451
|
Quan Y, Luo ZH, Yang QY, Li J, Zhu Q, Liu YM, Lv BM, Cui ZJ, Qin X, Xu YH, Zhu LD, Zhang HY. Systems Chemical Genetics-Based Drug Discovery: Prioritizing Agents Targeting Multiple/Reliable Disease-Associated Genes as Drug Candidates. Front Genet 2019; 10:474. [PMID: 31191604 PMCID: PMC6549477 DOI: 10.3389/fgene.2019.00474] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/01/2019] [Indexed: 01/10/2023] Open
Abstract
Genetic disease genes are considered a promising source of drug targets. Most diseases are caused by more than one pathogenic factor; thus, it is reasonable to consider that chemical agents targeting multiple disease genes are more likely to have desired activities. This is supported by a comprehensive analysis on the relationships between agent activity/druggability and target genetic characteristics. The therapeutic potential of agents increases steadily with increasing number of targeted disease genes, and can be further enhanced by strengthened genetic links between targets and diseases. By using the multi-label classification models for genetics-based drug activity prediction, we provide universal tools for prioritizing drug candidates. All of the documented data and the machine-learning prediction service are available at SCG-Drug (http://zhanglab.hzau.edu.cn/scgdrug).
Collapse
Affiliation(s)
- Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Zhi-Hui Luo
- College of Life Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qing-Yong Yang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Jiang Li
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Qiang Zhu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Ye-Mao Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Bo-Min Lv
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Ze-Jia Cui
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xuan Qin
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Yan-Hua Xu
- Sci-meds Biopharmaceutical Co., Ltd., Wuhan, China
| | - Li-Da Zhu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
452
|
Jiang ZC, Chen XJ, Zhou Q, Gong XH, Chen X, Wu WJ. Downregulated LRRK2 gene expression inhibits proliferation and migration while promoting the apoptosis of thyroid cancer cells by inhibiting activation of the JNK signaling pathway. Int J Oncol 2019; 55:21-34. [PMID: 31180559 PMCID: PMC6561619 DOI: 10.3892/ijo.2019.4816] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
Emerging studies have indicated that leucine-rich repeat kinase 2 (LRRK2) is associated with thyroid cancer (TC). The present study investigated the effect of LRRK2 on the cell cycle and apoptosis in TC, and examined the underlying mechanisms in vitro. To screen TC-associated differentially expressed genes, gene expression microarray analysis was conducted. Retrieval of pathways associated with TC from the Kyoto Encyclopedia of Genes and Genomes database indicated that the c-Jun N-terminal kinase (JNK) signaling pathway serves an essential role in TC. SW579, IHH-4, TFC-133, TPC-1 and Nthy-ori3-1 cell lines were used to screen cell lines with the highest and lowest LRRK2 expression for subsequent experiments. The two selected cell lines were transfected with pcDNA-LRRK2, or small interfering RNA against LRRK2 or SP600125 (a JNK inhibitor). Subsequently, flow cytometry, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling, a 5-ethynyl-2′-deoxyuridine assay and a scratch test was conducted to detect the cell cycle distribution, apoptosis, proliferation and migration, respectively, in each group. The LRRK2 gene was determined to be elevated in TC based on the microarray data of the GSE3678 dataset. The SW579 cell line was identified to exhibit the highest LRRK2 expression, while IHH-4 cells exhibited the lowest LRRK2 expression. LRRK2 silencing, through inhibiting the activation of the JNK signaling pathway, increased the expression levels of genes and proteins associated with cell cycle arrest and apoptosis in TC cells, promoted cell cycle arrest and apoptosis, and inhibited cell migration and proliferation in TC cells, indicating that LRRK2 repression could exert beneficial effects through the JNK signaling pathway on TC cells. These observations demonstrate that LRRK2 silencing promotes TC cell growth inhibition, and facilitates apoptosis and cell cycle arrest. The JNK signaling pathway may serve a crucial role in mediating the anti-carcinogenic activities of downregulated LRRK2 in TC.
Collapse
Affiliation(s)
- Zheng-Cai Jiang
- Department of General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiao-Jun Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, P.R. China
| | - Qi Zhou
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, P.R. China
| | - Xiao-Hua Gong
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, P.R. China
| | - Xiong Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, P.R. China
| | - Wen-Jun Wu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, P.R. China
| |
Collapse
|
453
|
Arbabi A, Adams DR, Fidler S, Brudno M. Identifying Clinical Terms in Medical Text Using Ontology-Guided Machine Learning. JMIR Med Inform 2019; 7:e12596. [PMID: 31094361 PMCID: PMC6533869 DOI: 10.2196/12596] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/20/2019] [Accepted: 03/29/2019] [Indexed: 01/22/2023] Open
Abstract
Background Automatic recognition of medical concepts in unstructured text is an important component of many clinical and research applications, and its accuracy has a large impact on electronic health record analysis. The mining of medical concepts is complicated by the broad use of synonyms and nonstandard terms in medical documents. Objective We present a machine learning model for concept recognition in large unstructured text, which optimizes the use of ontological structures and can identify previously unobserved synonyms for concepts in the ontology. Methods We present a neural dictionary model that can be used to predict if a phrase is synonymous to a concept in a reference ontology. Our model, called the Neural Concept Recognizer (NCR), uses a convolutional neural network to encode input phrases and then rank medical concepts based on the similarity in that space. It uses the hierarchical structure provided by the biomedical ontology as an implicit prior embedding to better learn embedding of various terms. We trained our model on two biomedical ontologies—the Human Phenotype Ontology (HPO) and Systematized Nomenclature of Medicine - Clinical Terms (SNOMED-CT). Results We tested our model trained on HPO by using two different data sets: 288 annotated PubMed abstracts and 39 clinical reports. We achieved 1.7%-3% higher F1-scores than those for our strongest manually engineered rule-based baselines (P=.003). We also tested our model trained on the SNOMED-CT by using 2000 Intensive Care Unit discharge summaries from MIMIC (Multiparameter Intelligent Monitoring in Intensive Care) and achieved 0.9%-1.3% higher F1-scores than those of our baseline. The results of our experiments show high accuracy of our model as well as the value of using the taxonomy structure of the ontology in concept recognition. Conclusion Most popular medical concept recognizers rely on rule-based models, which cannot generalize well to unseen synonyms. In addition, most machine learning methods typically require large corpora of annotated text that cover all classes of concepts, which can be extremely difficult to obtain for biomedical ontologies. Without relying on large-scale labeled training data or requiring any custom training, our model can be efficiently generalized to new synonyms and performs as well or better than state-of-the-art methods custom built for specific ontologies.
Collapse
Affiliation(s)
- Aryan Arbabi
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.,Centre for Computational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - David R Adams
- Section on Human Biochemical Genetics, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sanja Fidler
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Michael Brudno
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.,Centre for Computational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
454
|
Rosso M, Lapyckyj L, Besso MJ, Monge M, Reventós J, Canals F, Quevedo Cuenca JO, Matos ML, Vazquez-Levin MH. Characterization of the molecular changes associated with the overexpression of a novel epithelial cadherin splice variant mRNA in a breast cancer model using proteomics and bioinformatics approaches: identification of changes in cell metabolism and an increased expression of lactate dehydrogenase B. Cancer Metab 2019; 7:5. [PMID: 31086659 PMCID: PMC6507066 DOI: 10.1186/s40170-019-0196-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background Breast cancer (BC) is the most common female cancer and the leading cause of cancer death in women worldwide. Alterations in epithelial cadherin (E-cadherin) expression and functions are associated to BC, but the underlying molecular mechanisms have not been fully elucidated. We have previously reported a novel human E-cadherin splice variant (E-cadherin variant) mRNA. Stable transfectants in MCF-7 human BC cells (MCF7Ecadvar) depicted fibroblast-like cell morphology, E-cadherin wild-type downregulation, and other molecular changes characteristic of the epithelial-to-mesenchymal transition process, reduced cell-cell adhesion, and increased cell migration and invasion. In this study, a two-dimensional differential gel electrophoresis (2D-DIGE) combined with mass spectrometry (MS) protein identification and bioinformatics analyses were done to characterize biological processes and canonical pathways affected by E-cadherin variant expression. Results By 2D-DIGE and MS analysis, 50 proteins were found differentially expressed (≥ Δ1.5) in MCF7Ecadvar compared to control cells. Validation of transcript expression was done in the ten most overexpressed and underexpressed proteins. Bioinformatics analyses revealed that 39 of the 50 proteins identified had been previously associated to BC. Moreover, metabolic processes were the most affected, and glycolysis the canonical pathway most altered. The lactate dehydrogenase B (LDHB) was the highest overexpressed protein, and transcript levels were higher in MCF7Ecadvar than in control cells. In agreement with these findings, MCF7Ecadvar conditioned media had lower glucose and higher lactate levels than control cells. MCF7Ecadvar cell treatment with 5 mM of the glycolytic inhibitor 2-deoxy-glucose led to decreased cell viability, and modulation of LDHB expression in MCF7Ecadvar cells with a specific small interfering RNA resulted in decreased cell proliferation. Finally, a positive association between expression levels of the E-cadherin variant and LDHB transcripts was demonstrated in 21 human breast tumor tissues, and breast tumor samples with higher Ki67 expression showed higher LDHB mRNA levels. Conclusions Results from this investigation contributed to further characterize molecular changes associated to the novel E-cadherin splice variant expression in BC cells. They also revealed an association between expression of the novel variant and changes related to BC progression and aggressiveness, in particular those associated to cell metabolism.
Collapse
Affiliation(s)
- Marina Rosso
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Lara Lapyckyj
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - María José Besso
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Marta Monge
- 2Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jaume Reventós
- 3Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Francesc Canals
- 2Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jorge Oswaldo Quevedo Cuenca
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - María Laura Matos
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Mónica Hebe Vazquez-Levin
- Laboratorio de Estudios de Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| |
Collapse
|
455
|
Identify the Key Active Ingredients and Pharmacological Mechanisms of Compound XiongShao Capsule in Treating Diabetic Peripheral Neuropathy by Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:5801591. [PMID: 31210774 PMCID: PMC6532326 DOI: 10.1155/2019/5801591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/20/2019] [Accepted: 04/15/2019] [Indexed: 02/07/2023]
Abstract
Compound XiongShao Capsule (CXSC), a traditional herb mixture, has shown significant clinical efficacy against diabetic peripheral neuropathy (DPN). However, its multicomponent and multitarget features cause difficulty in deciphering its molecular mechanisms. Our study aimed to identify the key active ingredients and potential pharmacological mechanisms of CXSC in treating DPN by network pharmacology and provide scientific evidence of its clinical efficacy. CXSC active ingredients were identified from both the Traditional Chinese Medicine Systems Pharmacology database, with parameters of oral bioavailability ≥ 30% and drug-likeness ≥ 0.18, and the Herbal Ingredients' Targets (HIT) database. The targets of those active ingredients were identified using ChemMapper based on 3D-structure similarity and using HIT database. DPN-related genes were acquired from microarray dataset GSE95849 and five widely used databases (TTD, Drugbank, KEGG, DisGeNET, and OMIM). Next, we obtained candidate targets with therapeutic effects against DPN by mapping active ingredient targets and DPN-related genes and identifying the proteins interacting with those candidate targets using STITCH 5.0. We constructed an “active ingredients-candidate targets-proteins” network using Cytoscape 3.61 and identified key active ingredients and key targets in the network. We identified 172 active ingredients in CXSC, 898 targets of the active ingredients, 110 DPN-related genes, and 38 candidate targets with therapeutic effects against DPN. Three key active ingredients, namely, quercetin, kaempferol, and baicalein, and 25 key targets were identified. Next, we input all key targets into ClueGO plugin for KEGG enrichment and molecular function analyses. The AGE-RAGE signaling pathway in diabetic complications and MAP kinase activity were determined as the main KEGG pathway and molecular function involved, respectively. We determined quercetin, kaempferol, and baicalein as the key active ingredients of CXSC and the AGE-RAGE signaling pathway and MAP kinase activity as the main pharmacological mechanisms of CXSC against DPN, proving the clinical efficacy of CXSC against DPN.
