1
|
Wang M, Li B, Liu Y, Zhang M, Huang C, Cai T, Jia Y, Huang X, Ke H, Liu S, Yang S. Shu-Xie decoction alleviates oxidative stress and colon injury in acute sleep-deprived mice by suppressing p62/KEAP1/NRF2/HO1/NQO1 signaling. Front Pharmacol 2023; 14:1107507. [PMID: 36814500 PMCID: PMC9939528 DOI: 10.3389/fphar.2023.1107507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
Introduction: Sleep disorders are common clinical psychosomatic disorders that can co-exist with a variety of conditions. In humans and animal models, sleep deprivation (SD) is closely related with gastrointestinal diseases. Shu-Xie Decoction (SX) is a traditional Chinese medicine (TCM) with anti-nociceptive, anti-inflammatory, and antidepressant properties. SX is effective in the clinic for treating patients with abnormal sleep and/or gastrointestinal disorders, but the underlying mechanisms are not known. This study investigated the mechanisms by which SX alleviates SD-induced colon injury in vivo. Methods: C57BL/6 mice were placed on an automated sleep deprivation system for 72 h to generate an acute sleep deprivation (ASD) model, and low-dose SX (SXL), high-dose SX (SXH), or S-zopiclone (S-z) as a positive control using the oral gavage were given during the whole ASD-induced period for one time each day. The colon length was measured and the colon morphology was visualized using hematoxylin and eosin (H&E) staining. ROS and the redox biomarkers include reduced glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) were detected. Quantitative real-time PCR (qRT-PCR), molecular docking, immunofluorescence and western blotting assays were performed to detect the antioxidant signaling pathways. Results: ASD significantly increased FBG levels, decreased colon length, moderately increased the infiltration of inflammatory cells in the colon mucosa, altered the colon mucosal structure, increased the levels of ROS, GSH, MDA, and SOD activity compared with the controls. These adverse effects were significantly alleviated by SX treatment. ASD induced nuclear translocation of NRF2 in the colon mucosal cells and increased the expression levels of p62, NQO1, and HO1 transcripts and proteins, but these effects were reversed by SX treatment. Conclusion: SX decoction ameliorated ASD-induced oxidative stress and colon injury by suppressing the p62/KEAP1/NRF2/HO1/NQO1 signaling pathway. In conclusion, combined clinical experience, SX may be a promising drug for sleep disorder combined with colitis.
Collapse
Affiliation(s)
- Mengyuan Wang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Bo Li
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,*Correspondence: Bo Li, ; Suhuan Liu, ; Shuyu Yang,
| | - Yijiang Liu
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Mengting Zhang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Caoxin Huang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Teng Cai
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yibing Jia
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaoqing Huang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Hongfei Ke
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Suhuan Liu
- Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,*Correspondence: Bo Li, ; Suhuan Liu, ; Shuyu Yang,
| | - Shuyu Yang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,*Correspondence: Bo Li, ; Suhuan Liu, ; Shuyu Yang,
| |
Collapse
|
2
|
Sun G, Dong D, Dong Z, Zhang Q, Fang H, Wang C, Zhang S, Wu S, Dong Y, Wan Y. Drug repositioning: A bibliometric analysis. Front Pharmacol 2022; 13:974849. [PMID: 36225586 PMCID: PMC9549161 DOI: 10.3389/fphar.2022.974849] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Drug repurposing has become an effective approach to drug discovery, as it offers a new way to explore drugs. Based on the Science Citation Index Expanded (SCI-E) and Social Sciences Citation Index (SSCI) databases of the Web of Science core collection, this study presents a bibliometric analysis of drug repurposing publications from 2010 to 2020. Data were cleaned, mined, and visualized using Derwent Data Analyzer (DDA) software. An overview of the history and development trend of the number of publications, major journals, major countries, major institutions, author keywords, major contributors, and major research fields is provided. There were 2,978 publications included in the study. The findings show that the United States leads in this area of research, followed by China, the United Kingdom, and India. The Chinese Academy of Science published the most research studies, and NIH ranked first on the h-index. The Icahn School of Medicine at Mt Sinai leads in the average number of citations per study. Sci Rep, Drug Discov. Today, and Brief. Bioinform. are the three most productive journals evaluated from three separate perspectives, and pharmacology and pharmacy are unquestionably the most commonly used subject categories. Cheng, FX; Mucke, HAM; and Butte, AJ are the top 20 most prolific and influential authors. Keyword analysis shows that in recent years, most research has focused on drug discovery/drug development, COVID-19/SARS-CoV-2/coronavirus, molecular docking, virtual screening, cancer, and other research areas. The hotspots have changed in recent years, with COVID-19/SARS-CoV-2/coronavirus being the most popular topic for current drug repurposing research.
