1
|
Koul M, Kaushik S, Singh K, Sharma D. VITALdb: to select the best viroinformatics tools for a desired virus or application. Brief Bioinform 2025; 26:bbaf084. [PMID: 40063348 PMCID: PMC11892104 DOI: 10.1093/bib/bbaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/14/2025] [Accepted: 02/17/2025] [Indexed: 05/13/2025] Open
Abstract
The recent pandemics of viral diseases, COVID-19/mpox (humans) and lumpy skin disease (cattle), have kept us glued to viral research. These pandemics along with the recent human metapneumovirus outbreak have exposed the urgency for early diagnosis of viral infections, vaccine development, and discovery of novel antiviral drugs and therapeutics. To support this, there is an armamentarium of virus-specific computational tools that are currently available. VITALdb (VIroinformatics Tools and ALgorithms database) is a resource of ~360 viroinformatics tools encompassing all major viruses (SARS-CoV-2, influenza virus, human immunodeficiency virus, papillomavirus, herpes simplex virus, hepatitis virus, dengue virus, Ebola virus, Zika virus, etc.) and several diverse applications [structural and functional annotation, antiviral peptides development, subspecies characterization, recognition of viral recombination, inhibitors identification, phylogenetic analysis, virus-host prediction, viral metagenomics, detection of mutation(s), primer designing, etc.]. Resources, tools, and other utilities mentioned in this article will not only facilitate further developments in the realm of viroinformatics but also provide tremendous fillip to translate fundamental knowledge into applied research. Most importantly, VITALdb is an inevitable tool for selecting the best tool(s) to carry out a desired task and hence will prove to be a vital database (VITALdb) for the scientific community. Database URL: https://compbio.iitr.ac.in/vitaldb.
Collapse
Affiliation(s)
- Mira Koul
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Shalini Kaushik
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Kavya Singh
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Deepak Sharma
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
2
|
Yin H, Duo H, Li S, Qin D, Xie L, Xiao Y, Sun J, Tao J, Zhang X, Li Y, Zou Y, Yang Q, Yang X, Hao Y, Li B. Unlocking biological insights from differentially expressed genes: Concepts, methods, and future perspectives. J Adv Res 2024:S2090-1232(24)00560-5. [PMID: 39647635 DOI: 10.1016/j.jare.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Identifying differentially expressed genes (DEGs) is a core task of transcriptome analysis, as DEGs can reveal the molecular mechanisms underlying biological processes. However, interpreting the biological significance of large DEG lists is challenging. Currently, gene ontology, pathway enrichment and protein-protein interaction analysis are common strategies employed by biologists. Additionally, emerging analytical strategies/approaches (such as network module analysis, knowledge graph, drug repurposing, cell marker discovery, trajectory analysis, and cell communication analysis) have been proposed. Despite these advances, comprehensive guidelines for systematically and thoroughly mining the biological information within DEGs remain lacking. AIM OF REVIEW This review aims to provide an overview of essential concepts and methodologies for the biological interpretation of DEGs, enhancing the contextual understanding. It also addresses the current limitations and future perspectives of these approaches, highlighting their broad applications in deciphering the molecular mechanism of complex diseases and phenotypes. To assist users in extracting insights from extensive datasets, especially various DEG lists, we developed DEGMiner (https://www.ciblab.net/DEGMiner/), which integrates over 300 easily accessible databases and tools. KEY SCIENTIFIC CONCEPTS OF REVIEW This review offers strong support and guidance for exploring DEGs, and also will accelerate the discovery of hidden biological insights within genomes.
