdbase.org/) is a premier public resource for literature-based, manually curated associations between chemicals, gene products, phenotypes, diseases, and environmental exposures. In this biennial update, we present our new chemical–phenotype module that codes chemical-induced effects on phenotypes, curated using controlled vocabularies for chemicals, phenotypes, taxa, and anatomical descriptors; this module provides unique opportunities to explore cellular and system-level phenotypes of the pre-disease state and allows users to construct predictive adverse outcome pathways (linking chemical–gene molecular initiating events with phenotypic key events, diseases, and population-level health outcomes). We also report a 46% increase in CTD manually curated content, which when integrated with other datasets yields more than 38 million toxicogenomic relationships. We describe new querying and display features for our enhanced chemical–exposure science module, providing greater scope of content and utility. As well, we discuss an updated MEDIC disease vocabulary with over 1700 new terms and accession identifiers. To accommodate these increases in data content and functionality, CTD has upgraded its computational infrastructure. These updates continue to improve CTD and help inform new testable hypotheses about the etiology and mechanisms underlying environmentally influenced diseases.
Collapse
Affiliation(s)
- Allan Peter Davis
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Cynthia J Grondin
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Robin J Johnson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Daniela Sciaky
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Roy McMorran
- Department of Bioinformatics, The Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, USA
| | - Jolene Wiegers
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Thomas C Wiegers
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Carolyn J Mattingly
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
74
|
Tan X, Tang H, Gong L, Xie L, Lei Y, Luo Z, He C, Ma J, Han S. Integrating Genome-Wide Association Studies and Gene Expression Profiles With Chemical-Genes Interaction Networks to Identify Chemicals Associated With Colorectal Cancer. Front Genet 2020; 11:385. [PMID: 32391058 PMCID: PMC7193025 DOI: 10.3389/fgene.2020.00385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and has the second highest mortality rate in global cancer. Exploring the associations between chemicals and CRC has great significance in prophylaxis and therapy of tumor diseases. This study aims to explore the relationships between CRC and environmental chemicals on genetic basis by bioinformatics analysis. The genome-wide association study (GWAS) datasets for CRC were obtained from the UK Biobank. The GWAS data for colon cancer (category C18) includes 2,581 individuals and 449,683 controls, while that of rectal cancer (category C20) includes 1,244 individuals and 451,020 controls. In addition, we derived CRC gene expression datasets from the NCBI-GEO (GSE106582). The chemicals related gene sets were acquired from the comparative toxicogenomics database (CTD). Transcriptome-wide association study (TWAS) analysis was applied to CRC GWAS summary data and calculated the expression association testing statistics by FUSION software. We performed chemicals related gene set enrichment analysis (GSEA) by integrating GWAS summary data, mRNA expression profiles of CRC and the CTD chemical-gene interaction networks to identify relationships between chemicals and genes of CRC. We observed several significant correlations between chemicals and CRC. Meanwhile, we also detected 5 common chemicals between colon and rectal cancer, including methylnitronitrosoguanidine, isoniazid, PD 0325901, sulindac sulfide, and importazole. Our study performed TWAS and GSEA analysis, linked prior knowledge to newly generated data and thereby helped identifying chemicals related to tumor genes, which provides new clues for revealing the associations between environmental chemicals and cancer.
Collapse
Affiliation(s)
- Xinyue Tan
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hanmin Tang
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liuyun Gong
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lina Xie
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yutiantian Lei
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhenzhen Luo
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Chenchen He
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jinlu Ma
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Suxia Han
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
75
|
Ni P, Wang J, Zhong P, Li Y, Wu FX, Pan Y. Constructing Disease Similarity Networks Based on Disease Module Theory. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:906-915. [PMID: 29993782 DOI: 10.1109/tcbb.2018.2817624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Quantifying the associations between diseases is now playing an important role in modern biology and medicine. Actually discovering associations between diseases could help us gain deeper insights into pathogenic mechanisms of complex diseases, thus could lead to improvements in disease diagnosis, drug repositioning, and drug development. Due to the growing body of high-throughput biological data, a number of methods have been developed for computing similarity between diseases during the past decade. However, these methods rarely consider the interconnections of genes related to each disease in protein-protein interaction network (PPIN). Recently, the disease module theory has been proposed, which states that disease-related genes or proteins tend to interact with each other in the same neighborhood of a PPIN. In this study, we propose a new method called ModuleSim to measure associations between diseases by using disease-gene association data and PPIN data based on disease module theory. The experimental results show that by considering the interactions between disease modules and their modularity, the disease similarity calculated by ModuleSim has a significant correlation with disease classification of Disease Ontology (DO). Furthermore, ModuleSim outperforms other four popular methods which are all using disease-gene association data and PPIN data to measure disease-disease associations. In addition, the disease similarity network constructed by MoudleSim suggests that ModuleSim is capable of finding potential associations between diseases.
Collapse
|
76
|
Liu L, He D, Wang Y, Sheng M. Integrated analysis of DNA methylation and transcriptome profiling of polycystic ovary syndrome. Mol Med Rep 2020; 21:2138-2150. [PMID: 32323770 PMCID: PMC7115196 DOI: 10.3892/mmr.2020.11005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to identify potentially important biomarkers associated with polycystic ovary syndrome (PCOS) by integrating DNA methylation with transcriptome profiling. The transcription (E‑MTAB‑3768) and methylation (E‑MTAB‑3777) datasets were retrieved from ArrayExpress. Paired transcription and methylation profiling data of 10 cases of PCOS and 10 healthy controls were available for screening differentially expressed genes (DEGs) and differentially methylated genes (DMGs). Genes with a negative correlation between expression levels and methylation levels were retained by correlation analysis to construct a protein‑protein interaction (PPI) network. Subsequently, functional and pathway enrichment analyses were performed to identify genes in the PPI network. Additionally, a disease‑associated pathway network was also established. A total of 491 overlapping genes, and the expression levels of 237 genes, were negatively correlated with their methylation levels. Functional enrichment analysis revealed that genes in the PPI network were mainly involved with biological processes of cellular response to stress, negative regulation of the biosynthetic process, and regulation of cell proliferation. The constructed pathway network associated with PCOS led to the identification of four important genes (SPP1, F2R, IL12B and RBP4) and two important pathways (Jak‑STAT signaling pathway and neuroactive ligand‑receptor interaction). Taken, together, the results from the present study have revealed numerous important genes with abnormal DNA methylation levels and altered mRNA expression levels, along with their associated functions and pathways. These findings may contribute to an improved understanding of the possible pathophysiology of PCOS.