Collapse
|
456
|
Systems biology approach identifies key regulators and the interplay between miRNAs and transcription factors for pathological cardiac hypertrophy. Gene 2019; 698:157-169. [DOI: 10.1016/j.gene.2019.02.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/31/2019] [Accepted: 02/20/2019] [Indexed: 12/16/2022]
|
457
|
Wang X, Diao L, Sun D, Wang D, Zhu J, He Y, Liu Y, Xu H, Zhang Y, Liu J, Wang Y, He F, Li Y, Li D. OsteoporosAtlas: a human osteoporosis-related gene database. PeerJ 2019; 7:e6778. [PMID: 31086734 PMCID: PMC6487800 DOI: 10.7717/peerj.6778] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/13/2019] [Indexed: 01/12/2023] Open
Abstract
Background Osteoporosis is a common, complex disease of bone with a strong heritable component, characterized by low bone mineral density, microarchitectural deterioration of bone tissue and an increased risk of fracture. Due to limited drug selection for osteoporosis and increasing morbidity, mortality of osteoporotic fractures, osteoporosis has become a major health burden in aging societies. Current researches for identifying specific loci or genes involved in osteoporosis contribute to a greater understanding of the pathogenesis of osteoporosis and the development of better diagnosis, prevention and treatment strategies. However, little is known about how most causal genes work and interact to influence osteoporosis. Therefore, it is greatly significant to collect and analyze the studies involved in osteoporosis-related genes. Unfortunately, the information about all these osteoporosis-related genes is scattered in a large amount of extensive literature. Currently, there is no specialized database for easily accessing relevant information about osteoporosis-related genes and miRNAs. Methods We extracted data from literature abstracts in PubMed by text-mining and manual curation. Moreover, a local MySQL database containing all the data was developed with PHP on a Windows server. Results OsteoporosAtlas (http://biokb.ncpsb.org/osteoporosis/), the first specialized database for easily accessing relevant information such as osteoporosis-related genes and miRNAs, was constructed and served for researchers. OsteoporosAtlas enables users to retrieve, browse and download osteoporosis-related genes and miRNAs. Gene ontology and pathway analyses were integrated into OsteoporosAtlas. It currently includes 617 human encoding genes, 131 human non-coding miRNAs, and 128 functional roles. We think that OsteoporosAtlas will be an important bioinformatics resource to facilitate a better understanding of the pathogenesis of osteoporosis and developing better diagnosis, prevention and treatment strategies.
Collapse
Affiliation(s)
- Xun Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Lihong Diao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Dezhi Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Dan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Jiarun Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China.,College of life Sciences, Hebei University, Baoding, China
| | - Yangzhige He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China.,Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuan Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Hao Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yi Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China.,College of life Sciences, Hebei University, Baoding, China
| | - Jinying Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yang Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Dong Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
458
|
Zhai Y, Xu J, Feng L, Liu Q, Yao W, Li H, Cao Y, Cheng F, Bao B, Zhang L. Broad range metabolomics coupled with network analysis for explaining possible mechanisms of Er-Zhi-Wan in treating liver-kidney Yin deficiency syndrome of Traditional Chinese medicine. JOURNAL OF ETHNOPHARMACOLOGY 2019; 234:57-66. [PMID: 30690072 DOI: 10.1016/j.jep.2019.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/21/2018] [Accepted: 01/19/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Er-Zhi-Wan (EZW), a famous traditional Chinese formulation, is used to prevent, or to treat, various liver and kidney diseases for its actions of replenishing liver and kidney. However, the mechanisms of treating Liver-kidney Yin deficiency syndrome (LKYDS) of EZW have not been comprehensively investigated. AIM OF THE STUDY In this study, a broad range metabolomics strategy coupled with network analysis was established to investigate possible mechanisms of EZW in treating LKYDS. MATERIALS AND METHOD The rat models of LKYDS were established using the mixture of thyroxine and reserpine, and the changes of biochemical indices in serum and histopathology were detected to explore the effects of EZW. Next, a broad range metabolomics strategy based on RPLC-Q-TOF/MS and HILIC-Q-TOF/MS has been developed to find the possible significant metabolites in the serum and urine of LKYDS rats. Then, network analysis was applied to visualize the relationships between identified serum and urine metabolites and in detail to find hub metabolites, which might be responsible for the effect of EZW on rats of LKYDS. Furthermore, the shortest path of "disease gene-pathway protein-metabolite" was built to investigate the possible intervention path of EZW from the systematic perspective. RESULTS Five hub metabolites, namely, arachidonic acid, L-arginine, testosterone, taurine and oxoglutaric acid, were screened out and could be adjusted to recover by EZW. After that, the shortest path starting from disease genes and ending in metabolites were identified and disclosed, and the genes of aging such as CAV1 and ACO1 were selected to explain the pathological mechanism of LKYDS. CONCLUSION Broad range metabolomics coupled with network analysis could provide another perspective on systematically investigating the molecular mechanism of EZW in treating LKYDS at metabolomics level. In addition, EZW might prevent the pathological process of LKYDS through regulating the disturbed metabolic pathway and the aging genes such as CAV1 and ACO1, which may be potential targets for EZW in the treatment of LKYDS.
Collapse
Affiliation(s)
- Yuanyuan Zhai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jia Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Li Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Qinan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Weifeng Yao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hui Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yudan Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Fangfang Cheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Beihua Bao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Li Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
459
|
Kumar R, Nagpal G, Kumar V, Usmani SS, Agrawal P, Raghava GPS. HumCFS: a database of fragile sites in human chromosomes. BMC Genomics 2019; 19:985. [PMID: 30999860 PMCID: PMC7402404 DOI: 10.1186/s12864-018-5330-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/29/2018] [Indexed: 11/25/2022] Open
Abstract
Background Fragile sites are the chromosomal regions that are susceptible to breakage, and their frequency varies among the human population. Based on the frequency of fragile site induction, they are categorized as common and rare fragile sites. Common fragile sites are sensitive to replication stress and often rearranged in cancer. Rare fragile sites are the archetypal trinucleotide repeats. Fragile sites are known to be involved in chromosomal rearrangements in tumors. Human miRNA genes are also present at fragile sites. A better understanding of genes and miRNAs lying in the fragile site regions and their association with disease progression is required. Result HumCFS is a manually curated database of human chromosomal fragile sites. HumCFS provides useful information on fragile sites such as coordinates on the chromosome, cytoband, their chemical inducers and frequency of fragile site (rare or common), genes and miRNAs lying in fragile sites. Protein coding genes in the fragile sites were identified by mapping the coordinates of fragile sites with human genome Ensembl (GRCh38/hg38). Genes present in fragile sites were further mapped to DisGenNET database, to understand their possible link with human diseases. Human miRNAs from miRBase was also mapped on fragile site coordinates. In brief, HumCFS provides useful information about 125 human chromosomal fragile sites and their association with 4921 human protein-coding genes and 917 human miRNA’s. Conclusion User-friendly web-interface of HumCFS and hyper-linking with other resources will help researchers to search for genes, miRNAs efficiently and to intersect the relationship among them. For easy data retrieval and analysis, we have integrated standard web-based tools, such as JBrowse, BLAST etc. Also, the user can download the data in various file formats such as text files, gff3 files and Bed-format files which can be used on UCSC browser. Database URL:http://webs.iiitd.edu.in/raghava/humcfs/ Electronic supplementary material The online version of this article (10.1186/s12864-018-5330-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rajesh Kumar
- Center for Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110020, India.,Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Gandharva Nagpal
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Vinod Kumar
- Center for Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110020, India.,Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Salman Sadullah Usmani
- Center for Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110020, India.,Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Piyush Agrawal
- Center for Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110020, India.,Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Gajendra P S Raghava
- Center for Computational Biology, Indraprastha Institute of Information Technology, New Delhi, 110020, India.
| |
Collapse
|
460
|
Monnin P, Legrand J, Husson G, Ringot P, Tchechmedjiev A, Jonquet C, Napoli A, Coulet A. PGxO and PGxLOD: a reconciliation of pharmacogenomic knowledge of various provenances, enabling further comparison. BMC Bioinformatics 2019; 20:139. [PMID: 30999867 PMCID: PMC6471679 DOI: 10.1186/s12859-019-2693-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background Pharmacogenomics (PGx) studies how genomic variations impact variations in drug response phenotypes. Knowledge in pharmacogenomics is typically composed of units that have the form of ternary relationships gene variant – drug – adverse event. Such a relationship states that an adverse event may occur for patients having the specified gene variant and being exposed to the specified drug. State-of-the-art knowledge in PGx is mainly available in reference databases such as PharmGKB and reported in scientific biomedical literature. But, PGx knowledge can also be discovered from clinical data, such as Electronic Health Records (EHRs), and in this case, may either correspond to new knowledge or confirm state-of-the-art knowledge that lacks “clinical counterpart” or validation. For this reason, there is a need for automatic comparison of knowledge units from distinct sources. Results In this article, we propose an approach, based on Semantic Web technologies, to represent and compare PGx knowledge units. To this end, we developed PGxO, a simple ontology that represents PGx knowledge units and their components. Combined with PROV-O, an ontology developed by the W3C to represent provenance information, PGxO enables encoding and associating provenance information to PGx relationships. Additionally, we introduce a set of rules to reconcile PGx knowledge, i.e. to identify when two relationships, potentially expressed using different vocabularies and levels of granularity, refer to the same, or to different knowledge units. We evaluated our ontology and rules by populating PGxO with knowledge units extracted from PharmGKB (2701), the literature (65,720) and from discoveries reported in EHR analysis studies (only 10, manually extracted); and by testing their similarity. We called PGxLOD (PGx Linked Open Data) the resulting knowledge base that represents and reconciles knowledge units of those various origins. Conclusions The proposed ontology and reconciliation rules constitute a first step toward a more complete framework for knowledge comparison in PGx. In this direction, the experimental instantiation of PGxO, named PGxLOD, illustrates the ability and difficulties of reconciling various existing knowledge sources. Electronic supplementary material The online version of this article (10.1186/s12859-019-2693-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pierre Monnin
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France.
| | - Joël Legrand
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France
| | - Graziella Husson
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France
| | - Patrice Ringot
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France
| | | | - Clément Jonquet
- LIRMM, Université de Montpellier, CNRS, Montpellier, 34095, France.,Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, 94305, California, USA
| | - Amedeo Napoli
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France
| | - Adrien Coulet
- Université de Lorraine, CNRS, Inria, LORIA, Nancy, 54000, France.,Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, 94305, California, USA
| |
Collapse
|
461
|
Han SK, Kong J, Kim S, Lee JH, Han DH. Exomic and transcriptomic alterations of hereditary gingival fibromatosis. Oral Dis 2019; 25:1374-1383. [PMID: 30907493 DOI: 10.1111/odi.13093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/18/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Hereditary gingival fibromatosis (HGF) is a rare oral disease characterized by either localized or generalized gradual, benign, non-hemorrhagic enlargement of gingivae. Although several genetic causes of HGF are known, the genetic etiology of HGF as a non-syndromic and idiopathic entity remains uncertain. SUBJECTS AND METHODS We performed exome and RNA-seq of idiopathic HGF patients and controls, and then devised a computational framework that specifies exomic/transcriptomic alterations interconnected by a regulatory network to unravel genetic etiology of HGF. Moreover, given the lack of animal model or large-scale cohort data of HGF, we developed a strategy to cross-check their clinical relevance through in silico gene-phenotype mapping with biomedical literature mining and semantic analysis of disease phenotype similarities. RESULTS Exomic variants and differentially expressed genes of HGF were connected by members of TGF-β/SMAD signaling pathway and craniofacial development processes, accounting for the molecular mechanism of fibroblast overgrowth mimicking HGF. Our cross-check supports that genes derived from the regulatory network analysis have pathogenic roles in fibromatosis-related diseases. CONCLUSIONS The computational approach of connecting exomic and transcriptomic alterations through regulatory networks is applicable in the clinical interpretation of genetic variants in HGF patients.
Collapse
Affiliation(s)
- Seong Kyu Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Jungho Kong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea.,Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Korea
| | - Jae-Hoon Lee
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Korea
| | - Dong-Hoo Han
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Korea
| |
Collapse
|
462
|
FCMDAP: using miRNA family and cluster information to improve the prediction accuracy of disease related miRNAs. BMC SYSTEMS BIOLOGY 2019; 13:26. [PMID: 30953512 PMCID: PMC6449885 DOI: 10.1186/s12918-019-0696-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Biological experiments have confirmed the association between miRNAs and various diseases. However, such experiments are costly and time consuming. Computational methods help select potential disease-related miRNAs to improve the efficiency of biological experiments. Methods In this work, we develop a novel method using multiple types of data to calculate miRNA and disease similarity based on mutual information, and add miRNA family and cluster information to predict human disease-related miRNAs (FCMDAP). This method not only depends on known miRNA-diseases associations but also accurately measures miRNA and disease similarity and resolves the problem of overestimation. FCMDAP uses the k most similar neighbor recommendation algorithm to predict the association score between miRNA and disease. Information about miRNA cluster is also used to improve prediction accuracy. Result FCMDAP achieves an average AUC of 0.9165 based on leave-one-out cross validation. Results confirm the 100, 98 and 96% of the top 50 predicted miRNAs reported in case studies on colorectal, lung, and pancreatic neoplasms. FCMDAP also exhibits satisfactory performance in predicting diseases without any related miRNAs and miRNAs without any related diseases. Conclusions In this study, we present a computational method FCMDAP to improve the prediction accuracy of disease related miRNAs. FCMDAP could be an effective tool for further biological experiments. Electronic supplementary material The online version of this article (10.1186/s12918-019-0696-9) contains supplementary material, which is available to authorized users.