Collapse
Affiliation(s)
- Huachun Yin
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China; Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China; Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, The Army Medical University, Chongqing 400038, PR China
| | - Hongrui Duo
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China
| | - Dan Qin
- Department of Biology, College of Science, Northeastern University, Boston, MA 02115, USA
| | - Lingling Xie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yingxue Xiao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jing Sun
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Xiaoxi Zhang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yinghong Li
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, PR China
| | - Yue Zou
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xian Yang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| |
Collapse
|
3
|
Kersting J, Lazareva O, Louadi Z, Baumbach J, Blumenthal DB, List M. DysRegNet: Patient-specific and confounder-aware dysregulated network inference towards precision therapeutics. Br J Pharmacol 2024. [PMID: 39631757 DOI: 10.1111/bph.17395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/09/2024] [Accepted: 10/05/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND AND PURPOSE Gene regulation is frequently altered in diseases in unique and patient-specific ways. Hence, personalised strategies have been proposed to infer patient-specific gene-regulatory networks. However, existing methods do not scale well because they often require recomputing the entire network per sample. Moreover, they do not account for clinically important confounding factors such as age, sex or treatment history. Finally, a user-friendly implementation for the analysis and interpretation of such networks is missing. EXPERIMENTAL APPROACH We present DysRegNet, a method for inferring patient-specific regulatory alterations (dysregulations) from bulk gene expression profiles. We compared DysRegNet to the well-known SSN method, considering patient clustering, promoter methylation, mutations and cancer-stage data. KEY RESULTS We demonstrate that both SSN and DysRegNet produce interpretable and biologically meaningful networks across various cancer types. In contrast to SSN, DysRegNet can scale to arbitrary sample numbers and highlights the importance of confounders in network inference, revealing an age-specific bias in gene regulation in breast cancer. DysRegNet is available as a Python package (https://github.com/biomedbigdata/DysRegNet_package), and analysis results for 11 TCGA cancer types are available through an interactive web interface (https://exbio.wzw.tum.de/dysregnet). CONCLUSION AND IMPLICATIONS DysRegNet introduces a novel bioinformatics tool enabling confounder-aware and patient-specific network analysis to unravel regulatory alteration in complex diseases.
Collapse
Affiliation(s)
- Johannes Kersting
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Olga Lazareva
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Junior Clinical Cooperation Unit Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zakaria Louadi
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - David B Blumenthal
- Department Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus List
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Munich Data Science Institute, Technical University of Munich, Garching, Germany
| |
Collapse
|
4
|
Imani S, Aminnezhad S, Alikarami M, Abedi Z, Mosleh IS, Maghsoudloo M, Taheri Z. Exploration of drug repurposing for Mpox outbreaks targeting gene signatures and host-pathogen interactions. Sci Rep 2024; 14:29436. [PMID: 39604570 PMCID: PMC11603026 DOI: 10.1038/s41598-024-79897-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Monkeypox (Mpox) is a growing public health concern, with complex interactions within host systems contributing to its impact. This study employs multi-omics approaches to uncover therapeutic targets and potential drug repurposing opportunities to better understand Mpox's molecular pathogenesis. We developed an in silico host-pathogen interaction (HPI) network and applied weighted gene co-expression network analysis (WGCNA) to explore interactions between Mpox and host proteins. Subtype-specific host-pathogen protein-protein interaction networks were constructed, and key modules from the HPI and WGCNA were integrated to identify significant host proteins. To predict upstream signaling pathways and transcription factors, we used eXpression2Kinases and ChIP-X Enrichment Analysis. The multi-Steiner trees method was applied to compare our findings with those from FDA-approved antiviral drugs. Analysis of 55 differentially expressed genes in Mpox infection revealed 11 kinases and 15 transcription factors as key regulators. We identified 16 potential drug targets, categorized into 8 proviral genes (ESR2, ERK1, ERK2, P38, JNK1, CDK4, GSK3B, STAT3) designated for inhibition, and 8 antiviral genes (IKKA, HDAC1, HIPK2, TF65, CSK21, HIPK2, ESR2, GSK3B) designated for activation. Proviral genes are involved in the AKT, Wnt, and STAT3 pathways, while antiviral genes impact the AP-1, NF-κB, apoptosis, and IFN pathways. Promising FDA-approved candidates were identified, including kinase inhibitors, steroid hormone receptor agonists, STAT3 inhibitors, and notably Niclosamide. This study enhances our understanding of Mpox by identifying key therapeutic targets and potential repurposable drugs, providing a valuable framework for developing new treatments.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Sargol Aminnezhad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Moslem Alikarami
- Research and Development Center, Dina Pharmed Exir Salamat Co, Tehran, Iran
| | - Zahra Abedi
- School of Biotechnology College of Science, University of Tehran, Tehran, Iran
| | - Iman Samei Mosleh
- Plant Functional Genomics Lab, Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Vienna, Austria
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zahra Taheri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
5
|
Yang L, Chen R, Goodison S, Sun Y. A comprehensive benchmark study of methods for identifying significantly perturbed subnetworks in cancer. Brief Bioinform 2024; 26:bbae692. [PMID: 39737568 DOI: 10.1093/bib/bbae692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 12/02/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025] Open
Abstract
Network-based methods utilize protein-protein interaction information to identify significantly perturbed subnetworks in cancer and to propose key molecular pathways. Numerous methods have been developed, but to date, a rigorous benchmark analysis to compare the performance of existing approaches is lacking. In this paper, we proposed a novel benchmarking framework using synthetic data and conducted a comprehensive analysis to investigate the ability of existing methods to detect target genes and subnetworks and to control false positives, and how they perform in the presence of topological biases at both gene and subnetwork levels. Our analysis revealed insights into algorithmic performance that were previously unattainable. Based on the results of the benchmark study, we presented a practical guide for users on how to select appropriate detection methods and protein-protein interaction networks for cancer pathway identification, and provided suggestions for future algorithm development.