Collapse
Affiliation(s)
- Li Liu
- Reproductive Medical Center, Department of Gynecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Dongyun He
- Reproductive Medical Center, Department of Gynecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Yang Wang
- Department of Dermatology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130031, P.R. China
| | - Minjia Sheng
- Reproductive Medical Center, Department of Gynecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| |
Collapse
|
77
|
Huang F, Qiu Y, Li Q, Liu S, Ni F. Predicting Drug-Disease Associations via Multi-Task Learning Based on Collective Matrix Factorization. Front Bioeng Biotechnol 2020; 8:218. [PMID: 32373595 PMCID: PMC7179666 DOI: 10.3389/fbioe.2020.00218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/04/2020] [Indexed: 12/30/2022] Open
Abstract
Identifying drug-disease associations is integral to drug development. Computationally prioritizing candidate drug-disease associations has attracted growing attention due to its contribution to reducing the cost of laboratory screening. Drug-disease associations involve different association types, such as drug indications and drug side effects. However, the existing models for predicting drug-disease associations merely concentrate on independent tasks: recommending novel indications to benefit drug repositioning, predicting potential side effects to prevent drug-induced risk, or only determining the existence of drug-disease association. They ignore crucial prior knowledge of the correlations between different association types. Since the Comparative Toxicogenomics Database (CTD) annotates the drug-disease associations as therapeutic or marker/mechanism, we consider predicting the two types of association. To this end, we propose a collective matrix factorization-based multi-task learning method (CMFMTL) in this paper. CMFMTL handles the problem as multi-task learning where each task is to predict one type of association, and two tasks complement and improve each other by capturing the relatedness between them. First, drug-disease associations are represented as a bipartite network with two types of links representing therapeutic effects and non-therapeutic effects. Then, CMFMTL, respectively, approximates the association matrix regarding each link type by matrix tri-factorization, and shares the low-dimensional latent representations for drugs and diseases in the two related tasks for the goal of collective learning. Finally, CMFMTL puts the two tasks into a unified framework and an efficient algorithm is developed to solve our proposed optimization problem. In the computational experiments, CMFMTL outperforms several state-of-the-art methods both in the two tasks. Moreover, case studies show that CMFMTL helps to find out novel drug-disease associations that are not included in CTD, and simultaneously predicts their association types.
Collapse
Affiliation(s)
- Feng Huang
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Yang Qiu
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Qiaojun Li
- College of Informatics, Huazhong Agricultural University, Wuhan, China
- School of Electronic and Information Engineering, Henan Polytechnic Institute, Henan Nanyang, China
| | - Shichao Liu
- College of Informatics, Huazhong Agricultural University, Wuhan, China
- Hubei Engineering Technology Research Center of Agricultural Big Data, Wuhan, China
| | - Fuchuan Ni
- College of Informatics, Huazhong Agricultural University, Wuhan, China
- Hubei Engineering Technology Research Center of Agricultural Big Data, Wuhan, China
| |
Collapse
|
78
|
Tian R, Wang L, Zou H, Song M, Liu L, Zhang H. Role of the XIST-miR-181a-COL4A1 axis in the development and progression of keratoconus. Mol Vis 2020; 26:1-13. [PMID: 32165822 PMCID: PMC7043645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 01/31/2020] [Indexed: 10/26/2022] Open
Abstract
Background As a disorder occurs in the eyes, keratoconus (KC) is induced by the thinning of the corneal stroma. This study was designed to reveal the key long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and mRNAs involved in the mechanisms of KC. Methods Transcriptome RNA-seq data set GSE112155 was acquired from the Gene Expression Omnibus database, which contained 10 KC samples and 10 myopic control samples. Using the edgeR package, the differentially expressed (DE)-mRNAs between KC and control samples were screened. The DE-lncRNAs and DE-miRNAs in this data set were identified using the HUGO Gene Nomenclature Committee (HGNC). Using the pheatmap package, bidirectional hierarchical clustering of the DE-RNAs was conducted. Then, an enrichment analysis of the DE-mRNAs was performed using the DAVID tool. Moreover, a competitive endogenous RNA (ceRNA) regulatory network was built using the Cytoscape software. After KC-associated pathways were searched within the Comparative Toxicogenomics Database, a KC-associated ceRNA regulatory network was constructed. Results There were 282 DE-lncRNAs (192 upregulated and 90 downregulated), 40 DE-miRNAs (29 upregulated and 11 downregulated), and 910 DE-mRNAs (554 upregulated and 356 downregulated) between the KC and control samples. A total of 34 functional terms and 9 pathways were enriched for the DE-mRNAs. In addition, 6 mRNAs (including PPARG, HLA-B, COL4A1, and COL4A2), 5 miRNAs (including miR-181a), 9 lncRNAs (including XIST), and the XIST-miR-181a-COL4A1 axis were involved in the KC-associated ceRNA regulatory network. Conclusions PPARG, HLA-B, COL4A1, COL4A2, miR-181a, and XIST might be correlated with the development of KC. Further, the XIST-miR-181a-COL4A1 axis might be implicated in the pathogenesis of KC.
Collapse
Affiliation(s)
- Rui Tian
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Lufei Wang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - He Zou
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Meijiao Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Lu Liu
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Hui Zhang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| |
Collapse
|
79
|
Ma R, Li Y, Li C, Wan F, Hu H, Xu W, Zeng J. Secure multiparty computation for privacy-preserving drug discovery. Bioinformatics 2020; 36:2872-2880. [DOI: 10.1093/bioinformatics/btaa038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 01/24/2023] Open
Abstract
Abstract
Motivation
Quantitative structure–activity relationship (QSAR) and drug–target interaction (DTI) prediction are both commonly used in drug discovery. Collaboration among pharmaceutical institutions can lead to better performance in both QSAR and DTI prediction. However, the drug-related data privacy and intellectual property issues have become a noticeable hindrance for inter-institutional collaboration in drug discovery.