Collapse
|
463
|
Guo H, Wang J, Yao J, Sun S, Sheng N, Zhang X, Guo X, Guo Y, Sun Y, Dai J. Comparative Hepatotoxicity of Novel PFOA Alternatives (Perfluoropolyether Carboxylic Acids) on Male Mice. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:3929-3937. [PMID: 30865431 DOI: 10.1021/acs.est.9b00148] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
As novel alternatives to perfluorooctanoic acid (PFOA), perfluoropolyether carboxylic acids (multiether PFECAs, CF3(OCF2) nCOO-, n = 2-4) have been detected in various environmental matrices; however, public information regarding their toxicities remains unavailable. To compare the hepatotoxicity of multiether PFECAs (e.g., PFO2HxA, PFO3OA, and PFO4DA) with PFOA, male mice were exposed to 0.4, 2, or 10 mg/kg/d of each chemical for 28 d, respectively. Results demonstrated that PFO2HxA and PFO3OA exposure did not induce marked increases in relative liver weight; whereas 2 and 10 mg/kg/d of PFO4DA significantly increased relative liver weight. Furthermore, PFO2HxA and PFO3OA demonstrated almost no accumulation in the liver or serum; whereas PFO4DA was accumulated but with weaker potential than PFOA. Exposure to 10 mg/kg/d of PFO4DA led to 198 differentially expressed liver genes (56 down-regulated, 142 up-regulated), with bioinformatics analysis highlighting the urea cycle disorder. Like PFOA, 10 mg/kg/d of PFO4DA decreased the urea cycle-related enzyme protein levels (e.g., carbamoyl phosphate synthetase 1) and serum ammonia content in a dose-dependent manner. Both PFOA and PFO4DA treatment (highest concentration) caused a decrease in glutamate content and increase in both glutamine synthetase activity and aquaporin protein levels in the brain. Thus, we concluded that PFO4DA caused hepatotoxicity, as indicated by hepatomegaly and karyolysis, though to a lesser degree than PFOA, and induced urea cycle disorder, which may contribute to the observed toxic effects.
Collapse
Affiliation(s)
- Hua Guo
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| | - Jinghua Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| | - Jingzhi Yao
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| | - Sujie Sun
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| | - Nan Sheng
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| | - Xiaowen Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine , Nanjing Medical University , Nanjing 210029 , China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry , Chinese Academy of Sciences , Shanghai 200032 , China
| | - Yan Sun
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry , Chinese Academy of Sciences , Shanghai 200032 , China
| | - Jiayin Dai
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing 100101 , China
| |
Collapse
|
464
|
Gomez-Rubio P, Piñero J, Molina-Montes E, Gutiérrez-Sacristán A, Marquez M, Rava M, Michalski CW, Farré A, Molero X, Löhr M, Perea J, Greenhalf W, O'Rorke M, Tardón A, Gress T, Barberá VM, Crnogorac-Jurcevic T, Muñoz-Bellvís L, Domínguez-Muñoz E, Balsells J, Costello E, Yu J, Iglesias M, Ilzarbe L, Kleeff J, Kong B, Mora J, Murray L, O'Driscoll D, Poves I, Lawlor RT, Ye W, Hidalgo M, Scarpa A, Sharp L, Carrato A, Real FX, Furlong LI, Malats N. Pancreatic cancer and autoimmune diseases: An association sustained by computational and epidemiological case-control approaches. Int J Cancer 2019; 144:1540-1549. [PMID: 30229903 DOI: 10.1002/ijc.31866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/18/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
Abstract
Deciphering the underlying genetic basis behind pancreatic cancer (PC) and its associated multimorbidities will enhance our knowledge toward PC control. The study investigated the common genetic background of PC and different morbidities through a computational approach and further evaluated the less explored association between PC and autoimmune diseases (AIDs) through an epidemiological analysis. Gene-disease associations (GDAs) of 26 morbidities of interest and PC were obtained using the DisGeNET public discovery platform. The association between AIDs and PC pointed by the computational analysis was confirmed through multivariable logistic regression models in the PanGen European case-control study population of 1,705 PC cases and 1,084 controls. Fifteen morbidities shared at least one gene with PC in the DisGeNET database. Based on common genes, several AIDs were genetically associated with PC pointing to a potential link between them. An epidemiologic analysis confirmed that having any of the nine AIDs studied was significantly associated with a reduced risk of PC (Odds Ratio (OR) = 0.74, 95% confidence interval (CI) 0.58-0.93) which decreased in subjects having ≥2 AIDs (OR = 0.39, 95%CI 0.21-0.73). In independent analyses, polymyalgia rheumatica, and rheumatoid arthritis were significantly associated with low PC risk (OR = 0.40, 95%CI 0.19-0.89, and OR = 0.73, 95%CI 0.53-1.00, respectively). Several inflammatory-related morbidities shared a common genetic component with PC based on public databases. These molecular links could shed light into the molecular mechanisms underlying PC development and simultaneously generate novel hypotheses. In our study, we report sound findings pointing to an association between AIDs and a reduced risk of PC.
Collapse
Affiliation(s)
- Paulina Gomez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Janet Piñero
- Research Program on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM), Universidad Pompeu Fabra (UPF), Barcelona, Spain
| | - Esther Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Alba Gutiérrez-Sacristán
- Research Program on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM), Universidad Pompeu Fabra (UPF), Barcelona, Spain
| | - Mirari Marquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marta Rava
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Christoph W Michalski
- Department of Surgery, Technical University of Munich, Munich, Germany
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - Antoni Farré
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Xavier Molero
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital Universitaru Vall d'Hebron, Exocrine Pancreas Research Unit and Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Universitat Auntònoma de Barcelona, Campus de la UAB, Barcelona, Spain
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - José Perea
- Department of Surgery, University Hospital 12 de Octubre, Madrid, Spain
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Michael O'Rorke
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Adonina Tardón
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Department of Medicine, Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Thomas Gress
- Department of Gastroenterology, University Hospital of Giessen and Marburg, Marburg, Germany
| | - Victor M Barberá
- Laboratorio de Genética Molecular, Hospital General Universitario de Elche, Elche, Spain
| | - Tatjana Crnogorac-Jurcevic
- Centre for Molecular Oncology, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Luís Muñoz-Bellvís
- General and Digestive Surgery Department, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Enrique Domínguez-Muñoz
- Department of Gastroenterology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Joaquim Balsells
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital Universitaru Vall d'Hebron, Exocrine Pancreas Research Unit and Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Universitat Auntònoma de Barcelona, Campus de la UAB, Barcelona, Spain
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Jingru Yu
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Mar Iglesias
- Department of Gastroenterology, Hospital del Mar/Parc de Salut Mar, Barcelona, Spain
| | - Lucas Ilzarbe
- Department of Gastroenterology, Hospital del Mar/Parc de Salut Mar, Barcelona, Spain
| | - Jörg Kleeff
- Department of Surgery, Technical University of Munich, Munich, Germany
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, Halle, (Saale), Germany
| | - Bo Kong
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Josefina Mora
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Liam Murray
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Damian O'Driscoll
- Cancer Data Registrars, National Cancer Registry Ireland, Cork, Ireland
| | - Ignasi Poves
- Department of Gastroenterology, Hospital del Mar/Parc de Salut Mar, Barcelona, Spain
| | - Rita T Lawlor
- ARC-Net Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, University Hospital Trust of Verona, Verona, Italy
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet and University Hospital, Sweden
| | - Manuel Hidalgo
- Hospital Madrid-Norte-Sanchinarro and Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Rosenberg Clinical Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Aldo Scarpa
- ARC-Net Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, University Hospital Trust of Verona, Verona, Italy
| | - Linda Sharp
- Cancer Data Registrars, National Cancer Registry Ireland, Cork, Ireland
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alfredo Carrato
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Department of Oncology, Hospital Ramón y Cajal, Madrid, Spain
| | - Francisco X Real
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laura I Furlong
- Research Program on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM), Universidad Pompeu Fabra (UPF), Barcelona, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Enfermedades Hepáticas y Digestivas (CIBERHD), and Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
465
|
Iglesias V, Paladin L, Juan-Blanco T, Pallarès I, Aloy P, Tosatto SCE, Ventura S. In silico Characterization of Human Prion-Like Proteins: Beyond Neurological Diseases. Front Physiol 2019; 10:314. [PMID: 30971948 PMCID: PMC6445884 DOI: 10.3389/fphys.2019.00314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/07/2019] [Indexed: 12/18/2022] Open
Abstract
Prion-like behavior has been in the spotlight since it was first associated with the onset of mammalian neurodegenerative diseases. However, a growing body of evidence suggests that this mechanism could be behind the regulation of processes such as transcription and translation in multiple species. Here, we perform a stringent computational survey to identify prion-like proteins in the human proteome. We detected 242 candidate polypeptides and computationally assessed their function, protein–protein interaction networks, tissular expression, and their link to disease. Human prion-like proteins constitute a subset of modular polypeptides broadly expressed across different cell types and tissues, significantly associated with disease, embedded in highly connected interaction networks, and involved in the flow of genetic information in the cell. Our analysis suggests that these proteins might play a relevant role not only in neurological disorders, but also in different types of cancer and viral infections.
Collapse
Affiliation(s)
- Valentin Iglesias
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lisanna Paladin
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Teresa Juan-Blanco
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Irantzu Pallarès
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Silvio C E Tosatto
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,CNR Institute of Neuroscience, Padua, Italy
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
466
|
Saqi M, Lysenko A, Guo YK, Tsunoda T, Auffray C. Navigating the disease landscape: knowledge representations for contextualizing molecular signatures. Brief Bioinform 2019; 20:609-623. [PMID: 29684165 PMCID: PMC6556902 DOI: 10.1093/bib/bby025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/05/2018] [Indexed: 12/14/2022] Open
Abstract
Large amounts of data emerging from experiments in molecular medicine are leading to the identification of molecular signatures associated with disease subtypes. The contextualization of these patterns is important for obtaining mechanistic insight into the aberrant processes associated with a disease, and this typically involves the integration of multiple heterogeneous types of data. In this review, we discuss knowledge representations that can be useful to explore the biological context of molecular signatures, in particular three main approaches, namely, pathway mapping approaches, molecular network centric approaches and approaches that represent biological statements as knowledge graphs. We discuss the utility of each of these paradigms, illustrate how they can be leveraged with selected practical examples and identify ongoing challenges for this field of research.
Collapse
Affiliation(s)
- Mansoor Saqi
- Mansoor Saqi Data Science Institute, Imperial College London, UK
| | - Artem Lysenko
- Artem Lysenko Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yi-Ke Guo
- Yi-Ke Guo Data Science Institute, Imperial College London, UK
| | - Tatsuhiko Tsunoda
- Tatsuhiko Tsunoda Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan CREST, JST, Tokyo, Japan Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Charles Auffray
- Charles Auffray European Institute for Systems Biology and Medicine, Lyon, France
| |
Collapse
|
467
|
MorCVD: A Unified Database for Host-Pathogen Protein-Protein Interactions of Cardiovascular Diseases Related to Microbes. Sci Rep 2019; 9:4039. [PMID: 30858555 PMCID: PMC6411875 DOI: 10.1038/s41598-019-40704-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 02/20/2019] [Indexed: 01/07/2023] Open
Abstract
Microbe induced cardiovascular diseases (CVDs) are less studied at present. Host-pathogen interactions (HPIs) between human proteins and microbial proteins associated with CVD can be found dispersed in existing molecular interaction databases. MorCVD database is a curated resource that combines 23,377 protein interactions between human host and 432 unique pathogens involved in CVDs in a single intuitive web application. It covers endocarditis, myocarditis, pericarditis and 16 other microbe induced CVDs. The HPI information has been compiled, curated, and presented in a freely accessible web interface (http://morcvd.sblab-nsit.net/About). Apart from organization, enrichment of the HPI data was done by adding hyperlinked protein ID, PubMed, gene ontology records. For each protein in the database, drug target and interactors (same as well as different species) information has been provided. The database can be searched by disease, protein ID, pathogen name or interaction detection method. Interactions detected by more than one method can also be listed. The information can be presented in tabular form or downloaded. A comprehensive help file has been developed to explain the various options available. Hence, MorCVD acts as a unified resource for retrieval of HPI data for researchers in CVD and microbiology.
Collapse
|
468
|
Guo S, Zhou Y, Zeng P, Xu G, Wang G, Cui Q. Identification and analysis of the human sex-biased genes. Brief Bioinform 2019; 19:188-198. [PMID: 28028006 DOI: 10.1093/bib/bbw125] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Indexed: 01/28/2023] Open
Abstract
Tremendous differences between human sexes are universally observed. Therefore, identifying and analyzing the sex-biased genes are becoming basically important for uncovering the mystery of sex differences and personalized medicine. Here, we presented a computational method to identify sex-biased genes from public gene expression databases. We obtained 1407 female-biased genes (FGs) and 1096 male-biased genes (MGs) across 14 different tissues. Bioinformatics analysis revealed that compared with MGs, FGs have higher evolutionary rate, higher single-nucleotide polymorphism density, less homologous gene numbers and smaller phyletic age. FGs have lower expression level, higher tissue specificity and later expressed stage in body development. Moreover, FGs are highly involved in immune-related functions, whereas MGs are more enriched in metabolic process. In addition, cellular network analysis revealed that MGs have higher degree, more cellular activating signaling and tend to be located in cellular inner space, whereas FGs have lower degree, more cellular repressing signaling and tend to be located in cellular outer space. Finally, the identified sex-biased genes and the discovered biological insights together can be a valuable resource helpful for investigating sex-biased physiology and medicine, for example sex-biased disease diagnosis and therapy, which represents one important aspect of personalized and precision medicine.