Collapse
Affiliation(s)
- Guojun Sun
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Dashun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Zuojun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Qian Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Hui Fang
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| | - Chaojun Wang
- Hangzhou Aeronautical Sanatorium for Special Service of Chinese Air Force, Hangzhou, China
| | - Shaoya Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Shuaijun Wu
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yichen Dong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuehua Wan
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
3
|
Pavel A, Saarimäki LA, Möbus L, Federico A, Serra A, Greco D. The potential of a data centred approach & knowledge graph data representation in chemical safety and drug design. Comput Struct Biotechnol J 2022; 20:4837-4849. [PMID: 36147662 PMCID: PMC9464643 DOI: 10.1016/j.csbj.2022.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
Big Data pervades nearly all areas of life sciences, yet the analysis of large integrated data sets remains a major challenge. Moreover, the field of life sciences is highly fragmented and, consequently, so is its data, knowledge, and standards. This, in turn, makes integrated data analysis and knowledge gathering across sub-fields a demanding task. At the same time, the integration of various research angles and data types is crucial for modelling the complexity of organisms and biological processes in a holistic manner. This is especially valid in the context of drug development and chemical safety assessment where computational methods can provide solutions for the urgent need of fast, effective, and sustainable approaches. At the same time, such computational methods require the development of methodologies suitable for an integrated and data centred Big Data view. Here we discuss Knowledge Graphs (KG) as a solution to a data centred analysis approach for drug and chemical development and safety assessment. KGs are knowledge bases, data analysis engines, and knowledge discovery systems all in one, allowing them to be used from simple data retrieval, over meta-analysis to complex predictive and knowledge discovery systems. Therefore, KGs have immense potential to advance the data centred approach, the re-usability, and informativity of data. Furthermore, they can improve the power of analysis, and the complexity of modelled processes, all while providing knowledge in a natively human understandable network data model.
Collapse
Affiliation(s)
- Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Laura A Saarimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Aldahdooh J, Vähä-Koskela M, Tang J, Tanoli Z. Using BERT to identify drug-target interactions from whole PubMed. BMC Bioinformatics 2022; 23:245. [PMID: 35729494 PMCID: PMC9214985 DOI: 10.1186/s12859-022-04768-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Drug-target interactions (DTIs) are critical for drug repurposing and elucidation of drug mechanisms, and are manually curated by large databases, such as ChEMBL, BindingDB, DrugBank and DrugTargetCommons. However, the number of curated articles likely constitutes only a fraction of all the articles that contain experimentally determined DTIs. Finding such articles and extracting the experimental information is a challenging task, and there is a pressing need for systematic approaches to assist the curation of DTIs. To this end, we applied Bidirectional Encoder Representations from Transformers (BERT) to identify such articles. Because DTI data intimately depends on the type of assays used to generate it, we also aimed to incorporate functions to predict the assay format. RESULTS Our novel method identified 0.6 million articles (along with drug and protein information) which are not previously included in public DTI databases. Using 10-fold cross-validation, we obtained ~ 99% accuracy for identifying articles containing quantitative drug-target profiles. The F1 micro for the prediction of assay format is 88%, which leaves room for improvement in future studies. CONCLUSION The BERT model in this study is robust and the proposed pipeline can be used to identify previously overlooked articles containing quantitative DTIs. Overall, our method provides a significant advancement in machine-assisted DTI extraction and curation. We expect it to be a useful addition to drug mechanism discovery and repurposing.