Results
We have developed two novel algorithms under secure multiparty computation (MPC), including QSARMPC and DTIMPC, which enable pharmaceutical institutions to achieve high-quality collaboration to advance drug discovery without divulging private drug-related information. QSARMPC, a neural network model under MPC, displays good scalability and performance and is feasible for privacy-preserving collaboration on large-scale QSAR prediction. DTIMPC integrates drug-related heterogeneous network data and accurately predicts novel DTIs, while keeping the drug information confidential. Under several experimental settings that reflect the situations in real drug discovery scenarios, we have demonstrated that DTIMPC possesses significant performance improvement over the baseline methods, generates novel DTI predictions with supporting evidence from the literature and shows the feasible scalability to handle growing DTI data. All these results indicate that QSARMPC and DTIMPC can provide practically useful tools for advancing privacy-preserving drug discovery.
Availability and implementation
The source codes of QSARMPC and DTIMPC are available on the GitHub: https://github.com/rongma6/QSARMPC_DTIMPC.git.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Rong Ma
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Yi Li
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Chenxing Li
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Fangping Wan
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Hailin Hu
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wei Xu
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Jianyang Zeng
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
80
|
Shi H, Sun H, Li J, Bai Z, Wu J, Li X, Lv Y, Zhang G. Systematic analysis of lncRNA and microRNA dynamic features reveals diagnostic and prognostic biomarkers of myocardial infarction. Aging (Albany NY) 2020; 12:945-964. [PMID: 31927529 PMCID: PMC6977700 DOI: 10.18632/aging.102667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022]
Abstract
Analyses of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) implicated in myocardial infarction (MI) have increased our understanding of gene regulatory mechanisms in MI. However, it is not known how their expression fluctuates over the different stages of MI progression. In this study, we used time-series gene expression data to examine global lncRNA and miRNA expression patterns during the acute phase of MI and at three different time points thereafter. We observed that the largest expression peak for mRNAs, lncRNAs, and miRNAs occurred during the acute phase of MI and involved mainly protein-coding, rather than non-coding RNAs. Functional analysis indicated that the lncRNAs and miRNAs most sensitive to MI and most unstable during MI progression were usually related to fewer biological functions. Additionally, we developed a novel computational method for identifying dysregulated competing endogenous lncRNA-miRNA-mRNA triplets (LmiRM-CTs) during MI onset and progression. As a result, a new panel of candidate diagnostic biomarkers defined by seven lncRNAs was suggested to have high classification performance for patients with or without MI, and a new panel of prognostic biomarkers defined by two lncRNAs evidenced high discriminatory capability for MI patients who developed heart failure from those who did not.
Collapse
Affiliation(s)
- Hongbo Shi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Haoran Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiayao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyi Bai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jie Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiuhong Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingli Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Guangde Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
81
|
Ma S, Yang K, Wang N, Zhu Q, Gao Z, Zhang R, Liu B, Zhou X. Disease phenotype synonymous prediction through network representation learning from PubMed database. Artif Intell Med 2020; 102:101745. [DOI: 10.1016/j.artmed.2019.101745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 03/04/2019] [Accepted: 10/23/2019] [Indexed: 11/15/2022]
|
82
|
Yang M, Luo H, Li Y, Wu FX, Wang J. Overlap matrix completion for predicting drug-associated indications. PLoS Comput Biol 2019; 15:e1007541. [PMID: 31869322 PMCID: PMC6946175 DOI: 10.1371/journal.pcbi.1007541] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 01/07/2020] [Accepted: 11/12/2019] [Indexed: 11/19/2022] Open
Abstract
Identification of potential drug–associated indications is critical for either approved or novel drugs in drug repositioning. Current computational methods based on drug similarity and disease similarity have been developed to predict drug–disease associations. When more reliable drug- or disease-related information becomes available and is integrated, the prediction precision can be continuously improved. However, it is a challenging problem to effectively incorporate multiple types of prior information, representing different characteristics of drugs and diseases, to identify promising drug–disease associations. In this study, we propose an overlap matrix completion (OMC) for bilayer networks (OMC2) and tri-layer networks (OMC3) to predict potential drug-associated indications, respectively. OMC is able to efficiently exploit the underlying low-rank structures of the drug–disease association matrices. In OMC2, first of all, we construct one bilayer network from drug-side aspect and one from disease-side aspect, and then obtain their corresponding block adjacency matrices. We then propose the OMC2 algorithm to fill out the values of the missing entries in these two adjacency matrices, and predict the scores of unknown drug–disease pairs. Moreover, we further extend OMC2 to OMC3 to handle tri-layer networks. Computational experiments on various datasets indicate that our OMC methods can effectively predict the potential drug–disease associations. Compared with the other state-of-the-art approaches, our methods yield higher prediction accuracy in 10-fold cross-validation and de novo experiments. In addition, case studies also confirm the effectiveness of our methods in identifying promising indications for existing drugs in practical applications. This work introduces a computational approach, namely overlap matrix completion (OMC), to predict potential associations between drugs and diseases. The novelty of OMC lies in constructing an efficient framework of incorporating multiple types of prior information in bilayer and tri-layer networks. OMC for bilayer networks (OMC2) can approximate the low-rank structures of the drug–disease association matrices from both drug-side and disease-side. In addition, we further improve the prediction accuracy by extending OMC to handle tri-layer networks and develop its corresponding algorithm (OMC3). To evaluate the performance of OMC2 and OMC3, we conduct 10-fold cross-validation and de novo experiments on three datasets. Our computational results demonstrate that both OMC2 and OMC3 generally outperform five state-of-the-art methods in terms of ROC curve, PR curve, and top-ranked predictions.
Collapse
Affiliation(s)
- Mengyun Yang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R. China
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang, Hunan, P.R. China
| | - Huimin Luo
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R. China
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, Virginia, United States of America
| | - Fang-Xiang Wu
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R. China
- * E-mail:
| |
Collapse
|
83
|
Wang R, Li S, Cheng L, Wong MH, Leung KS. Predicting associations among drugs, targets and diseases by tensor decomposition for drug repositioning. BMC Bioinformatics 2019; 20:628. [PMID: 31839008 PMCID: PMC6912989 DOI: 10.1186/s12859-019-3283-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Development of new drugs is a time-consuming and costly process, and the cost is still increasing in recent years. However, the number of drugs approved by FDA every year per dollar spent on development is declining. Drug repositioning, which aims to find new use of existing drugs, attracts attention of pharmaceutical researchers due to its high efficiency. A variety of computational methods for drug repositioning have been proposed based on machine learning approaches, network-based approaches, matrix decomposition approaches, etc. RESULTS: We propose a novel computational method for drug repositioning. We construct and decompose three-dimensional tensors, which consist of the associations among drugs, targets and diseases, to derive latent factors reflecting the functional patterns of the three kinds of entities. The proposed method outperforms several baseline methods in recovering missing associations. Most of the top predictions are validated by literature search and computational docking. Latent factors are used to cluster the drugs, targets and diseases into functional groups. Topological Data Analysis (TDA) is applied to investigate the properties of the clusters. We find that the latent factors are able to capture the functional patterns and underlying molecular mechanisms of drugs, targets and diseases. In addition, we focus on repurposing drugs for cancer and discover not only new therapeutic use but also adverse effects of the drugs. In the in-depth study of associations among the clusters of drugs, targets and cancer subtypes, we find there exist strong associations between particular clusters. CONCLUSIONS The proposed method is able to recover missing associations, discover new predictions and uncover functional clusters of drugs, targets and diseases. The clustering of drugs, targets and diseases, as well as the associations among the clusters, provides a new guiding framework for drug repositioning.