Collapse
Affiliation(s)
- Sisi Guo
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, China
| | - Pan Zeng
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, China
| | - Guoheng Xu
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, China
| | - Guoqing Wang
- Department of Pathogenobiology, College of Basic Medicine, Jilin University, Changchun, Jilin, China
| | - Qinghua Cui
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Beijing, China
| |
Collapse
|
469
|
He F, Berg A, Imamura Kawasawa Y, Bixler EO, Fernandez-Mendoza J, Whitsel EA, Liao D. Association between DNA methylation in obesity-related genes and body mass index percentile in adolescents. Sci Rep 2019; 9:2079. [PMID: 30765773 PMCID: PMC6375997 DOI: 10.1038/s41598-019-38587-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/21/2018] [Indexed: 12/16/2022] Open
Abstract
Childhood obesity remains an epidemic in the U.S. and worldwide. However, little is understood regarding the epigenetic basis of obesity in adolescents. To investigate the cross-sectional association between DNA methylation level in obesity-related genes and body mass index (BMI) percentile, data from 263 adolescents in the population-based Penn State Child Cohort follow-up exam was analysed. Using DNA extracted from peripheral leukocytes, epigenome-wide single nucleotide resolution of DNA methylation in cytosine-phosphate-guanine (CpG) sites and surrounding regions was obtained. We used multivariable-adjusted linear regression models to assess the association between site-specific methylation level and age- and sex-specific BMI percentile. Hypergeometric and permutation tests were used to determine if obesity-related genes were significantly enriched among all intragenic sites that achieved a p < 0.05 throughout the epigenome. Among the 5,669 sites related to BMI percentile with p < 0.05, 28 were identified within obesity-related genes. Obesity-related genes were significantly enriched among 103,466 intragenic sites (Phypergeometric = 0.006; Ppermutation = 0.006). Moreover, increased methylation on one site within SIM1 was significantly related to higher BMI percentile (P = 4.2E-05). If externally validated, our data would suggest that DNA methylation in obesity-related genes may relate to obesity risk in adolescents.
Collapse
Affiliation(s)
- Fan He
- Department of Public Health Sciences, the Pennsylvania State University College of Medicine, Hershey, 17033, Pennsylvania, USA
| | - Arthur Berg
- Department of Public Health Sciences, the Pennsylvania State University College of Medicine, Hershey, 17033, Pennsylvania, USA
| | - Yuka Imamura Kawasawa
- Institute for Personalized Medicine, Departments of Biochemistry and Molecular Biology and Pharmacology, the Pennsylvania State University College of Medicine, Hershey, 17033, Pennsylvania, USA
| | - Edward O Bixler
- Sleep Research and Treatment Center, Department of Psychiatry, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania, 17033, USA
| | - Julio Fernandez-Mendoza
- Sleep Research and Treatment Center, Department of Psychiatry, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania, 17033, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health; Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Duanping Liao
- Department of Public Health Sciences, the Pennsylvania State University College of Medicine, Hershey, 17033, Pennsylvania, USA.
| |
Collapse
|
470
|
Hu Y, Zhao T, Zang T, Zhang Y, Cheng L. Identification of Alzheimer's Disease-Related Genes Based on Data Integration Method. Front Genet 2019; 9:703. [PMID: 30740125 PMCID: PMC6355707 DOI: 10.3389/fgene.2018.00703] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/14/2018] [Indexed: 01/18/2023] Open
Abstract
Alzheimer disease (AD) is the fourth major cause of death in the elderly following cancer, heart disease and cerebrovascular disease. Finding candidate causal genes can help in the design of Gene targeted drugs and effectively reduce the risk of the disease. Complex diseases such as AD are usually caused by multiple genes. The Genome-wide association study (GWAS), has identified the potential genetic variants for most diseases. However, because of linkage disequilibrium (LD), it is difficult to identify the causative mutations that directly cause diseases. In this study, we combined expression quantitative trait locus (eQTL) studies with the GWAS, to comprehensively define the genes that cause Alzheimer disease. The method used was the Summary Mendelian randomization (SMR), which is a novel method to integrate summarized data. Two GWAS studies and five eQTL studies were referenced in this paper. We found several candidate SNPs that have a strong relationship with AD. Most of these SNPs overlap in different data sets, providing relatively strong reliability. We also explain the function of the novel AD-related genes we have discovered.
Collapse
Affiliation(s)
- Yang Hu
- Department of Computer Science and Technology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Tianyi Zhao
- Department of Computer Science and Technology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Tianyi Zang
- Department of Computer Science and Technology, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ying Zhang
- Department of Rehabilitation, Heilongjiang Province Land Reclamation Headquarters General Hospital, Harbin, China
| | - Liang Cheng
- Department of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
471
|
Baig MH, Rashid I, Srivastava P, Ahmad K, Jan AT, Rabbani G, Choi D, Barreto GE, Ashraf GM, Lee EJ, Choi I. NeuroMuscleDB: a Database of Genes Associated with Muscle Development, Neuromuscular Diseases, Ageing, and Neurodegeneration. Mol Neurobiol 2019; 56:5835-5843. [PMID: 30684219 DOI: 10.1007/s12035-019-1478-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/10/2019] [Indexed: 12/25/2022]
Abstract
Skeletal muscle is a highly complex, heterogeneous tissue that serves a multitude of biological functions in living organisms. With the advent of methods, such as microarrays, transcriptome analysis, and proteomics, studies have been performed at the genome level to gain insight of changes in the expression profiles of genes during different stages of muscle development and of associated diseases. In the present study, a database was conceived for the straightforward retrieval of information on genes involved in skeletal muscle formation, neuromuscular diseases (NMDs), ageing, and neurodegenerative disorders (NDs). The resulting database named NeuroMuscleDB ( http://yu-mbl-muscledb.com/NeuroMuscleDB ) is the result of a wide literature survey, database searches, and data curation. NeuroMuscleDB contains information of genes in Homo sapiens, Mus musculus, and Bos Taurus, and their promoter sequences and specified roles at different stages of muscle development and in associated myopathies. The database contains information on ~ 1102 genes, 6030 mRNAs, and 5687 proteins, and embedded analytical tools that can be used to perform tasks related to gene sequence usage. The authors believe NeuroMuscleDB provides a platform for obtaining desired information on genes related to myogenesis and their associations with various diseases (NMDs, ageing, and NDs). NeuroMuscleDB is freely available on the web at http://yu-mbl-muscledb.com/NeuroMuscleDB and supports all major browsers.
Collapse
Affiliation(s)
- Mohammad Hassan Baig
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Iliyas Rashid
- Amity Institute of Biotechnology, Amity University, Lucknow, Uttar Pradesh, 226 028, India
| | - Prachi Srivastava
- Amity Institute of Biotechnology, Amity University, Lucknow, Uttar Pradesh, 226 028, India
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, 185236, India
| | - Gulam Rabbani
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Dukhwan Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
472
|
Michaelovsky E, Carmel M, Frisch A, Salmon-Divon M, Pasmanik-Chor M, Weizman A, Gothelf D. Risk gene-set and pathways in 22q11.2 deletion-related schizophrenia: a genealogical molecular approach. Transl Psychiatry 2019; 9:15. [PMID: 30710087 PMCID: PMC6358611 DOI: 10.1038/s41398-018-0354-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 12/05/2018] [Accepted: 12/10/2018] [Indexed: 11/15/2022] Open
Abstract
The 22q11.2 deletion is a strong, but insufficient, "first hit" genetic risk factor for schizophrenia (SZ). We attempted to identify "second hits" from the entire genome in a unique multiplex 22q11.2 deletion syndrome (DS) family. Bioinformatic analysis of whole-exome sequencing and comparative-genomic hybridization array identified de novo and inherited, rare and damaging variants, including copy number variations, outside the 22q11.2 region. A specific 22q11.2-haplotype was associated with psychosis. The interaction of the identified "second hits" with the 22q11.2 haploinsufficiency may affect neurodevelopmental processes, including neuron projection, cytoskeleton activity, and histone modification in 22q11.2DS-ralated psychosis. A larger load of variants, involved in neurodevelopment, in combination with additional molecular events that affect sensory perception, olfactory transduction and G-protein-coupled receptor signaling may account for the development of 22q11.2DS-related SZ. Comprehensive analysis of multiplex families is a promising approach to the elucidation of the molecular pathophysiology of 22q11.2DS-related SZ with potential relevance to treatment.
Collapse
Affiliation(s)
- Elena Michaelovsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Felsenstein Medical Research Center, Petah Tikva, Israel.
| | - Miri Carmel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Amos Frisch
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Petah Tikva, Israel
| | | | - Metsada Pasmanik-Chor
- Bioinformatics Unit, G.S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Abraham Weizman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Petah Tikva, Israel
- Geha Mental Health Center, Petah Tikva, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Doron Gothelf
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Behavioral Neurogenetics Center, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
473
|
Bo C, Wang J, Zhang H, Cao Y, Lu X, Wang T, Wang Y, Li S, Kong X, Sun X, Liu Z, Ning S, Wang L. Global pathway view analysis of microRNA clusters in myasthenia gravis. Mol Med Rep 2019; 19:2350-2360. [PMID: 30664201 DOI: 10.3892/mmr.2019.9845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/25/2018] [Indexed: 11/05/2022] Open
Abstract
The significant roles of microRNAs (miRNAs) in the pathogenesis of myasthenia gravis (MG) have been observed in numerous previous studies. The impact of miRNA clusters on immunity has been demonstrated in previous years; however, the regulation of miRNA clusters in MG remains to be elucidated. In the present study, 245 MG risk genes were collected and 99 MG risk pathways enriched by these genes were identified. A catalog of 126 MG risk miRNAs was then created; the MG risk miRNAs were located on each chromosome and a miRNA cluster was defined as a number of miRNAs with a relative distance of <6 kb on the same sub‑band, same band, same region and same chromosome. Furthermore, enrichment analyses were performed using the target genes of the MG risk miRNA clusters, and a number of risk pathways of each miRNA clusters were identified. As a result, 15 significant miRNA clusters associated with MG were identified. Additionally, the most significant pathways of the miRNA clusters were identified to be enriched on chromosomes 9, 19 and 22, characterized by immunity, infection and carcinoma, suggesting that the mechanism of MG may be associated with certain abnormalities of miRNA clusters on chromosomes 9, 19 and 22. The present study provides novel insight into a global pathway view of miRNA clusters in the pathogenesis of MG.
Collapse
Affiliation(s)
- Chunrui Bo
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Huixue Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yuze Cao
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoyu Lu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Tianfeng Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yu Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shuang Li
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaotong Kong
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xuesong Sun
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhaojun Liu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
474
|
Wang J, Dai X, Berry LD, Cogan JD, Liu Q, Shyr Y. HACER: an atlas of human active enhancers to interpret regulatory variants. Nucleic Acids Res 2019; 47:D106-D112. [PMID: 30247654 PMCID: PMC6323890 DOI: 10.1093/nar/gky864] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 12/19/2022] Open
Abstract
Recent studies have shown that disease-susceptibility variants frequently lie in cell-type-specific enhancer elements. To identify, interpret, and prioritize such risk variants, we must identify the enhancers active in disease-relevant cell types, their upstream transcription factor (TF) binding, and their downstream target genes. To address this need, we built HACER (http://bioinfo.vanderbilt.edu/AE/HACER/), an atlas of Human ACtive Enhancers to interpret Regulatory variants. The HACER atlas catalogues and annotates in-vivo transcribed cell-type-specific enhancers, as well as placing enhancers within transcriptional regulatory networks by integrating ENCODE TF ChIP-Seq and predicted/validated chromatin interaction data. We demonstrate the utility of HACER in (i) offering a mechanistic hypothesis to explain the association of SNP rs614367 with ER-positive breast cancer risk, (ii) exploring tumor-specific enhancers in selective MYC dysregulation and (iii) prioritizing/annotating non-coding regulatory regions targeting CCND1. HACER provides a valuable resource for studies of GWAS, non-coding variants, and enhancer-mediated regulation.