Collapse
Affiliation(s)
- Jehad Aldahdooh
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Doctoral Programme in Computer Science, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Ziaurrehman Tanoli
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,BioICAWtech, Helsinki, Finland.
| |
Collapse
|
5
|
Mavridou D, Psatha K, Aivaliotis M. Proteomics and Drug Repurposing in CLL towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13143391. [PMID: 34298607 PMCID: PMC8303629 DOI: 10.3390/cancers13143391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Despite continued efforts, the current status of knowledge in CLL molecular pathobiology, diagnosis, prognosis and treatment remains elusive and imprecise. Proteomics approaches combined with advanced bioinformatics and drug repurposing promise to shed light on the complex proteome heterogeneity of CLL patients and mitigate, improve, or even eliminate the knowledge stagnation. In relation to this concept, this review presents a brief overview of all the available proteomics and drug repurposing studies in CLL and suggests the way such studies can be exploited to find effective therapeutic options combined with drug repurposing strategies to adopt and accost a more “precision medicine” spectrum. Abstract CLL is a hematological malignancy considered as the most frequent lymphoproliferative disease in the western world. It is characterized by high molecular heterogeneity and despite the available therapeutic options, there are many patient subgroups showing the insufficient effectiveness of disease treatment. The challenge is to investigate the individual molecular characteristics and heterogeneity of these patients. Proteomics analysis is a powerful approach that monitors the constant state of flux operators of genetic information and can unravel the proteome heterogeneity and rewiring into protein pathways in CLL patients. This review essences all the available proteomics studies in CLL and suggests the way these studies can be exploited to find effective therapeutic options combined with drug repurposing approaches. Drug repurposing utilizes all the existing knowledge of the safety and efficacy of FDA-approved or investigational drugs and anticipates drug alignment to crucial CLL therapeutic targets, leading to a better disease outcome. The drug repurposing studies in CLL are also discussed in this review. The next goal involves the integration of proteomics-based drug repurposing in precision medicine, as well as the application of this procedure into clinical practice to predict the most appropriate drugs combination that could ensure therapy and the long-term survival of each CLL patient.
Collapse
Affiliation(s)
- Dimitra Mavridou
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantina Psatha
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Greece
- Correspondence: (K.P.); (M.A.)
| | - Michalis Aivaliotis
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Greece
- Correspondence: (K.P.); (M.A.)
| |
Collapse
|
6
|
NOD: a web server to predict New use of Old Drugs to facilitate drug repurposing. Sci Rep 2021; 11:13540. [PMID: 34188160 PMCID: PMC8241987 DOI: 10.1038/s41598-021-92903-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/15/2021] [Indexed: 11/08/2022] Open
Abstract
Computational methods accelerate the drug repurposing pipelines that are a quicker and cost-effective alternative to discovering new molecules. However, there is a paucity of web servers to conduct fast, focussed, and customized investigations for identifying new uses of old drugs. We present the NOD web server, which has the mentioned characteristics. NOD uses a sensitive sequence-guided approach to identify close and distant homologs of a protein of interest. NOD then exploits this evolutionary information to suggest potential compounds from the DrugBank database that can be repurposed against the input protein. NOD also allows expansion of the chemical space of the potential candidates through similarity searches. We have validated the performance of NOD against available experimental and/or clinical reports. In 65.6% of the investigated cases in a control study, NOD is able to identify drugs more effectively than the searches made in DrugBank. NOD is freely-available at http://pauling.mbu.iisc.ac.in/NOD/NOD/ .
Collapse
|
7
|
Dotolo S, Marabotti A, Rachiglio AM, Esposito Abate R, Benedetto M, Ciardiello F, De Luca A, Normanno N, Facchiano A, Tagliaferri R. A multiple network-based bioinformatics pipeline for the study of molecular mechanisms in oncological diseases for personalized medicine. Brief Bioinform 2021; 22:6287337. [PMID: 34050359 PMCID: PMC8574709 DOI: 10.1093/bib/bbab180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 01/03/2023] Open
Abstract
Motivation Assessment of genetic mutations is an essential element in the modern era of personalized cancer treatment. Our strategy is focused on ‘multiple network analysis’ in which we try to improve cancer diagnostics by using biological networks. Genetic alterations in some important hubs or in driver genes such as BRAF and TP53 play a critical role in regulating many important molecular processes. Most of the studies are focused on the analysis of the effects of single mutations, while tumors often carry mutations of multiple driver genes. The aim of this work is to define an innovative bioinformatics pipeline focused on the design and analysis of networks (such as biomedical and molecular networks), in order to: (1) improve the disease diagnosis; (2) identify the patients that could better respond to a given drug treatment; and (3) predict what are the primary and secondary effects of gene mutations involved in human diseases. Results By using our pipeline based on a multiple network approach, it has been possible to demonstrate and validate what are the joint effects and changes of the molecular profile that occur in patients with metastatic colorectal carcinoma (mCRC) carrying mutations in multiple genes. In this way, we can identify the most suitable drugs for the therapy for the individual patient. This information is useful to improve precision medicine in cancer patients. As an application of our pipeline, the clinically significant case studies of a cohort of mCRC patients with the BRAF V600E-TP53 I195N missense combined mutation were considered. Availability The procedures used in this paper are part of the Cytoscape Core, available at (www.cytoscape.org). Data used here on mCRC patients have been published in [55]. Supplementary Information A supplementary file containing a more detailed discussion of this case study and other cases is available at the journal site as Supplementary Data.