Collapse
Affiliation(s)
- Ran Wang
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuai Li
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Lixin Cheng
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medicine College of Ji’nan University, Shenzhen, China
| | - Man Hon Wong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwong Sak Leung
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
84
|
Noori E, Kazemi B, Bandehpour M, Zali H, Khalesi B, Khalili S. Deciphering crucial genes in coeliac disease by bioinformatics analysis. Autoimmunity 2019; 53:102-113. [PMID: 31809599 DOI: 10.1080/08916934.2019.1698552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coeliac disease (CD) is a chronic autoimmune disease that is characterized by malabsorption in sensitive individuals. CD is triggered by the ingestion of grains containing gluten. CD is concomitant with several other disorders, including dermatitis herpetiformis, selective IgA deficiency, thyroid disorders, diabetes mellitus, various connective tissue disorders, inflammatory bowel disease, and rheumatoid arthritis. The advent of high throughput technologies has provided a massive wealth of data which are processed in various omics scale fields. These approaches have revolutionized the medical research and monitoring of the biological systems. In this regard, omics scaled analyses of CD by Comparative Toxicogenomics Database (CTD), DISEASES, and GeneCards databases have retrieved 2656 CD associated genes. Amongst, 54 genes were assigned by Venn Diagram of the intersection to be shared by these 3 databases for CD. These common genes were subjected to further analysis and screening. The Enrich database, GeneMANIA, Cytoscape, and WebGestalt (WEB-based GEne SeT AnaLysis Toolkit) were employed for functional analysis. These analyses indicated that the obtained genes are mainly involved in the immune system and signalling pathways related to autoimmune diseases. The STAT1, ALB, IL10, IL2, IL4, IL17A, TGFB1, IL1B, IL6, TNF, IFNG hub genes were particularly indicated to have significant roles in CD. Functional analyses of these hub genes by GeneMANIA indicated that they are involved in immune systems regulation. Moreover, 25 out of 54 genes were identified to be seed genes by the WebGestalt database. Gene set analysis with GEO2R tool from Gene Expression Omnibus (GEO) showed that there were 15 significant genes in GSE76168, 29 significant genes in GSE87460, 12 significant genes in GSE87458, 9 significant genes in GSE87457, 3753 significant genes in GSE112102 and 1043 significant genes in GSE102991 with differential expression in coeliac patients compared to controls. The IRF1and STAT1 genes were common between the significant genes from GEO and the 54 CD related genes from three public databases. In the light these results, nine key genes, including IRF1, STAT1, IL17A, TGFB1, ALB, IL10, IL2, IL4, and IL1B, were identified to be associated with CD. These findings could be used to find novel diagnostic biomarkers, understand the pathology of disease, and devise more efficient treatments.
Collapse
Affiliation(s)
- Effat Noori
- Department of Biotechnology School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandehpour
- Department of Biotechnology School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute Agriculture Research Education and Extension Organization(AREEO), Karaj, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
85
|
Gu J, Zhu H, Zhu D, Li M, Xiao M, Yan D, Shen S. VWF, CXCL8 and IL6 might be potential druggable genes for acute coronary syndrome (ACS). Comput Biol Chem 2019; 83:107125. [DOI: 10.1016/j.compbiolchem.2019.107125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 01/31/2023]
|
86
|
Hu Y, Zhao T, Zhang N, Zhang Y, Cheng L. A Review of Recent Advances and Research on Drug Target Identification Methods. Curr Drug Metab 2019; 20:209-216. [PMID: 30251599 DOI: 10.2174/1389200219666180925091851] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 01/01/2018] [Accepted: 08/02/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND From a therapeutic viewpoint, understanding how drugs bind and regulate the functions of their target proteins to protect against disease is crucial. The identification of drug targets plays a significant role in drug discovery and studying the mechanisms of diseases. Therefore the development of methods to identify drug targets has become a popular issue. METHODS We systematically review the recent work on identifying drug targets from the view of data and method. We compiled several databases that collect data more comprehensively and introduced several commonly used databases. Then divided the methods into two categories: biological experiments and machine learning, each of which is subdivided into different subclasses and described in detail. RESULTS Machine learning algorithms are the majority of new methods. Generally, an optimal set of features is chosen to predict successful new drug targets with similar properties. The most widely used features include sequence properties, network topological features, structural properties, and subcellular locations. Since various machine learning methods exist, improving their performance requires combining a better subset of features and choosing the appropriate model for the various datasets involved. CONCLUSION The application of experimental and computational methods in protein drug target identification has become increasingly popular in recent years. Current biological and computational methods still have many limitations due to unbalanced and incomplete datasets or imperfect feature selection methods.