Collapse
Affiliation(s)
- Jing Wang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xizhen Dai
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lynne D Berry
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joy D Cogan
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
475
|
Network Pharmacology Integrated Molecular Docking Reveals the Antiosteosarcoma Mechanism of Biochanin A. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1410495. [PMID: 30723510 PMCID: PMC6339762 DOI: 10.1155/2019/1410495] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
Background As the malignant tumor with the highest incidence in teenagers, osteosarcoma has become a major problem in oncology research. In addition to surgical management, the pharmacotherapeutic strategy for osteosarcoma treatment is an attractive way to explore. It has been demonstrated that biochanin A has an antitumor capacity on multiple kinds of solid tumor, including osteosarcoma. But the precise mechanism of biochanin A against osteosarcoma is still needed to be discovered. Objective To identify the potential therapeutic targets of biochanin A in treating osteosarcoma. Methods In present study, an integrated approach including network pharmacology and molecular docking technique was conducted, which mainly comprises target prediction, network construction, gene ontology, and pathway enrichment. CCK8 test was employed to evaluate the cell viability of MG63 cells. Western-blot was used to verify the target proteins of biochanin A. Results Ninety-six and 114 proteins were obtained as the targets of biochanin A and osteosarcoma, respectively. TP53, IGF1, JUN, BGLAP, ATM, MAPK1, ATF3, H2AFX, BAX, CDKN2A, and EGF were identified as the potential targets of biochanin A against osteosarcoma. Based on the western-blot detection, the expression of BGLAP, BAX, and ATF3 in MG63 cell line changed under the treatment of biochanin A. Conclusion Biochanin A can effectively suppress the proliferation of osteosarcoma and regulate the expression of BGLAP, BAX, and ATF3, which may act as the potential therapeutic targets of osteosarcoma.
Collapse
|
476
|
Hu DL, Wang G, Yu J, Zhang LH, Huang YF, Wang D, Zhou HH. Epigallocatechin‑3‑gallate modulates long non‑coding RNA and mRNA expression profiles in lung cancer cells. Mol Med Rep 2019; 19:1509-1520. [PMID: 30628683 PMCID: PMC6390008 DOI: 10.3892/mmr.2019.9816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 10/26/2018] [Indexed: 01/23/2023] Open
Abstract
(−)-Epigallocatechin-3-gallate (EGCG), a major constituent of green tea, is a potential anticancer agent, but the molecular mechanisms of its effects are not well-understood. The present study was conducted to examine the mechanism of EGCG in lung cancer cells. Alterations in long non-coding RNAs (lncRNAs) and mRNAs were investigated in lung cancer cells treated with EGCG by lncRNA microarray analysis. Furthermore, the functions and signaling pathways regulated by EGCG were predicted by bioinformatics analysis. A total of 960 lncRNAs and 1,434 mRNAs were significantly altered following EGCG treatment. These lncRNAs were distributed across nearly all human chromosomes and the mRNAs were involved in the cell cycle and the mitotic cell cycle process. Through a combination of microarray and bioinformatics analysis, 20 mRNAs predicted to serve a key role in the EGCG regulation were identified, and certain regulatory networks involving EGCG-regulated lncRNAs were predicted. In conclusion, EGCG affects the expression of various lncRNAs and mRNAs in the cells, therefore affecting cell functions. The results of the present study provide an insight into the mechanism of EGCG, which may be useful for therapeutic development.
Collapse
Affiliation(s)
- Dong-Li Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jing Yu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Li-Hua Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yuan-Fei Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Dan Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
477
|
Zhang SY, Zhang SW, Fan XN, Meng J, Chen Y, Gao SJ, Huang Y. Global analysis of N6-methyladenosine functions and its disease association using deep learning and network-based methods. PLoS Comput Biol 2019; 15:e1006663. [PMID: 30601803 PMCID: PMC6331136 DOI: 10.1371/journal.pcbi.1006663] [Citation(s) in RCA: 35] [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: 06/01/2018] [Revised: 01/14/2019] [Accepted: 11/21/2018] [Indexed: 02/03/2023] Open
Abstract
N6-methyladenosine (m6A) is the most abundant methylation, existing in >25% of human mRNAs. Exciting recent discoveries indicate the close involvement of m6A in regulating many different aspects of mRNA metabolism and diseases like cancer. However, our current knowledge about how m6A levels are controlled and whether and how regulation of m6A levels of a specific gene can play a role in cancer and other diseases is mostly elusive. We propose in this paper a computational scheme for predicting m6A-regulated genes and m6A-associated disease, which includes Deep-m6A, the first model for detecting condition-specific m6A sites from MeRIP-Seq data with a single base resolution using deep learning and Hot-m6A, a new network-based pipeline that prioritizes functional significant m6A genes and its associated diseases using the Protein-Protein Interaction (PPI) and gene-disease heterogeneous networks. We applied Deep-m6A and this pipeline to 75 MeRIP-seq human samples, which produced a compact set of 709 functionally significant m6A-regulated genes and nine functionally enriched subnetworks. The functional enrichment analysis of these genes and networks reveal that m6A targets key genes of many critical biological processes including transcription, cell organization and transport, and cell proliferation and cancer-related pathways such as Wnt pathway. The m6A-associated disease analysis prioritized five significantly associated diseases including leukemia and renal cell carcinoma. These results demonstrate the power of our proposed computational scheme and provide new leads for understanding m6A regulatory functions and its roles in diseases.
Collapse
Affiliation(s)
- Song-Yao Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi’an, China
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi’an, China
| | - Xiao-Nan Fan
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi’an, China
| | - Jia Meng
- Department of Biological Sciences, HRINU, SUERI, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu, China
| | - Yidong Chen
- Department of Epidemiology and Biostatistics, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Shou-Jiang Gao
- UPMC Hillman Cancer Center and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yufei Huang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
- Department of Epidemiology and Biostatistics, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
478
|
Yan C, Wang J, Ni P, Lan W, Wu FX, Pan Y. DNRLMF-MDA:Predicting microRNA-Disease Associations Based on Similarities of microRNAs and Diseases. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:233-243. [PMID: 29990253 DOI: 10.1109/tcbb.2017.2776101] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
MicroRNAs (miRNAs) are a class of non-coding RNAs about ∼ 22nt nucleotides. Studies have proven that miRNAs play key roles in many human complex diseases. Therefore, discovering miRNA-disease associations is beneficial to understanding disease mechanisms, developing drugs, and treating complex diseases. It is well known that it is a time-consuming and expensive process to discover the miRNA-disease associations via biological experiments. Alternatively, computational models could provide a low-cost and high-efficiency way for predicting miRNA-disease associations. In this study, we propose a method (called DNRLMF-MDA) to predict miRNA-disease associations based on dynamic neighborhood regularized logistic matrix factorization. DNRLMF-MDA integrates known miRNA-disease associations, functional similarity and Gaussian Interaction Profile (GIP) kernel similarity of miRNAs, and functional similarity and GIP kernel similarity of diseases. Especially, positive observations (known miRNA-disease associations) are assigned higher importance levels than negative observations (unknown miRNA-disease associations).DNRLMF-MDA computes the probability that a miRNA would interact with a disease by a logistic matrix factorization method, where latent vectors of miRNAs and diseases represent the properties of miRNAs and diseases, respectively, and further improve prediction performance via dynamic neighborhood regularized. The 5-fold cross validation is adopted to assess the performance of our DNRLMF-MDA, as well as other competing methods for comparison. The computational experiments show that DNRLMF-MDA outperforms the state-of-art method PBMDA. The AUC values of DNRLMF-MDA on three datasets are 0.9357, 0.9411, and 0.9416, respectively, which are superior to the PBMDA's results of 0.9218, 0.9187, and 0.9262. The average computation times per 5-fold cross validation of DNRLMF-MDA on three datasets are 38, 46, and 50 seconds, which are shorter than the PBMDA's average computation times of 10869, 916, and 8448 seconds, respectively. DNRLMF-MDA also can predict potential diseases for new miRNAs. Furthermore, case studies illustrate that DNRLMF-MDA is an effective method to predict miRNA-disease associations.
Collapse
|
479
|
Abstract
Computational prediction of the clinical success or failure of a potential drug target for therapeutic use is a challenging problem. Novel network propagation algorithms that integrate heterogeneous biological networks are proving useful for drug target identification and prioritization. These approaches typically utilize a network describing relationships between targets, a method to disseminate the relevant information through the network, and a method to elucidate new associations between targets and diseases. Here, we utilize one such network propagation-based approach, DTINet, which starts with diffusion component analysis of networks of both potential drug targets and diseases. Then an inductive matrix completion algorithm is applied to identify novel disease targets based on their network topological similarities with known disease targets with successfully launched drugs. DTINet performed well as assessed with area under the precision-recall curve (AUPR = 0.88 ± 0.007) and area under the receiver operating characteristic curve (AUROC = 0.86 ± 0.008). These metrics improved when we combined data from multiple networks in the target space but reduced significantly when we used a more conservative method to define negative controls (AUPR = 0.56 ± 0.007, AUROC = 0.57 ± 0.007). We are optimistic that integration of more relevant and cleaner datasets and networks, careful calibration of model parameters, as well as algorithmic improvements will improve prediction accuracy. However, we also recognize that predicting drug targets that are likely to be successful is an extremely challenging problem due to its complex nature and sparsity of known disease targets.
Collapse
|
480
|
Cheng F. Cardio-oncology: Network-Based Prediction of Cancer Therapy-Induced Cardiotoxicity. CHALLENGES AND ADVANCES IN COMPUTATIONAL CHEMISTRY AND PHYSICS 2019:75-97. [DOI: 10.1007/978-3-030-16443-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
481
|
Izadi F. Differential Connectivity in Colorectal Cancer Gene Expression Network. IRANIAN BIOMEDICAL JOURNAL 2019; 23. [PMID: 29843204 PMCID: PMC6305824 DOI: 10.29252/.23.1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the challenging types of cancers; thus, exploring effective biomarkers related to colorectal could lead to significant progresses toward the treatment of this disease. METHODS In the present study, CRC gene expression datasets have been reanalyzed. Mutual differentially expressed genes across 294 normal mucosa and adjacent tumoral samples were then utilized in order to build two independent transcriptional regulatory networks. By analyzing the networks topologically, genes with differential global connectivity related to cancer state were determined for which the potential transcriptional regulators including transcription factors were identified. RESULTS The majority of differentially connected genes (DCGs) were up-regulated in colorectal transcriptome experiments. Moreover, a number of these genes have been experimentally validated as cancer or CRC-associated genes. The DCGs, including GART, TGFB1, ITGA2, SLC16A5, SOX9, and MMP7, were investigated across 12 cancer types. Functional enrichment analysis followed by detailed data mining exhibited that these candidate genes could be related to CRC by mediating in metastatic cascade in addition to shared pathways with 12 cancer types by triggering the inflammatory events. DISCUSSION Our study uncovered correlated alterations in gene expression related to CRC susceptibility and progression that the potent candidate biomarkers could provide a link to disease.
Collapse
Affiliation(s)
- Fereshteh Izadi
- Sari Agricultural Sciences and Natural Resources University (SANRU), Farah Abad Road, Mazandaran 4818168984, Iran,Corresponding Author: Fereshteh Izadi Sari Agricultural Sciences and Natural Resources University (SANRU), Farah Abad Road, Mazandaran 4818168984, Iran; Mobile: (+98-918) 6291302; E-mail:
| |
Collapse
|
482
|
Damle NP, Köhn M. The human DEPhOsphorylation Database DEPOD: 2019 update. Database (Oxford) 2019; 2019:baz133. [PMID: 31836896 PMCID: PMC6911163 DOI: 10.1093/database/baz133] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/06/2019] [Accepted: 10/29/2019] [Indexed: 01/04/2023]
Abstract
The human Dephosphorylation Database (DEPOD) is a manually curated resource that harbors human phosphatases, their protein and non-protein substrates, dephosphorylation sites and the associated signaling pathways. We report here an update to DEPOD by integrating and/or linking to annotations from 69 other open access databases including disease associations, phosphorylating kinases, protein interactions, and also genome browsers. We also provide tools to visualize protein interactions, protein structures, phosphorylation networks, evolutionary conservation of proteins, dephosphorylation sites, and short linear motifs within various proteins. The updated version of DEPOD contains 254 human phosphatases, 336 protein and 83 non-protein substrates, and 1215 manually curated phosphatase-substrate relationships. In addition, we have improved the data access as all the data in DEPOD can now be easily downloaded in a user-friendly format. With multiple significant improvements, DEPOD continues serving as a key resource for research on phosphatase-kinase networks. Database URL: www.depod.org.