Collapse
Affiliation(s)
- Serena Dotolo
- Dipartimento di Scienze Aziendali, Management & Innovation Systems, Università degli Studi di Salerno, Fisciano (SA), Italy
| | - Anna Marabotti
- Dipartimento di Chimica e Biologia "A. Zambelli", Università degli Studi di Salerno, Fisciano (SA), Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori -IRCCS - Fondazione G. Pascale, Naples, Italy
| | | | - Fortunato Ciardiello
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Angelo Facchiano
- Institute of Food Sciences, Italian National Research Council (CNR), Avellino, Italy
| | - Roberto Tagliaferri
- Dipartimento di Scienze Aziendali, Management & Innovation Systems, Università degli Studi di Salerno, Fisciano (SA), Italy
| |
Collapse
|
8
|
Barh D, Aljabali AA, Tambuwala MM, Tiwari S, Serrano-Aroca Á, Alzahrani KJ, Silva Andrade B, Azevedo V, Ganguly NK, Lundstrom K. Predicting COVID-19-Comorbidity Pathway Crosstalk-Based Targets and Drugs: Towards Personalized COVID-19 Management. Biomedicines 2021; 9:556. [PMID: 34067609 PMCID: PMC8156524 DOI: 10.3390/biomedicines9050556] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/12/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
It is well established that pre-existing comorbid conditions such as hypertension, diabetes, obesity, cardiovascular diseases (CVDs), chronic kidney diseases (CKDs), cancers, and chronic obstructive pulmonary disease (COPD) are associated with increased severity and fatality of COVID-19. The increased death from COVID-19 is due to the unavailability of a gold standard therapeutic and, more importantly, the lack of understanding of how the comorbid conditions and COVID-19 interact at the molecular level, so that personalized management strategies can be adopted. Here, using multi-omics data sets and bioinformatics strategy, we identified the pathway crosstalk between COVID-19 and diabetes, hypertension, CVDs, CKDs, and cancers. Further, shared pathways and hub gene-based targets for COVID-19 and its associated specific and combination of comorbid conditions are also predicted towards developing personalized management strategies. The approved drugs for most of these identified targets are also provided towards drug repurposing. Literature supports the involvement of our identified shared pathways in pathogenesis of COVID-19 and development of the specific comorbid condition of interest. Similarly, shared pathways- and hub gene-based targets are also found to have potential implementations in managing COVID-19 patients. However, the identified targets and drugs need further careful evaluation for their repurposing towards personalized treatment of COVID-19 cases having pre-existing specific comorbid conditions we have considered in this analysis. The method applied here may also be helpful in identifying common pathway components and targets in other disease-disease interactions too.
Collapse
Affiliation(s)
- Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.T.); (V.A.)
| | - Alaa A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK;
| | - Sandeep Tiwari
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.T.); (V.A.)
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia;
| | - Bruno Silva Andrade
- Laboratório de Bioinformática e Química Computacional, Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia (UESB), Jequié 45206-190, Brazil;
| | - Vasco Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.T.); (V.A.)