Collapse
Affiliation(s)
- Yang Hu
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Tianyi Zhao
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ningyi Zhang
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ying Zhang
- Department of Pharmacy, Heilongjiang Province Land Reclamation Headquarters General Hospital, Harbin 150088, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
87
|
Network Pharmacology of Yougui Pill Combined with Buzhong Yiqi Decoction for the Treatment of Sexual Dysfunction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1243743. [PMID: 31814838 PMCID: PMC6877955 DOI: 10.1155/2019/1243743] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/10/2019] [Indexed: 12/16/2022]
Abstract
Purpose We aimed to find the possible key targets of Yougui pill and Buzhong Yiqi decoction for the treatment of sexual dysfunction. Materials and Methods The composition of Yougui pill combined with Buzhong Yiqi decoction was obtained, and its effective components of medicine were screened using ADME; the component target proteins were predicted and screened based on the TCMSP and BATMAN databases. Target proteins were cross-validated using the CTD database. We performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses for target proteins using the Cytoscape plugin ClueGO + CluePedia and the R package clusterProfiler, respectively. Subsequently, protein-protein interaction (PPI) analyses were conducted using the STRING database. Finally, a pharmacological network was constructed. Results The pharmacological network contained 89 nodes and 176 relation pairs. Among these nodes, there were 12 for herbal medicines (orange peel, licorice, Eucommia, Aconite, Astragalus, Chinese wolfberry, yam, dodder seed, ginseng, Cornus officinalis, Rehmannia, and Angelica), 9 for chemical components (18-beta-glycyrrhetinic acid, carvacrol, glycyrrhetinic acid, higenamine, nobilin, quercetin, stigmasterol, synephrine, and thymol), 62 for target proteins (e.g., NR3C1, ESR1, PTGS2, CAT, TNF, INS, and TP53), and 6 for pathways (MAPK signaling pathway, proteoglycans in cancer, dopaminergic synapse, thyroid hormone signaling pathway, cAMP signaling pathway, and neuroactive ligand-receptor interaction). Conclusion NR3C1, ESR1, PTGS2, CAT, TNF, INS, and TP53 may be important targets for the key active elements in the decoction combining Yougui pill and Buzhong Yiqi. Furthermore, these target proteins are relevant to the treatment of sexual dysfunction, probably via pathways associated with cancer and signal transduction.
Collapse
|
88
|
Kosnik MB, Planchart A, Marvel SW, Reif DM, Mattingly CJ. Integration of curated and high-throughput screening data to elucidate environmental influences on disease pathways. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 12:100094. [PMID: 31453412 PMCID: PMC6709694 DOI: 10.1016/j.comtox.2019.100094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Addressing the complex relationship between public health and environmental exposure requires multiple types and sources of data. An important source of chemical data derives from high-throughput screening (HTS) efforts, such as the Tox21/ToxCast program, which aim to identify chemical hazard using primarily in vitro assays to probe toxicity. While most of these assays target specific genes, assessing the disease-relevance of these assays remains challenging. Integration with additional data sets may help to resolve these questions by providing broader context for individual assay results. The Comparative Toxicogenomics Database (CTD), a publicly available database that builds networks of chemical, gene, and disease information from manually curated literature sources, offers a promising solution for contextual integration with HTS data. Here, we tested the value of integrating data across Tox21/ToxCast and CTD by linking elements common to both databases (i.e., assays, genes, and chemicals). Using polymarcine and Parkinson's disease as a case study, we found that their union significantly increased chemical-gene associations and disease-pathway coverage. Integration also enabled new disease associations to be made with HTS assays, expanding coverage of chemical-gene data associated with diseases. We demonstrate how integration enables development of predictive adverse outcome pathways using 4-nonylphenol, branched as an example. Thus, we demonstrate enhancements to each data source through database integration, including scenarios where HTS data can efficiently probe chemical space that may be understudied in the literature, as well as how CTD can add biological context to those results.
Collapse
Affiliation(s)
- Marissa B. Kosnik
- Toxicology Program, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Bioinformatics Research Center, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Department of Biological Sciences, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
| | - Antonio Planchart
- Toxicology Program, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Department of Biological Sciences, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7617, United States
| | - Skylar W. Marvel
- Bioinformatics Research Center, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Department of Biological Sciences, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
| | - David M. Reif
- Toxicology Program, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Bioinformatics Research Center, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Department of Biological Sciences, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7617, United States
| | - Carolyn J. Mattingly
- Toxicology Program, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Department of Biological Sciences, North Carolina State University, North Carolina State University, Raleigh, NC 27695-7617, United States
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7617, United States
| |
Collapse
|
89
|
Zhou W, Chen Z, Wang Y, Li X, Lu A, Sun X, Liu Z. Systems Pharmacology-Based Method to Assess the Mechanism of Action of Weight-Loss Herbal Intervention Therapy for Obesity. Front Pharmacol 2019; 10:1165. [PMID: 31680953 PMCID: PMC6802489 DOI: 10.3389/fphar.2019.01165] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is a multi-factorial chronic disease that has become a serious, prevalent, and refractory public health challenge globally because of high rates of various complications. Traditional Chinese medicines (TCMs) as a functional food are considered to be a valuable and readily available resource for treating obesity because of their better therapeutic effects and reduced side effects. However, their "multi-compound" and "multi-target" features make it extremely difficult to interpret the potential mechanism underlying the anti-obesity effects of TCMs from a holistic perspective. An innovative systems-pharmacology approach was employed, which combined absorption, distribution, metabolism, and excretion screening and multiple target fishing, gene ontology enrichment analysis, network pharmacology, and pathway analysis to explore the potential therapeutic mechanism of weight-loss herbal intervention therapy in obesity and related diseases. The current study provides a promising approach to facilitate the development and discovery of new botanical drugs.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Respirology and Allergy, The Third Affiliated Hospital of ShenZhen University, Shenzhen, China
- School of Basic Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ziyi Chen
- Department of Respirology and Allergy, The Third Affiliated Hospital of ShenZhen University, Shenzhen, China
| | - Yonghua Wang
- College of Life Sciences, Northwest University, Xi’an, China
| | - Xiumin Li
- Department of Respirology and Allergy, The Third Affiliated Hospital of ShenZhen University, Shenzhen, China
- School of Basic Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Xizhuo Sun
- Department of Respirology and Allergy, The Third Affiliated Hospital of ShenZhen University, Shenzhen, China
| | - Zhigang Liu
- Department of Respirology and Allergy, The Third Affiliated Hospital of ShenZhen University, Shenzhen, China
- School of Basic Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
90
|
Corsi SR, De Cicco LA, Villeneuve DL, Blackwell BR, Fay KA, Ankley GT, Baldwin AK. Prioritizing chemicals of ecological concern in Great Lakes tributaries using high-throughput screening data and adverse outcome pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 686:995-1009. [PMID: 31412529 DOI: 10.1016/j.scitotenv.2019.05.457] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 04/15/2023]
Abstract
Chemical monitoring data were collected in surface waters from 57 Great Lakes tributaries from 2010 to 13 to identify chemicals of potential biological relevance and sites at which these chemicals occur. Traditional water-quality benchmarks for aquatic life based on in vivo toxicity data were available for 34 of 67 evaluated chemicals. To expand evaluation of potential biological effects, measured chemical concentrations were compared to chemical-specific biological activities determined in high-throughput (ToxCast) in vitro assays. Resulting exposure-activity ratios (EARs) were used to prioritize the chemicals of greatest potential concern: 4‑nonylphenol, bisphenol A, metolachlor, atrazine, DEET, caffeine, tris(2‑butoxyethyl) phosphate, tributyl phosphate, triphenyl phosphate, benzo(a)pyrene, fluoranthene, and benzophenone. Water-quality benchmarks were unavailable for five of these chemicals, but for the remaining seven, EAR-based prioritization was consistent with that based on toxicity quotients calculated from benchmarks. Water-quality benchmarks identified three additional PAHs (anthracene, phenanthrene, and pyrene) not prioritized using EARs. Through this analysis, an EAR of 10-3 was identified as a reasonable threshold above which a chemical might be of potential concern. To better understand apical hazards potentially associated with biological activities captured in ToxCast assays, in vitro bioactivity data were matched with available adverse outcome pathway (AOP) information. The 49 ToxCast assays prioritized via EAR analysis aligned with 23 potentially-relevant AOPs present in the AOP-Wiki. Mixture effects at monitored sites were estimated by summation of EAR values for multiple chemicals by individual assay or individual AOP. Commonly predicted adverse outcomes included impacts on reproduction and mitochondrial function. The EAR approach provided a screening-level assessment for evidence-based prioritization of chemicals and sites with potential for adverse biological effects. The approach aids prioritization of future monitoring activities and provides testable hypotheses to help focus those efforts. This also expands the fraction of detected chemicals for which biologically-based benchmark concentrations are available to help contextualize chemical monitoring results.