Collapse
Affiliation(s)
- Nikhil P Damle
- Signalling Research Centres BIOSS and CIBSS, Faculty of Biology, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Maja Köhn
- Signalling Research Centres BIOSS and CIBSS, Faculty of Biology, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| |
Collapse
|
483
|
Osmak GJ, Matveeva NA, Titov BV, Favorova OO. The Myocardial Infarction Associated Variant in the MIR196A2 Gene and Presumable Signaling Pathways to Involve miR-196a2 in the Pathological Phenotype. Mol Biol 2018. [DOI: 10.1134/s0026893318060146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
484
|
Perscheid C, Grasnick B, Uflacker M. Integrative Gene Selection on Gene Expression Data: Providing Biological Context to Traditional Approaches. J Integr Bioinform 2018; 16:/j/jib.ahead-of-print/jib-2018-0064/jib-2018-0064.xml. [PMID: 30785707 PMCID: PMC6798862 DOI: 10.1515/jib-2018-0064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/12/2018] [Indexed: 12/30/2022] Open
Abstract
The advance of high-throughput RNA-Sequencing techniques enables researchers to analyze the complete gene activity in particular cells. From the insights of such analyses, researchers can identify disease-specific expression profiles, thus understand complex diseases like cancer, and eventually develop effective measures for diagnosis and treatment. The high dimensionality of gene expression data poses challenges to its computational analysis, which is addressed with measures of gene selection. Traditional gene selection approaches base their findings on statistical analyses of the actual expression levels, which implies several drawbacks when it comes to accurately identifying the underlying biological processes. In turn, integrative approaches include curated information on biological processes from external knowledge bases during gene selection, which promises to lead to better interpretability and improved predictive performance. Our work compares the performance of traditional and integrative gene selection approaches. Moreover, we propose a straightforward approach to integrate external knowledge with traditional gene selection approaches. We introduce a framework enabling the automatic external knowledge integration, gene selection, and evaluation. Evaluation results prove our framework to be a useful tool for evaluation and show that integration of external knowledge improves overall analysis results.
Collapse
Affiliation(s)
- Cindy Perscheid
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| | - Bastien Grasnick
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| | - Matthias Uflacker
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| |
Collapse
|
485
|
Zimmermann MT. The Importance of Biologic Knowledge and Gene Expression Context for Genomic Data Interpretation. Front Genet 2018; 9:670. [PMID: 30619486 PMCID: PMC6305277 DOI: 10.3389/fgene.2018.00670] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/04/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Genomic sequencing, including whole exome sequencing (WES), is enabling a higher resolution for defining diseases, understand mechanisms, and improving the practice of clinical care. However, WES routinely identifies genomic variants with uncertain functional effects. Furthering uncertainty in WES data interpretation is that many genes can express multiple transcripts and their relative expression may differ by body tissue. In order to interpret WES data, we not only need to understand which transcript is most relevant, but what tissue is most relevant. Methods: In this work, we quantify how frequently differences in transcript and tissue expression affect WES data interpretation at gene, pathway, disease, and biologic network levels. We combined and analyzed multiple large and publically available datasets to inform genomic data interpretation. Results: Across well-established biologic pathways and genes with pathogenic disease variants, 54 and 40% have a different protein coding effect by transcript selection for, respectively, 25 and 50% of the genes contained. Additionally, strong differences in human tissue expression levels affect 33 and 19% of the same set of pathways and diseases for, respectively, 25 and 50% of the genes contained. Conclusion: Whole exome sequencing identifies genomic variants, but to interpret the functional effects of those variants in high-resolution, we recommend building transcript selection and cross-tissue gene expression levels into hypotheses and analyses. Using current large-scale data, we show how extensively interpretation of genomic variants may differ according to transcript and tissue, across most pathways and disease. Thus, their inclusion is necessary for WES data interpretation.
Collapse
Affiliation(s)
- Michael T. Zimmermann
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
486
|
Leng L, Zhang C, Ren L, Li Q. Construction of a long non‑coding RNA-mediated competitive endogenous RNA network reveals global patterns and regulatory markers in gestational diabetes. Int J Mol Med 2018; 43:927-935. [PMID: 30569156 PMCID: PMC6317690 DOI: 10.3892/ijmm.2018.4026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a common disease affecting pregnant women. Recent studies have suggested that competing endogenous RNAs (ceRNAs), which compete with long non‑coding RNAs (lncRNAs) for microRNA (miRNA or miR) binding and indirectly regulate miRNA targets through competing interactions, play a critical role in disease. In this study, we present a computationally integrated approach with which to construct a lncRNA‑mediated ceRNA network (LCEN) in GDM by integrating RNA interactions and expression data. lncRNAs exhibited specific features and played critical roles in GDM‑associated LCEN. The construction of a global functional score profile revealed that ceRNAs had a high activity in GDM. We extracted several ceRNA modules and demonstrated that these modules had increased close interactions. We further discovered that these ceRNA modules may be utilized as specific and effective circulating biomarkers for GDM. Finally, functional analyses demonstrated that the GDM‑associated ceRNAs participated in the regulation of irisin and the thyroid hormone signaling pathway. It was suggested that there were close associations between the thyroid hormone and GDM. Collectively, ceRNAs may accelerate biomarker discovery and therapeutic development in GDM.
Collapse
Affiliation(s)
- Lei Leng
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Chengwei Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Lihong Ren
- Department of Endocrinology, The Second Hospital of Harbin, Harbin, Heilongjiang 150006, P.R. China
| | - Qiang Li
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| |
Collapse
|
487
|
Westwood S, Baird AL, Hye A, Ashton NJ, Nevado-Holgado AJ, Anand SN, Liu B, Newby D, Bazenet C, Kiddle SJ, Ward M, Newton B, Desai K, Tan Hehir C, Zanette M, Galimberti D, Parnetti L, Lleó A, Baker S, Narayan VA, van der Flier WM, Scheltens P, Teunissen CE, Visser PJ, Lovestone S. Plasma Protein Biomarkers for the Prediction of CSF Amyloid and Tau and [ 18F]-Flutemetamol PET Scan Result. Front Aging Neurosci 2018; 10:409. [PMID: 30618716 PMCID: PMC6297196 DOI: 10.3389/fnagi.2018.00409] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Blood biomarkers may aid in recruitment to clinical trials of Alzheimer's disease (AD) modifying therapeutics by triaging potential trials participants for amyloid positron emission tomography (PET) or cerebrospinal fluid (CSF) Aβ and tau tests. Objective: To discover a plasma proteomic signature associated with CSF and PET measures of AD pathology. Methods: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) based proteomics were performed in plasma from participants with subjective cognitive decline (SCD), mild cognitive impairment (MCI), and AD, recruited to the Amsterdam Dementia Cohort, stratified by CSF Tau/Aβ42 (n = 50). Technical replication and independent validation were performed by immunoassay in plasma from SCD, MCI, and AD participants recruited to the Amsterdam Dementia Cohort with CSF measures (n = 100), MCI participants enrolled in the GE067-005 study with [18F]-Flutemetamol PET amyloid measures (n = 173), and AD, MCI and cognitively healthy participants from the EMIF 500 study with CSF Aβ42 measurements (n = 494). Results: 25 discovery proteins were nominally associated with CSF Tau/Aβ42 (P < 0.05) with associations of ficolin-2 (FCN2), apolipoprotein C-IV and fibrinogen β chain confirmed by immunoassay (P < 0.05). In the GE067-005 cohort, FCN2 was nominally associated with PET amyloid (P < 0.05) replicating the association with CSF Tau/Aβ42. There were nominally significant associations of complement component 3 with PET amyloid, and apolipoprotein(a), apolipoprotein A-I, ceruloplasmin, and PPY with MCI conversion to AD (all P < 0.05). In the EMIF 500 cohort FCN2 was trending toward a significant relationship with CSF Aβ42 (P ≈ 0.05), while both A1AT and clusterin were nominally significantly associated with CSF Aβ42 (both P < 0.05). Conclusion: Associations of plasma proteins with multiple measures of AD pathology and progression are demonstrated. To our knowledge this is the first study to report an association of FCN2 with AD pathology. Further testing of the proteins in larger independent cohorts will be important.
Collapse
Affiliation(s)
- Sarah Westwood
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Alison L. Baird
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kigndom
- Biomedical Research Unit for Dementia, NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Nicholas J. Ashton
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kigndom
- Biomedical Research Unit for Dementia, NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Foundation Trust, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | | | - Sneha N. Anand
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Benjamine Liu
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Chantal Bazenet
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kigndom
| | - Steven J. Kiddle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- MRC Biostatistics Unit, Cambridge Biomedical Campus, Cambridge Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Malcolm Ward
- Proteomics Facility, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Ben Newton
- GE Healthcare Life Sciences Core Imaging, London, United Kingdom
| | - Keyur Desai
- Biosciences, GE Global Research, Niskayuna, NY, United States
| | | | - Michelle Zanette
- GE Healthcare Life Sciences Core Imaging, Marlborough, MA, United States
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Centro Dino Ferrari, University of Milan, Milan, Italy
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lucilla Parnetti
- Center for Memory Disorders and Laboratory of Clinical Neurochemistry, Neurology Clinic, University of Perugia, Perugia, Italy
| | - Alberto Lleó
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susan Baker
- Janssen Neuroscience Research & Development, Titusville, NJ, United States
| | - Vaibhav A. Narayan
- Janssen Neuroscience Research & Development, Titusville, NJ, United States
| | - Wiesje M. van der Flier
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
488
|
Swindell WR, Bojanowski K, Kindy MS, Chau RMW, Ko D. GM604 regulates developmental neurogenesis pathways and the expression of genes associated with amyotrophic lateral sclerosis. Transl Neurodegener 2018; 7:30. [PMID: 30524706 PMCID: PMC6276193 DOI: 10.1186/s40035-018-0135-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/21/2018] [Indexed: 12/11/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is currently an incurable disease without highly effective pharmacological treatments. The peptide drug GM604 (GM6 or Alirinetide) was developed as a candidate ALS therapy, which has demonstrated safety and good drug-like properties with a favorable pharmacokinetic profile. GM6 is hypothesized to bolster neuron survival through the multi-target regulation of developmental pathways, but mechanisms of action are not fully understood. Methods This study used RNA-seq to evaluate transcriptome responses in SH-SY5Y neuroblastoma cells following GM6 treatment (6, 24 and 48 h). Results We identified 2867 protein-coding genes with expression significantly altered by GM6 (FDR < 0.10). Early (6 h) responses included up-regulation of Notch and hedgehog signaling components, with increased expression of developmental genes mediating neurogenesis and axon growth. Prolonged GM6 treatment (24 and 48 h) altered the expression of genes contributing to cell adhesion and the extracellular matrix. GM6 further down-regulated the expression of genes associated with mitochondria, inflammatory responses, mRNA processing and chromatin organization. GM6-increased genes were located near GC-rich motifs interacting with C2H2 zinc finger transcription factors, whereas GM6-decreased genes were located near AT-rich motifs associated with helix-turn-helix homeodomain factors. Such motifs interacted with a diverse network of transcription factors encoded by GM6-regulated genes (STAT3, HOXD11, HES7, GLI1). We identified 77 ALS-associated genes with expression significantly altered by GM6 treatment (FDR < 0.10), which were known to function in neurogenesis, axon guidance and the intrinsic apoptosis pathway. Conclusions Our findings support the hypothesis that GM6 acts through developmental-stage pathways to influence neuron survival. Gene expression responses were consistent with neurotrophic effects, ECM modulation, and activation of the Notch and hedgehog neurodevelopmental pathways. This multifaceted mechanism of action is unique among existing ALS drug candidates and may be applicable to multiple neurodegenerative diseases. Electronic supplementary material The online version of this article (10.1186/s40035-018-0135-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William R Swindell
- 1Heritage College of Osteopathic Medicine, Ohio University, Athens, OH USA
| | | | - Mark S Kindy
- 3Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL USA.,4James A. Haley VAMC, Tampa, FL USA
| | | | - Dorothy Ko
- Genervon Biopharmaceuticals LLC, Pasadena, CA USA
| |
Collapse
|
489
|
Wilson JL, Racz R, Liu T, Adeniyi O, Sun J, Ramamoorthy A, Pacanowski M, Altman R. PathFX provides mechanistic insights into drug efficacy and safety for regulatory review and therapeutic development. PLoS Comput Biol 2018; 14:e1006614. [PMID: 30532240 PMCID: PMC6285459 DOI: 10.1371/journal.pcbi.1006614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Failure to demonstrate efficacy and safety issues are important reasons that drugs do not reach the market. An incomplete understanding of how drugs exert their effects hinders regulatory and pharmaceutical industry projections of a drug's benefits and risks. Signaling pathways mediate drug response and while many signaling molecules have been characterized for their contribution to disease or their role in drug side effects, our knowledge of these pathways is incomplete. To better understand all signaling molecules involved in drug response and the phenotype associations of these molecules, we created a novel method, PathFX, a non-commercial entity, to identify these pathways and drug-related phenotypes. We benchmarked PathFX by identifying drugs' marketed disease indications and reported a sensitivity of 41%, a 2.7-fold improvement over similar approaches. We then used PathFX to strengthen signals for drug-adverse event pairs occurring in the FDA Adverse Event Reporting System (FAERS) and also identified opportunities for drug repurposing for new diseases based on interaction paths that associated a marketed drug to that disease. By discovering molecular interaction pathways, PathFX improved our understanding of drug associations to safety and efficacy phenotypes. The algorithm may provide a new means to improve regulatory and therapeutic development decisions.