| | - Nirmal Kumar Ganguly
- National Institute of Immunology, Aruna Asaf Ali Marg, Jawaharlal Nehru University, New Delhi 110067, India;
- Institute of Liver and Biliary Science, New Delhi 110070, India
- Policy Center for Biomedical Research, Translational Health Science & Technology Institute, Faridabad 121001, India
| | | |
Collapse
|
9
|
Tanoli Z, Seemab U, Scherer A, Wennerberg K, Tang J, Vähä-Koskela M. Exploration of databases and methods supporting drug repurposing: a comprehensive survey. Brief Bioinform 2021; 22:1656-1678. [PMID: 32055842 PMCID: PMC7986597 DOI: 10.1093/bib/bbaa003] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/09/2019] [Indexed: 02/07/2023] Open
Abstract
Drug development involves a deep understanding of the mechanisms of action and possible side effects of each drug, and sometimes results in the identification of new and unexpected uses for drugs, termed as drug repurposing. Both in case of serendipitous observations and systematic mechanistic explorations, confirmation of new indications for a drug requires hypothesis building around relevant drug-related data, such as molecular targets involved, and patient and cellular responses. These datasets are available in public repositories, but apart from sifting through the sheer amount of data imposing computational bottleneck, a major challenge is the difficulty in selecting which databases to use from an increasingly large number of available databases. The database selection is made harder by the lack of an overview of the types of data offered in each database. In order to alleviate these problems and to guide the end user through the drug repurposing efforts, we provide here a survey of 102 of the most promising and drug-relevant databases reported to date. We summarize the target coverage and types of data available in each database and provide several examples of how multi-database exploration can facilitate drug repurposing.
Collapse
Affiliation(s)
- Ziaurrehman Tanoli
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - Umair Seemab
- Haartman Institute, University of Helsinki, Finland
| | - Andreas Scherer
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - Krister Wennerberg
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Denmark
| | - Jing Tang
- Faculty of medicine, University of Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| |
Collapse
|
10
|
Tanoli Z, Vähä-Koskela M, Aittokallio T. Artificial intelligence, machine learning, and drug repurposing in cancer. Expert Opin Drug Discov 2021; 16:977-989. [PMID: 33543671 DOI: 10.1080/17460441.2021.1883585] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Drug repurposing provides a cost-effective strategy to re-use approved drugs for new medical indications. Several machine learning (ML) and artificial intelligence (AI) approaches have been developed for systematic identification of drug repurposing leads based on big data resources, hence further accelerating and de-risking the drug development process by computational means.Areas covered: The authors focus on supervised ML and AI methods that make use of publicly available databases and information resources. While most of the example applications are in the field of anticancer drug therapies, the methods and resources reviewed are widely applicable also to other indications including COVID-19 treatment. A particular emphasis is placed on the use of comprehensive target activity profiles that enable a systematic repurposing process by extending the target profile of drugs to include potent off-targets with therapeutic potential for a new indication.Expert opinion: The scarcity of clinical patient data and the current focus on genetic aberrations as primary drug targets may limit the performance of anticancer drug repurposing approaches that rely solely on genomics-based information. Functional testing of cancer patient cells exposed to a large number of targeted therapies and their combinations provides an additional source of repurposing information for tissue-aware AI approaches.
Collapse
Affiliation(s)
- Ziaurrehman Tanoli
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLife, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLife, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLife, University of Helsinki, Helsinki, Finland.,Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.,Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Tuerkova A, Zdrazil B. A ligand-based computational drug repurposing pipeline using KNIME and Programmatic Data Access: case studies for rare diseases and COVID-19. J Cheminform 2020; 12:71. [PMID: 33250934 PMCID: PMC7686838 DOI: 10.1186/s13321-020-00474-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/09/2020] [Indexed: 01/01/2023] Open
Abstract
Biomedical information mining is increasingly recognized as a promising technique to accelerate drug discovery and development. Especially, integrative approaches which mine data from several (open) data sources have become more attractive with the increasing possibilities to programmatically access data through Application Programming Interfaces (APIs). The use of open data in conjunction with free, platform-independent analytic tools provides the additional advantage of flexibility, re-usability, and transparency. Here, we present a strategy for performing ligand-based in silico drug repurposing with the analytics platform KNIME. We demonstrate the usefulness of the developed workflow on the basis of two different use cases: a rare disease (here: Glucose Transporter Type 1 (GLUT-1) deficiency), and a new disease (here: COVID 19). The workflow includes a targeted download of data through web services, data curation, detection of enriched structural patterns, as well as substructure searches in DrugBank and a recently deposited data set of antiviral drugs provided by Chemical Abstracts Service. Developed workflows, tutorials with detailed step-by-step instructions, and the information gained by the analysis of data for GLUT-1 deficiency syndrome and COVID-19 are made freely available to the scientific community. The provided framework can be reused by researchers for other in silico drug repurposing projects, and it should serve as a valuable teaching resource for conveying integrative data mining strategies.