Collapse
Affiliation(s)
- Steven R Corsi
- U.S. Geological Survey, Middleton, WI 53562, United States.
| | | | - Daniel L Villeneuve
- U.S. Environmental Protection Agency, Office of Research and Development, Duluth, MN 55804, United States
| | - Brett R Blackwell
- U.S. Environmental Protection Agency, Office of Research and Development, Duluth, MN 55804, United States
| | - Kellie A Fay
- General Dynamics Information Technology, Duluth, MN 55804, United States
| | - Gerald T Ankley
- U.S. Environmental Protection Agency, Office of Research and Development, Duluth, MN 55804, United States
| | | |
Collapse
|
91
|
Alofe O, Kisanga E, Inayat-Hussain SH, Fukumura M, Garcia-Milian R, Perera L, Vasiliou V, Whirledge S. Determining the endocrine disruption potential of industrial chemicals using an integrative approach: Public databases, in vitro exposure, and modeling receptor interactions. ENVIRONMENT INTERNATIONAL 2019; 131:104969. [PMID: 31310931 PMCID: PMC6728168 DOI: 10.1016/j.envint.2019.104969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 05/18/2023]
Abstract
Environmental and occupational exposure to industrial chemicals has been linked to toxic and carcinogenic effects in animal models and human studies. However, current toxicology testing does not thoroughly explore the endocrine disrupting effects of industrial chemicals, which may have low dose effects not predicted when determining the limit of toxicity. The objective of this study was to evaluate the endocrine disrupting potential of a broad range of chemicals used in the petrochemical sector. Therefore, 139 chemicals were classified for reproductive toxicity based on the United Nations Globally Harmonized System for hazard classification. These chemicals were evaluated in PubMed for reported endocrine disrupting activity, and their endocrine disrupting potential was estimated by identifying chemicals with active nuclear receptor endpoints publicly available databases. Evaluation of ToxCast data suggested that these chemicals preferentially alter the activity of the estrogen receptor (ER). Four chemicals were prioritized for in vitro testing using the ER-positive, immortalized human uterine Ishikawa cell line and a range of concentrations below the reported limit of toxicity in humans. We found that 2,6-di-tert-butyl-p-cresol (BHT) and diethanolamine (DEA) repressed the basal expression of estrogen-responsive genes PGR, NPPC, and GREB1 in Ishikawa cells, while tetrachloroethylene (PCE) and 2,2'-methyliminodiethanol (MDEA) induced the expression of these genes. Furthermore, low-dose combinations of PCE and MDEA produced additive effects. All four chemicals interfered with estradiol-mediated induction of PGR, NPPC, and GREB1. Molecular docking demonstrated that these chemicals could bind to the ligand binding site of ERα, suggesting the potential for direct stimulatory or inhibitory effects. We found that these chemicals altered rates of proliferation and regulated the expression of cell proliferation associated genes. These findings demonstrate previously unappreciated endocrine disrupting effects and underscore the importance of testing the endocrine disrupting potential of chemicals in the future to better understand their potential to impact public health.
Collapse
Affiliation(s)
- Olubusayo Alofe
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Edwina Kisanga
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Salmaan H Inayat-Hussain
- Department of Product Stewardship and Toxicology, Group Health, Safety, Security and Environment, Petroliam Nasional Berhad, Kuala Lumpur, Malaysia; Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Masao Fukumura
- Department of Product Stewardship and Toxicology, Group Health, Safety, Security and Environment, Petroliam Nasional Berhad, Kuala Lumpur, Malaysia
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, USA
| | - Lalith Perera
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Shannon Whirledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA; Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
92
|
Kosnik MB, Reif DM. Determination of chemical-disease risk values to prioritize connections between environmental factors, genetic variants, and human diseases. Toxicol Appl Pharmacol 2019; 379:114674. [PMID: 31323264 PMCID: PMC6708494 DOI: 10.1016/j.taap.2019.114674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 12/18/2022]
Abstract
Traditional methods for chemical risk assessment are too time-consuming and resource-intensive to characterize either the diversity of chemicals to which humans are exposed or how that diversity may manifest in population susceptibility differences. The advent of novel toxicological data sources and their integration with bioinformatic databases affords opportunities for modern approaches that consider gene-environment (GxE) interactions in population risk assessment. Here, we present an approach that systematically links multiple data sources to relate chemical risk values to diseases and gene-disease variants. These data sources include high-throughput screening (HTS) results from Tox21/ToxCast, chemical-disease relationships from the Comparative Toxicogenomics Database (CTD), hazard data from resources like the Integrated Risk Information System, exposure data from the ExpoCast initiative, and gene-variant-disease information from the DisGeNET database. We use these integrated data to identify variants implicated in chemical-disease enrichments and develop a new value that estimates the risk of these associations toward differential population responses. Finally, we use this value to prioritize chemical-disease associations by exploring the genomic distribution of variants implicated in high-risk diseases. We offer this modular approach, termed DisQGOS (Disease Quotient Genetic Overview Score), for relating overall chemical-disease risk to potential for population variable responses, as a complement to methods aiming to modernize aspects of risk assessment.