Collapse
Affiliation(s)
- Jennifer L. Wilson
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
| | - Rebecca Racz
- Division of Applied Regulatory Science, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Tianyun Liu
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
| | - Oluseyi Adeniyi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Jielin Sun
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Anuradha Ramamoorthy
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Michael Pacanowski
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Russ Altman
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
- Department of Genetics, Stanford University, Palo Alto California, United States of America
| |
Collapse
|
490
|
Kilicoglu H. Biomedical text mining for research rigor and integrity: tasks, challenges, directions. Brief Bioinform 2018; 19:1400-1414. [PMID: 28633401 PMCID: PMC6291799 DOI: 10.1093/bib/bbx057] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/10/2017] [Indexed: 01/01/2023] Open
Abstract
An estimated quarter of a trillion US dollars is invested in the biomedical research enterprise annually. There is growing alarm that a significant portion of this investment is wasted because of problems in reproducibility of research findings and in the rigor and integrity of research conduct and reporting. Recent years have seen a flurry of activities focusing on standardization and guideline development to enhance the reproducibility and rigor of biomedical research. Research activity is primarily communicated via textual artifacts, ranging from grant applications to journal publications. These artifacts can be both the source and the manifestation of practices leading to research waste. For example, an article may describe a poorly designed experiment, or the authors may reach conclusions not supported by the evidence presented. In this article, we pose the question of whether biomedical text mining techniques can assist the stakeholders in the biomedical research enterprise in doing their part toward enhancing research integrity and rigor. In particular, we identify four key areas in which text mining techniques can make a significant contribution: plagiarism/fraud detection, ensuring adherence to reporting guidelines, managing information overload and accurate citation/enhanced bibliometrics. We review the existing methods and tools for specific tasks, if they exist, or discuss relevant research that can provide guidance for future work. With the exponential increase in biomedical research output and the ability of text mining approaches to perform automatic tasks at large scale, we propose that such approaches can support tools that promote responsible research practices, providing significant benefits for the biomedical research enterprise.
Collapse
Affiliation(s)
- Halil Kilicoglu
- Lister Hill National Center for Biomedical Communications, US National Library of Medicine
| |
Collapse
|
491
|
Lee T, Yoon Y. Drug repositioning using drug-disease vectors based on an integrated network. BMC Bioinformatics 2018; 19:446. [PMID: 30463505 PMCID: PMC6249928 DOI: 10.1186/s12859-018-2490-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/12/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diverse interactions occur between biomolecules, such as activation, inhibition, expression, or repression. However, previous network-based studies of drug repositioning have employed interaction on the binary protein-protein interaction (PPI) network without considering the characteristics of the interactions. Recently, some studies of drug repositioning using gene expression data found that associations between drug and disease genes are useful information for identifying novel drugs to treat diseases. However, the gene expression profiles for drugs and diseases are not always available. Although gene expression profiles of drugs and diseases are available, existing methods cannot use the drugs or diseases, when differentially expressed genes in the profiles are not included in their network. RESULTS We developed a novel method for identifying candidate indications of existing drugs considering types of interactions between biomolecules based on known drug-disease associations. To obtain associations between drug and disease genes, we constructed a directed network using protein interaction and gene regulation data obtained from various public databases providing diverse biological pathways. The network includes three types of edges depending on relationships between biomolecules. To quantify the association between a target gene and a disease gene, we explored the shortest paths from the target gene to the disease gene and calculated the types and weights of the shortest paths. For each drug-disease pair, we built a vector consisting of values for each disease gene influenced by the drug. Using the vectors and known drug-disease associations, we constructed classifiers to identify novel drugs for each disease. CONCLUSION We propose a method for exploring candidate drugs of diseases using associations between drugs and disease genes derived from a directed gene network instead of gene regulation data obtained from gene expression profiles. Compared to existing methods that require information on gene relationships and gene expression data, our method can be applied to a greater number of drugs and diseases. Furthermore, to validate our predictions, we compared the predictions with drug-disease pairs in clinical trials using the hypergeometric test, which showed significant results. Our method also showed better performance compared to existing methods for the area under the receiver operating characteristic curve (AUC).
Collapse
Affiliation(s)
- Taekeon Lee
- Department of Computer Engineering, Gachon University, 5-22Ho, IT college, 1324 Seongnam-daero, Seongnam-si, 13120 South Korea
| | - Youngmi Yoon
- Department of Computer Engineering, Gachon University, 5-22Ho, IT college, 1324 Seongnam-daero, Seongnam-si, 13120 South Korea
| |
Collapse
|
492
|
Zhou WM, Wu GL, Huang J, Li JG, Hao C, He QM, Chen XD, Wang GX, Tu XH. Low expression of PDK1 inhibits renal cell carcinoma cell proliferation, migration, invasion and epithelial mesenchymal transition through inhibition of the PI3K-PDK1-Akt pathway. Cell Signal 2018; 56:1-14. [PMID: 30465826 DOI: 10.1016/j.cellsig.2018.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 11/18/2022]
Abstract
As the most commonly occurring form of primary renal tumor, renal cell carcinoma (RCC) is a malignancy accompanied by a high mortality rate. 3-phosphoinositide-dependent protein kinase 1 (PDK1) has been established as a protein target and generated considerable interest in both the pharmaceutical and academia industry. The aim of the current study was to investigate the effect of si-PDK1 on the RCC cell apoptosis, proliferation, migration, invasion and epithelial mesenchymal transition (EMT) in connection with the PI3K-PDK1-Akt pathway. Microarray analysis from the GEO database was adopted to identify differentially expressed genes (DEGs) related to RCC, after which the positive expression of the PDK1 protein in tissue was determined accordingly. The optimal silencing si-RNA was subsequently selected and RCC cell lines 786-O and A498 were selected and transfected with either a si-PDK1 or activator of the PI3K-PDK1-Akt pathway for grouping purposes. The mRNA and protein expressions of PDK1, the PI3K-PDK1-Akt pathway-, EMT- and apoptosis-related genes were then evaluated. The effect of si-PDK1 on cell proliferation, apoptosis, invasion and migration was then analyzed. Through microarray analysis of GSE6344, GSE53757, GSE14762 and GSE781, PDK1 was examined. PDK1 was determined to be highly expressed in RCC tissues. Si-PDK1 exhibited marked reductions in relation to the mRNA and protein expression of PDK1, PI3K, AKT as well as Vimentin while elevated mRNA and protein expressions of E-cadherin were detected, which ultimately suggested that cell migration, proliferation and invasion had been inhibited coupled with enhanced levels of cell apoptosis. While a notable observation was made highlighting that the PI3K-PDK1-Akt pathway antagonized the effect of PDK1 silencing. Taken together, the key observations of this study provide evidence suggesting that high expressions of PDK1 are found in RCC, while highlighting that silencing PDK1 could inhibit RCC cell proliferation, migration, invasion and EMT by repressing the PI3K-PDK1-Akt pathway.
Collapse
Affiliation(s)
- Wei-Min Zhou
- Jiangxi Medical College, Nanchang University, Nanchang 330006, PR China; Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Gao-Liang Wu
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Ji Huang
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Jin-Gao Li
- Department of Radiotherapy, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Chao Hao
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Qiu-Ming He
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Xiao-Dan Chen
- Department of Science and Education, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Gong-Xian Wang
- Jiangxi Medical College, Nanchang University, Nanchang 330006, PR China; Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| | - Xin-Hua Tu
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China.
| |
Collapse
|
493
|
Fernandes M, Patel A, Husi H. C/VDdb: A multi-omics expression profiling database for a knowledge-driven approach in cardiovascular disease (CVD). PLoS One 2018; 13:e0207371. [PMID: 30419069 PMCID: PMC6231654 DOI: 10.1371/journal.pone.0207371] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022] Open
Abstract
The cardiovascular disease (C/VD) database is an integrated and clustered information resource that covers multi-omic studies (microRNA, genomics, proteomics and metabolomics) of cardiovascular-related traits with special emphasis on coronary artery disease (CAD). This resource was built by mining existing literature and public databases and thereafter manual biocuration was performed. To enable integration of omic data from distinct platforms and species, a specific ontology was applied to tie together and harmonise multi-level omic studies based on gene and protein clusters (CluSO) and mapping of orthologous genes (OMAP) across species. CAD continues to be a leading cause of death in the population worldwide, and it is generally thought to be an age-related disease. However, CAD incidence rates are now known to be highly influenced by environmental factors and interactions, in addition to genetic determinants. With the complexity of CAD aetiology, there is a difficulty in research studies to elucidate general elements compared to other cardiovascular diseases. Data from 92 studies, covering 13945 molecular entries (4353 unique molecules) is described, including data descriptors for experimental setup, study design, discovery-validation sample size and associated fold-changes of the differentially expressed molecular features (p-value<0.05). A dedicated interactive web interface, equipped with a multi-parametric search engine, data export and indexing menus are provided for a user-accessible browsing experience. The main aim of this work was the development of a data repository linking clinical information and molecular differential expression in several CVD-related traits from multi-omics studies (genomics, transcriptomics, proteomics and metabolomics). As an example case of how to query and identify data sets within the database framework and concomitantly demonstrate the database utility, we queried CAD-associated studies and performed a systems-level integrative analysis. URL: www.padb.org/cvd
Collapse
Affiliation(s)
- Marco Fernandes
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Alisha Patel
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- Division of Biomedical Sciences, Centre for Health Science, University of the Highlands and Islands, Inverness, United Kingdom
- * E-mail:
| |
Collapse
|
494
|
López-Cortés A, Paz-Y-Miño C, Cabrera-Andrade A, Barigye SJ, Munteanu CR, González-Díaz H, Pazos A, Pérez-Castillo Y, Tejera E. Gene prioritization, communality analysis, networking and metabolic integrated pathway to better understand breast cancer pathogenesis. Sci Rep 2018; 8:16679. [PMID: 30420728 PMCID: PMC6232116 DOI: 10.1038/s41598-018-35149-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/16/2018] [Indexed: 12/30/2022] Open
Abstract
Consensus strategy was proved to be highly efficient in the recognition of gene-disease association. Therefore, the main objective of this study was to apply theoretical approaches to explore genes and communities directly involved in breast cancer (BC) pathogenesis. We evaluated the consensus between 8 prioritization strategies for the early recognition of pathogenic genes. A communality analysis in the protein-protein interaction (PPi) network of previously selected genes was enriched with gene ontology, metabolic pathways, as well as oncogenomics validation with the OncoPPi and DRIVE projects. The consensus genes were rationally filtered to 1842 genes. The communality analysis showed an enrichment of 14 communities specially connected with ERBB, PI3K-AKT, mTOR, FOXO, p53, HIF-1, VEGF, MAPK and prolactin signaling pathways. Genes with highest ranking were TP53, ESR1, BRCA2, BRCA1 and ERBB2. Genes with highest connectivity degree were TP53, AKT1, SRC, CREBBP and EP300. The connectivity degree allowed to establish a significant correlation between the OncoPPi network and our BC integrated network conformed by 51 genes and 62 PPi. In addition, CCND1, RAD51, CDC42, YAP1 and RPA1 were functional genes with significant sensitivity score in BC cell lines. In conclusion, the consensus strategy identifies both well-known pathogenic genes and prioritized genes that need to be further explored.
Collapse
Affiliation(s)
- Andrés López-Cortés
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, 170129, Quito, Ecuador.
- RNASA-IMEDIR, Computer Sciences Faculty, University of Coruna, 15071, Coruna, Spain.
| | - César Paz-Y-Miño
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, 170129, Quito, Ecuador
| | - Alejandro Cabrera-Andrade
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de las Américas, Avenue de los Granados, 170125, Quito, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de las Américas, Avenue de los Granados, 170125, Quito, Ecuador
| | - Stephen J Barigye
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC, H3A 0B8, Canada
| | - Cristian R Munteanu
- RNASA-IMEDIR, Computer Sciences Faculty, University of Coruna, 15071, Coruna, Spain
- INIBIC, Institute of Biomedical Research, CHUAC, UDC, 15006, Coruna, Spain
| | - Humberto González-Díaz
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940, Leioa, Biscay, Spain
- IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Biscay, Spain
| | - Alejandro Pazos
- RNASA-IMEDIR, Computer Sciences Faculty, University of Coruna, 15071, Coruna, Spain
- INIBIC, Institute of Biomedical Research, CHUAC, UDC, 15006, Coruna, Spain
| | - Yunierkis Pérez-Castillo
- Grupo de Bio-Quimioinformática, Universidad de las Américas, Avenue de los Granados, 170125, Quito, Ecuador
- Escuela de Ciencias Físicas y Matemáticas, Universidad de las Américas, Avenue de los Granados, 170125, Quito, Ecuador
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de las Américas, Avenue de los Granados, 170125, Quito, Ecuador.