Collapse
Affiliation(s)
- Alzbeta Tuerkova
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Barbara Zdrazil
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| |
Collapse
|
12
|
Uncovering New Drug Properties in Target-Based Drug-Drug Similarity Networks. Pharmaceutics 2020; 12:pharmaceutics12090879. [PMID: 32947845 PMCID: PMC7557376 DOI: 10.3390/pharmaceutics12090879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 01/19/2023] Open
Abstract
Despite recent advances in bioinformatics, systems biology, and machine learning, the accurate prediction of drug properties remains an open problem. Indeed, because the biological environment is a complex system, the traditional approach—based on knowledge about the chemical structures—can not fully explain the nature of interactions between drugs and biological targets. Consequently, in this paper, we propose an unsupervised machine learning approach that uses the information we know about drug–target interactions to infer drug properties. To this end, we define drug similarity based on drug–target interactions and build a weighted Drug–Drug Similarity Network according to the drug–drug similarity relationships. Using an energy-model network layout, we generate drug communities associated with specific, dominant drug properties. DrugBank confirms the properties of 59.52% of the drugs in these communities, and 26.98% are existing drug repositioning hints we reconstruct with our DDSN approach. The remaining 13.49% of the drugs seem not to match the dominant pharmacologic property; thus, we consider them potential drug repurposing hints. The resources required to test all these repurposing hints are considerable. Therefore we introduce a mechanism of prioritization based on the betweenness/degree node centrality. Using betweenness/degree as an indicator of drug repurposing potential, we select Azelaic acid and Meprobamate as a possible antineoplastic and antifungal, respectively. Finally, we use a test procedure based on molecular docking to analyze Azelaic acid and Meprobamate’s repurposing.
Collapse
|
13
|
Abstract
Aim: The druggability of epigenetic targets has prompted researchers to develop small-molecule therapeutics. However, no systematic assessment has ever been done to investigate the chemical space of epigenetic modulators. Herein, we report a comprehensive chemoinformatic analysis of epigenetic ligands from EpiDBase, HEMD, ChEMBL and PubChem databases. Results: Nearly, 0.45 × 106 ligands were analyzed for assay interference compounds, target profiling, drug-like properties and hit prioritization. After eliminating approximately 96,000 problematic compounds, the remaining 0.36 × 106 compounds were studied for their physicochemical distributions, principal component analysis and hit prioritization. More than 30% of assay interference compounds were determined for many proteins. Conclusion: This systematic assessment of epigenetic ligands will help in the enrichment of screening libraries with high-quality compounds and thus, the generation of efficacious drug candidates.
Collapse
|
14
|
Giri AK, Ianevski A, Aittokallio T. Genome-wide off-targets of drugs: risks and opportunities. Cell Biol Toxicol 2019; 35:485-487. [PMID: 31432301 DOI: 10.1007/s10565-019-09491-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/11/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290, Helsinki, Finland
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290, Helsinki, Finland.,Helsinki Institute for Information Technology (HIIT), Aalto University, FI-02150, Espoo, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290, Helsinki, Finland. .,Helsinki Institute for Information Technology (HIIT), Aalto University, FI-02150, Espoo, Finland. .,Department of Mathematics and Statistics, University of Turku, FI-20014, Turku, Finland.
| |
Collapse
|
15
|
Sabetian S, Shamsir MS. Computer aided analysis of disease linked protein networks. Bioinformation 2019; 15:513-522. [PMID: 31485137 PMCID: PMC6704336 DOI: 10.6026/97320630015513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/26/2022] Open
Abstract
Proteins can interact in various ways, ranging from direct physical relationships to indirect interactions in a formation of protein-protein interaction network. Diagnosis of the protein connections is critical to identify various cellular pathways. Today constructing and analyzing the protein interaction network is being developed as a powerful approach to create network pharmacology toward detecting unknown genes and proteins associated with diseases. Discovery drug targets regarding therapeutic decisions are exciting outcomes of studying disease networks. Protein connections may be identified by experimental and recent new computational approaches. Due to difficulties in analyzing in-vivo proteins interactions, many researchers have encouraged improving computational methods to design protein interaction network. In this review, the experimental and computational approaches and also advantages and disadvantages of these methods regarding the identification of new interactions in a molecular mechanism have been reviewed. Systematic analysis of complex biological systems including network pharmacology and disease network has also been discussed in this review.