Collapse
Affiliation(s)
- Marissa B Kosnik
- Toxicology Program, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7617, United States of America.
| | - David M Reif
- Toxicology Program, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7617, United States of America.
| |
Collapse
|
93
|
Kamdar MR, Fernández JD, Polleres A, Tudorache T, Musen MA. Enabling Web-scale data integration in biomedicine through Linked Open Data. NPJ Digit Med 2019; 2:90. [PMID: 31531395 PMCID: PMC6736878 DOI: 10.1038/s41746-019-0162-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/06/2019] [Indexed: 01/17/2023] Open
Abstract
The biomedical data landscape is fragmented with several isolated, heterogeneous data and knowledge sources, which use varying formats, syntaxes, schemas, and entity notations, existing on the Web. Biomedical researchers face severe logistical and technical challenges to query, integrate, analyze, and visualize data from multiple diverse sources in the context of available biomedical knowledge. Semantic Web technologies and Linked Data principles may aid toward Web-scale semantic processing and data integration in biomedicine. The biomedical research community has been one of the earliest adopters of these technologies and principles to publish data and knowledge on the Web as linked graphs and ontologies, hence creating the Life Sciences Linked Open Data (LSLOD) cloud. In this paper, we provide our perspective on some opportunities proffered by the use of LSLOD to integrate biomedical data and knowledge in three domains: (1) pharmacology, (2) cancer research, and (3) infectious diseases. We will discuss some of the major challenges that hinder the wide-spread use and consumption of LSLOD by the biomedical research community. Finally, we provide a few technical solutions and insights that can address these challenges. Eventually, LSLOD can enable the development of scalable, intelligent infrastructures that support artificial intelligence methods for augmenting human intelligence to achieve better clinical outcomes for patients, to enhance the quality of biomedical research, and to improve our understanding of living systems.
Collapse
Affiliation(s)
- Maulik R. Kamdar
- Center for Biomedical Informatics Research, Stanford University, Stanford, CA USA
| | - Javier D. Fernández
- Vienna University of Economics & Business, Vienna, Austria
- Complexity Science Hub Vienna, Vienna, Austria
| | - Axel Polleres
- Vienna University of Economics & Business, Vienna, Austria
- Complexity Science Hub Vienna, Vienna, Austria
| | - Tania Tudorache
- Center for Biomedical Informatics Research, Stanford University, Stanford, CA USA
| | - Mark A. Musen
- Center for Biomedical Informatics Research, Stanford University, Stanford, CA USA
| |
Collapse
|
94
|
Comparison of Target Features for Predicting Drug-Target Interactions by Deep Neural Network Based on Large-Scale Drug-Induced Transcriptome Data. Pharmaceutics 2019; 11:pharmaceutics11080377. [PMID: 31382356 PMCID: PMC6723794 DOI: 10.3390/pharmaceutics11080377] [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: 06/12/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/31/2022] Open
Abstract
Uncovering drug-target interactions (DTIs) is pivotal to understand drug mode-of-action (MoA), avoid adverse drug reaction (ADR), and seek opportunities for drug repositioning (DR). For decades, in silico predictions for DTIs have largely depended on structural information of both targets and compounds, e.g., docking or ligand-based virtual screening. Recently, the application of deep neural network (DNN) is opening a new path to uncover novel DTIs for thousands of targets. One important question is which features for targets are most relevant to DTI prediction. As an early attempt to answer this question, we objectively compared three canonical target features extracted from: (i) the expression profiles by gene knockdown (GEPs); (ii) the protein–protein interaction network (PPI network); and (iii) the pathway membership (PM) of a target gene. For drug features, the large-scale drug-induced transcriptome dataset, or the Library of Integrated Network-based Cellular Signatures (LINCS) L1000 dataset was used. All these features are closely related to protein function or drug MoA, of which utility is only sparsely investigated. In particular, few studies have compared the three types of target features in DNN-based DTI prediction under the same evaluation scheme. Among the three target features, the PM and the PPI network show similar performances superior to GEPs. DNN models based on both features consistently outperformed other machine learning methods such as naïve Bayes, random forest, or logistic regression.
Collapse
|
95
|
Cheng L, Hu Y, Sun J, Zhou M, Jiang Q. DincRNA: a comprehensive web-based bioinformatics toolkit for exploring disease associations and ncRNA function. Bioinformatics 2019; 34:1953-1956. [PMID: 29365045 DOI: 10.1093/bioinformatics/bty002] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 01/22/2018] [Indexed: 01/09/2023] Open
Abstract
Summary DincRNA aims to provide a comprehensive web-based bioinformatics toolkit to elucidate the entangled relationships among diseases and non-coding RNAs (ncRNAs) from the perspective of disease similarity. The quantitative way to illustrate relationships of pair-wise diseases always depends on their molecular mechanisms, and structures of the directed acyclic graph of Disease Ontology (DO). Corresponding methods for calculating similarity of pair-wise diseases involve Resnik's, Lin's, Wang's, PSB and SemFunSim methods. Recently, disease similarity was validated suitable for calculating functional similarities of ncRNAs and prioritizing ncRNA-disease pairs, and it has been widely applied for predicting the ncRNA function due to the limited biological knowledge from wet lab experiments of these RNAs. For this purpose, a large number of algorithms and priori knowledge need to be integrated. e.g. 'pair-wise best, pairs-average' (PBPA) and 'pair-wise all, pairs-maximum' (PAPM) methods for calculating functional similarities of ncRNAs, and random walk with restart (RWR) method for prioritizing ncRNA-disease pairs. To facilitate the exploration of disease associations and ncRNA function, DincRNA implemented all of the above eight algorithms based on DO and disease-related genes. Currently, it provides the function to query disease similarity scores, miRNA and lncRNA functional similarity scores, and the prioritization scores of lncRNA-disease and miRNA-disease pairs. Availability and implementation http://bio-annotation.cn:18080/DincRNAClient/. Contact biofomeng@hotmail.com or qhjiang@hit.edu.cn. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Liang Cheng
- Department of College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Sheng 150081, China
| | - Yang Hu
- Department of Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Sheng 150001, China
| | - Jie Sun
- Department of College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Sheng 150081, China
| | - Meng Zhou
- Department of College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Sheng 150081, China
| | - Qinghua Jiang
- Department of Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Sheng 150001, China
| |
Collapse
|
96
|