- Facultad de Ingeniería y Ciencias Agropecuarias, Universidad de las Américas, Avenue de los Granados, 170125, Quito, Ecuador.
| |
Collapse
|
495
|
Smaili FZ, Gao X, Hoehndorf R. OPA2Vec: combining formal and informal content of biomedical ontologies to improve similarity-based prediction. Bioinformatics 2018; 35:2133-2140. [DOI: 10.1093/bioinformatics/bty933] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Fatima Zohra Smaili
- Computer, Electrical & Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Computer, Electrical & Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Robert Hoehndorf
- Computer, Electrical & Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
496
|
Yang K, Wang N, Liu G, Wang R, Yu J, Zhang R, Chen J, Zhou X. Heterogeneous network embedding for identifying symptom candidate genes. J Am Med Inform Assoc 2018; 25:1452-1459. [PMID: 30357378 PMCID: PMC7646926 DOI: 10.1093/jamia/ocy117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/24/2018] [Accepted: 08/11/2018] [Indexed: 11/12/2022] Open
Abstract
Objective Investigating the molecular mechanisms of symptoms is a vital task in precision medicine to refine disease taxonomy and improve the personalized management of chronic diseases. Although there are abundant experimental studies and computational efforts to obtain the candidate genes of diseases, the identification of symptom genes is rarely addressed. We curated a high-quality benchmark dataset of symptom-gene associations and proposed a heterogeneous network embedding for identifying symptom genes. Methods We proposed a heterogeneous network embedding representation algorithm, which constructed a heterogeneous symptom-related network that integrated symptom-related associations and applied an embedding representation algorithm to obtain the low-dimensional vector representation of nodes. By measuring the relevance between symptoms and genes via calculating the similarities of their vectors, the candidate genes of given symptoms can be obtained. Results A benchmark dataset of 18 270 symptom-gene associations between 505 symptoms and 4549 genes was curated. We compared our method to baseline algorithms (FSGER and PRINCE). The experimental results indicated our algorithm achieved a significant improvement over the state-of-the-art method, with precision and recall improved by 66.80% (0.844 vs 0.506) and 53.96% (0.311 vs 0.202), respectively, for TOP@3 and association precision improved by 37.71% (0.723 vs 0.525) over the PRINCE. Conclusions The experimental validation of the algorithms and the literature validation of typical symptoms indicated our method achieved excellent performance. Hence, we curated a prediction dataset of 17 479 symptom-candidate genes. The benchmark and prediction datasets have the potential to promote investigations of the molecular mechanisms of symptoms and provide candidate genes for validation in experimental settings.
Collapse
Affiliation(s)
- Kuo Yang
- School of Computer and Information Technology and Beijing Key Laboratory of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Ning Wang
- School of Computer and Information Technology and Beijing Key Laboratory of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Guangming Liu
- School of Computer and Information Technology and Beijing Key Laboratory of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Ruyu Wang
- School of Computer and Information Technology and Beijing Key Laboratory of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Jian Yu
- School of Computer and Information Technology and Beijing Key Laboratory of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Runshun Zhang
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing, China
| | - Xuezhong Zhou
- School of Computer and Information Technology and Beijing Key Laboratory of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
- Data Center of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
497
|
PWCDA: Path Weighted Method for Predicting circRNA-Disease Associations. Int J Mol Sci 2018; 19:ijms19113410. [PMID: 30384427 PMCID: PMC6274797 DOI: 10.3390/ijms19113410] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
CircRNAs have particular biological structure and have proven to play important roles in diseases. It is time-consuming and costly to identify circRNA-disease associations by biological experiments. Therefore, it is appealing to develop computational methods for predicting circRNA-disease associations. In this study, we propose a new computational path weighted method for predicting circRNA-disease associations. Firstly, we calculate the functional similarity scores of diseases based on disease-related gene annotations and the semantic similarity scores of circRNAs based on circRNA-related gene ontology, respectively. To address missing similarity scores of diseases and circRNAs, we calculate the Gaussian Interaction Profile (GIP) kernel similarity scores for diseases and circRNAs, respectively, based on the circRNA-disease associations downloaded from circR2Disease database (http://bioinfo.snnu.edu.cn/CircR2Disease/). Then, we integrate disease functional similarity scores and circRNA semantic similarity scores with their related GIP kernel similarity scores to construct a heterogeneous network made up of three sub-networks: disease similarity network, circRNA similarity network and circRNA-disease association network. Finally, we compute an association score for each circRNA-disease pair based on paths connecting them in the heterogeneous network to determine whether this circRNA-disease pair is associated. We adopt leave one out cross validation (LOOCV) and five-fold cross validations to evaluate the performance of our proposed method. In addition, three common diseases, Breast Cancer, Gastric Cancer and Colorectal Cancer, are used for case studies. Experimental results illustrate the reliability and usefulness of our computational method in terms of different validation measures, which indicates PWCDA can effectively predict potential circRNA-disease associations.
Collapse
|
498
|
Lin J, Wu YJ, Liang X, Ji M, Ying HM, Wang XY, Sun X, Shao CH, Zhan LX, Zhang Y. Network-based integration of mRNA and miRNA profiles reveals new target genes involved in pancreatic cancer. Mol Carcinog 2018; 58:206-218. [PMID: 30294829 DOI: 10.1002/mc.22920] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/31/2018] [Accepted: 10/03/2018] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer is regarded as the most fatal and aggressive malignancy cancer due to its low 5-year survival rate and poor prognosis. The approaches of early diagnosis and treatment are limited, which makes it urgent to identify the complex mechanism of pancreatic oncogenesis. In this study, we used RNA-seq to investigate the transcriptomic (mRNA and miRNA) profiles of pancreatic cancer in paired tumor and normal pancreatic samples from ten patients. More than 1000 differentially expressed genes were identified, nearly half of which were also found to be differentially expressed in the majority of examined patients. Functional enrichment analysis revealed that these genes were significantly enriched in multicellular organismal and metabolic process, secretion, mineral transport, and intercellular communication. In addition, only 24 differentially expressed miRNAs were found, all of which have been reported to be associated with pancreatic cancer. Furthermore, an integrated miRNA-mRNA interaction network was generated using multiple resources. Based on the calculation of disease correlation scores developed here, several genes present in the largest connected subnetwork, such as albumin, ATPase H+ /K+ exchanging alpha polypeptide and carcinoembryonic antigen-related cell adhesion molecule 1, were considered as novel genes that play important roles in the development of pancreatic cancer. Overall, our data provide new insights into further understanding of key molecular mechanisms underlying pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Jie Lin
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong Province, P. R. China.,Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Yan-Jun Wu
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Xing Liang
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, P. R. China
| | - Meng Ji
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, P. R. China
| | - Hui-Min Ying
- Department of Endocrinology, Hangzhou Xixi Hospital, Hangzhou, Zhejiang, P. R. China
| | - Xin-Yu Wang
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Xia Sun
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Cheng-Hao Shao
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, P. R. China
| | - Li-Xing Zhan
- Key Laboratory of Nutrition, Metabolism, and Food Safety, Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Yan Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong Province, P. R. China
| |
Collapse
|
499
|
Kurt Z, Barrere-Cain R, LaGuardia J, Mehrabian M, Pan C, Hui ST, Norheim F, Zhou Z, Hasin Y, Lusis AJ, Yang X. Tissue-specific pathways and networks underlying sexual dimorphism in non-alcoholic fatty liver disease. Biol Sex Differ 2018; 9:46. [PMID: 30343673 PMCID: PMC6196429 DOI: 10.1186/s13293-018-0205-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) encompasses benign steatosis and more severe conditions such as non-alcoholic steatohepatitis (NASH), cirrhosis, and liver cancer. This chronic liver disease has a poorly understood etiology and demonstrates sexual dimorphisms. We aim to examine the molecular mechanisms underlying sexual dimorphisms in NAFLD pathogenesis through a comprehensive multi-omics study. We integrated genomics (DNA variations), transcriptomics of liver and adipose tissue, and phenotypic data of NAFLD derived from female mice of ~ 100 strains included in the hybrid mouse diversity panel (HMDP) and compared the NAFLD molecular pathways and gene networks between sexes. RESULTS We identified both shared and sex-specific biological processes for NAFLD. Adaptive immunity, branched chain amino acid metabolism, oxidative phosphorylation, and cell cycle/apoptosis were shared between sexes. Among the sex-specific pathways were vitamins and cofactors metabolism and ion channel transport for females, and phospholipid, lysophospholipid, and phosphatidylinositol metabolism and insulin signaling for males. Additionally, numerous lipid and insulin-related pathways and inflammatory processes in the adipose and liver tissue appeared to show more prominent association with NAFLD in male HMDP. Using data-driven network modeling, we identified plausible sex-specific and tissue-specific regulatory genes as well as those that are shared between sexes. These key regulators orchestrate the NAFLD pathways in a sex- and tissue-specific manner. Gonadectomy experiments support that sex hormones may partially underlie the sexually dimorphic genes and pathways involved in NAFLD. CONCLUSIONS Our multi-omics integrative study reveals sex- and tissue-specific genes, processes, and networks underlying sexual dimorphism in NAFLD and may facilitate sex-specific precision medicine.
Collapse
Affiliation(s)
- Zeyneb Kurt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA USA
| | - Rio Barrere-Cain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA USA
| | - Jonnby LaGuardia
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA USA
| | - Margarete Mehrabian
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Simon T Hui
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Frode Norheim
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Zhiqiang Zhou
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Yehudit Hasin
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA USA
| |
Collapse
|
500
|
Yamada Y, Kato K, Oguri M, Horibe H, Fujimaki T, Yasukochi Y, Takeuchi I, Sakuma J. Identification of 12 novel loci that confer susceptibility to early-onset dyslipidemia. Int J Mol Med 2018; 43:57-82. [PMID: 30365130 PMCID: PMC6257857 DOI: 10.3892/ijmm.2018.3943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/26/2018] [Indexed: 01/14/2023] Open
Abstract
The circulating concentrations of triglycerides, high density lipoprotein (HDL)-cholesterol, and low density lipoprotein (LDL)-cholesterol have a substantial genetic component, and the heritability of early-onset dyslipidemia might be expected to be higher compared with late-onset forms. In the present study, exome-wide association studies (EWASs) were performed for early-onset hypertriglyceridemia, hypo-HDL-cholesterolemia, and hyper-LDL-cholesterolemia, with the aim to identify genetic variants that confer susceptibility to these conditions in the Japanese population. A total of 8,073 individuals aged ≤65 years were enrolled in the study. The EWASs for hypertriglyceridemia (2,664 cases and 5,294 controls), hypo-HDL-cholesterolemia (974 cases and 7,085 controls), and hyper-LDL-cholesterolemia (2,911 cases and 5,111 controls) were performed with Illumina Human Exome-12 v1.2 DNA Analysis BeadChip or Infinium Exome-24 v1.0 BeadChip arrays. The association of allele frequencies for 31,198, 31,133, or 31,175 single nucleotide polymorphisms (SNPs) to hypertriglyceridemia, hypo-HDL-cholesterolemia, or hyper-LDL-cholesterolemia, respectively, was examined with Fisher’s exact test. To compensate for multiple comparisons of genotypes with each of the three conditions, Bonferroni’s correction was applied for statistical significance of association. The results demonstrated that 25, 28 and 65 SNPs were significantly associated with hypertriglyceridemia, hypo-HDL-cholesterolemia and hyper-LDL-cholesterolemia, respectively. Multivariable logistic regression analysis with adjustment for age and sex revealed that all 25, 28 and 65 of these SNPs were significantly associated with hypertriglyceridemia, hypo-HDL-cholesterolemia and hyper-LDL-cholesterolemia, respectively. Following examination of the association of the identified SNPs to serum concentrations of triglycerides, HDL-cholesterol, or LDL-cholesterol, linkage disequilibrium of the SNPs, and results of previous genome-wide association studies, we newly identified chromosomal region 19p12 as a susceptibility locus for hypertriglyceridemia, eight loci (MOB3C-TMOD4, LPGAT1, EHD3, COL6A3, ZNF860-CACNA1D, COL6A5, DCLRE1C, ZNF77) for hypo-HDL-cholesterolemia, and three loci (KIAA0319-FAM65B, UBD, LOC105375015) for hyper-LDL-cholesterolemia. The present study thus identified 12 novel loci that may confer susceptibility to early-onset dyslipidemia. Determination of genotypes for the SNPs at these loci may prove informative for assessment of genetic risk for hypertriglyceridemia, hypo-HDL-cholesterolemia, or hyper-LDL-cholesterolemia in the Japanese population.
Collapse
Affiliation(s)
- Yoshiji Yamada
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Kimihiko Kato
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Mitsutoshi Oguri
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Hideki Horibe
- Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu 507‑8522, Japan
| | - Tetsuo Fujimaki
- Department of Cardiovascular Medicine, Northern Mie Medical Center Inabe General Hospital, Inabe, Mie 511‑0428, Japan
| | - Yoshiki Yasukochi
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Ichiro Takeuchi
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332‑0012, Japan
| | - Jun Sakuma
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332‑0012, Japan
| |
Collapse
|