Collapse
Affiliation(s)
- Soudabeh Sabetian
- Department of Biological and Health Sciences, Faculty of Bioscience and Medical Engineering, Universiti Teknologi Malaysia, 81310 Johor, Malaysia
- Infertility Research Center, Shiraz University, Shiraz 71454, Iran, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohd Shahir Shamsir
- Department of Biological and Health Sciences, Faculty of Bioscience and Medical Engineering, Universiti Teknologi Malaysia, 81310 Johor, Malaysia
| |
Collapse
|
16
|
Blucher AS, McWeeney SK, Stein L, Wu G. Visualization of drug target interactions in the contexts of pathways and networks with ReactomeFIViz. F1000Res 2019; 8:908. [PMID: 31372215 PMCID: PMC6644836 DOI: 10.12688/f1000research.19592.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
The precision medicine paradigm is centered on therapies targeted to particular molecular entities that will elicit an anticipated and controlled therapeutic response. However, genetic alterations in the drug targets themselves or in genes whose products interact with the targets can affect how well a drug actually works for an individual patient. To better understand the effects of targeted therapies in patients, we need software tools capable of simultaneously visualizing patient-specific variations and drug targets in their biological context. This context can be provided using pathways, which are process-oriented representations of biological reactions, or biological networks, which represent pathway-spanning interactions among genes, proteins, and other biological entities. To address this need, we have recently enhanced the Reactome Cytoscape app, ReactomeFIViz, to assist researchers in visualizing and modeling drug and target interactions. ReactomeFIViz integrates drug-target interaction information with high quality manually curated pathways and a genome-wide human functional interaction network. Both the pathways and the functional interaction network are provided by Reactome, the most comprehensive open source biological pathway knowledgebase. We describe several examples demonstrating the application of these new features to the visualization of drugs in the contexts of pathways and networks. Complementing previous features in ReactomeFIViz, these new features enable researchers to ask focused questions about targeted therapies, such as drug sensitivity for patients with different mutation profiles, using a pathway or network perspective.
Collapse
Affiliation(s)
- Aurora S Blucher
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.,Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Lincoln Stein
- Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
17
|
Zhou L, Li Z, Yang J, Tian G, Liu F, Wen H, Peng L, Chen M, Xiang J, Peng L. Revealing Drug-Target Interactions with Computational Models and Algorithms. Molecules 2019; 24:molecules24091714. [PMID: 31052598 PMCID: PMC6540161 DOI: 10.3390/molecules24091714] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/02/2022] Open
Abstract
Background: Identifying possible drug-target interactions (DTIs) has become an important task in drug research and development. Although high-throughput screening is becoming available, experimental methods narrow down the validation space because of extremely high cost, low success rate, and time consumption. Therefore, various computational models have been exploited to infer DTI candidates. Methods: We introduced relevant databases and packages, mainly provided a comprehensive review of computational models for DTI identification, including network-based algorithms and machine learning-based methods. Specially, machine learning-based methods mainly include bipartite local model, matrix factorization, regularized least squares, and deep learning. Results: Although computational methods have obtained significant improvement in the process of DTI prediction, these models have their limitations. We discussed potential avenues for boosting DTI prediction accuracy as well as further directions.
Collapse
Affiliation(s)
- Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| | - Zejun Li
- School of Computer Science, Hunan Institute of Technology, Henyang 421002, Hunan, China.
| | | | - Geng Tian
- Geneis (Beijing) Co. Ltd., Beijing 100102, China.
| | - Fuxing Liu
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| | - Hong Wen
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| | - Li Peng
- School of Computer Science, University of Science and Technology of Hunan, Xiangtan 411201, Hunan, China.
| | - Min Chen
- School of Computer Science, Hunan Institute of Technology, Henyang 421002, Hunan, China.
| | - Ju Xiang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China.
- Neuroscience Research Center, Department of Basic Medical Sciences, Changsha Medical University, Changsha 410219, Hunan, China.
| | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| |
Collapse
|