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
|
97
|
PIM1, CYP1B1, and HSPA2 Targeted by Quercetin Play Important Roles in Osteoarthritis Treatment by Achyranthes bidentata. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1205942. [PMID: 31998395 PMCID: PMC6964619 DOI: 10.1155/2019/1205942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/18/2019] [Indexed: 01/02/2023]
Abstract
Aim Achyranthes bidentata is one of the most commonly used Chinese herbal medicines (CHM) that is currently considered for the treatment of osteoarthritis. The purpose of this study was to reveal the mechanism of Achyranthes bidentata in osteoarthritis treatment based on the network pharmacology. Methods The effective components of Achyranthes bidentata were firstly screened out from the TCMSP database with ADME property parameters. Then, osteoarthritis-related proteins targeted by the effective components were predicted based on the DrugBank and CTD databases. Subsequently, enrichment analysis and interaction network between targets of effective components and pathways were also studied. In addition, the differentially expressed genes (DEGs) of GSE55457 were used for validation of the osteoarthritis-related target proteins. Finally, the effective components-target molecular docking models were predicted. Results A total of 10 effective components were identified, of which kaempferol and quercetin had 1 and 29 targets, respectively. There were 26 target proteins of quercetin related to the osteoarthritis. These targets were mainly enriched in mitochondrial ATP synthesis coupled proton transport, cellular response to estradiol stimulus, and nitric oxide biosynthetic process. In addition, there were three common proteins, PIM1, CYP1B1, and HSPA2 based on the DEGs of GSE55457, which were considered as the key targeted proteins of the quercetin. Conclusion The docking of PIM1-quercetin, CYP1B1-quercetin, and HSPA2-quercetin may play important roles during the treatment of osteoarthritis by Achyranthes bidentata.
Collapse
|
98
|
Lin C, Ni S, Liang Y, Zeng X, Liu X. Learning to Predict Drug Target Interaction From Missing Not at Random Labels. IEEE Trans Nanobioscience 2019; 18:353-359. [DOI: 10.1109/tnb.2019.2909293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
99
|
Luo H, Li M, Wang S, Liu Q, Li Y, Wang J. Computational drug repositioning using low-rank matrix approximation and randomized algorithms. Bioinformatics 2019; 34:1904-1912. [PMID: 29365057 DOI: 10.1093/bioinformatics/bty013] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 01/18/2018] [Indexed: 12/26/2022] Open
Abstract
Motivation Computational drug repositioning is an important and efficient approach towards identifying novel treatments for diseases in drug discovery. The emergence of large-scale, heterogeneous biological and biomedical datasets has provided an unprecedented opportunity for developing computational drug repositioning methods. The drug repositioning problem can be modeled as a recommendation system that recommends novel treatments based on known drug-disease associations. The formulation under this recommendation system is matrix completion, assuming that the hidden factors contributing to drug-disease associations are highly correlated and thus the corresponding data matrix is low-rank. Under this assumption, the matrix completion algorithm fills out the unknown entries in the drug-disease matrix by constructing a low-rank matrix approximation, where new drug-disease associations having not been validated can be screened. Results In this work, we propose a drug repositioning recommendation system (DRRS) to predict novel drug indications by integrating related data sources and validated information of drugs and diseases. Firstly, we construct a heterogeneous drug-disease interaction network by integrating drug-drug, disease-disease and drug-disease networks. The heterogeneous network is represented by a large drug-disease adjacency matrix, whose entries include drug pairs, disease pairs, known drug-disease interaction pairs and unknown drug-disease pairs. Then, we adopt a fast Singular Value Thresholding (SVT) algorithm to complete the drug-disease adjacency matrix with predicted scores for unknown drug-disease pairs. The comprehensive experimental results show that DRRS improves the prediction accuracy compared with the other state-of-the-art approaches. In addition, case studies for several selected drugs further demonstrate the practical usefulness of the proposed method. Availability and implementation http://bioinformatics.csu.edu.cn/resources/softs/DrugRepositioning/DRRS/index.html. Contact yaohang@cs.odu.edu or jxwang@mail.csu.edu.cn. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Huimin Luo
- School of Information Science and Engineering, Central South University, ChangSha 410083, China.,School of Computer and Information Engineering, Henan University, KaiFeng 475001, China
| | - Min Li
- School of Information Science and Engineering, Central South University, ChangSha 410083, China
| | - Shaokai Wang
- School of Information Science and Engineering, Central South University, ChangSha 410083, China
| | - Quan Liu
- School of Information Science and Engineering, Central South University, ChangSha 410083, China
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA 23529, USA
| | - Jianxin Wang
- School of Information Science and Engineering, Central South University, ChangSha 410083, China
| |
Collapse
|
100
|
Gokhman D, Kelman G, Amartely A, Gershon G, Tsur S, Carmel L. Gene ORGANizer: linking genes to the organs they affect. Nucleic Acids Res 2019; 45:W138-W145. [PMID: 28444223 PMCID: PMC5570240 DOI: 10.1093/nar/gkx302] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/14/2017] [Indexed: 12/16/2022] Open
Abstract
One of the biggest challenges in studying how genes work is understanding their effect on the physiology and anatomy of the body. Existing tools try to address this using indirect features, such as expression levels and biochemical pathways. Here, we present Gene ORGANizer (geneorganizer.huji.ac.il), a phenotype-based tool that directly links human genes to the body parts they affect. It is built upon an exhaustive curated database that links >7000 genes to ∼150 anatomical parts using >150 000 gene-organ associations. The tool offers user-friendly platforms to analyze the anatomical effects of individual genes, and identify trends within groups of genes. We demonstrate how Gene ORGANizer can be used to make new discoveries, showing that chromosome X is enriched with genes affecting facial features, that positive selection targets genes with more constrained phenotypic effects, and more. We expect Gene ORGANizer to be useful in a variety of evolutionary, medical and molecular studies aimed at understanding the phenotypic effects of genes.
Collapse
Affiliation(s)
- David Gokhman
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Guy Kelman
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Adir Amartely
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Guy Gershon
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Shira Tsur
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Liran Carmel
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel.,Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|