Collapse
Affiliation(s)
- Le Yang
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, New York, NY 14203, United States
| | - Runpu Chen
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, New York, NY 14203, United States
| | - Steve Goodison
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, United States
| | - Yijun Sun
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, New York, NY 14203, United States
- Department of Computer Science and Engineering, University at Buffalo, The State University of New York, 12 Capen Hall, Buffalo, New York, NY 14260, United States
| |
Collapse
|
6
|
Duarte T, Omage FB, Rieder GS, Rocha JBT, Dalla Corte CL. Investigating SARS-CoV-2 virus-host interactions and mRNA expression: Insights using three models of D. melanogaster. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167324. [PMID: 38925484 DOI: 10.1016/j.bbadis.2024.167324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/22/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Responsible for COVID-19, SARS-CoV-2 is a coronavirus in which contagious variants continue to appear. Therefore, some population groups have demonstrated greater susceptibility to contagion and disease progression. For these reasons, several researchers have been studying the SARS-CoV-2/human interactome to understand the pathophysiology of COVID-19 and develop new pharmacological strategies. D. melanogaster is a versatile animal model with approximately 90 % human protein orthology related to SARS-CoV-2/human interactome and is widely used in metabolic studies. In this context, our work assessed the potential interaction between human proteins (ZNF10, NUP88, BCL2L1, UBC9, and RBX1) and their orthologous proteins in D. melanogaster (gl, Nup88, Buffy, ubc9, and Rbx1a) with proteins from SARS-CoV-2 (nsp3, nsp9, E, ORF7a, N, and ORF10) using computational approaches. Our results demonstrated that all the proteins have the potential to interact, and we compared the binding sites between humans and fruit flies. The stability and consistency in the structure of the gl_nsp3 complex, specifically, could be crucial for its specific biological functions. Lastly, to enhance the understanding of the influence of host factors on coronavirus infection, we also analyse the mRNA expression of the five genes (mbo, gl, lwr, Buffy, and Roc1a) responsible for encoding the fruit fly proteins. Briefly, we demonstrated that those genes were differentially regulated according to diets, sex, and age. Two groups showed higher positive gene regulation than others: females in the HSD group and males in the aging group, which could imply a higher virus-host susceptibility. Overall, while preliminary, our work contributes to the understanding of host defense mechanisms and potentially identifies candidate proteins and genes for in vivo viral studies against SARS-CoV-2.
Collapse
Affiliation(s)
- Tâmie Duarte
- Laboratory of Experimental Biochemistry and Toxicology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Folorunsho Bright Omage
- Biological Chemistry Laboratory, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil; Computational Biology Research Group, Embrapa Agricultural Informatics, Campinas, SP, Brazil
| | - Guilherme Schmitt Rieder
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - João B T Rocha
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Cristiane Lenz Dalla Corte
- Laboratory of Experimental Biochemistry and Toxicology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
7
|
Majidifar S, Zabihian A, Hooshmand M. Combination therapy synergism prediction for virus treatment using machine learning models. PLoS One 2024; 19:e0309733. [PMID: 39231124 PMCID: PMC11373828 DOI: 10.1371/journal.pone.0309733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
Combining different drugs synergistically is an essential aspect of developing effective treatments. Although there is a plethora of research on computational prediction for new combination therapies, there is limited to no research on combination therapies in the treatment of viral diseases. This paper proposes AI-based models for predicting novel antiviral combinations to treat virus diseases synergistically. To do this, we assembled a comprehensive dataset comprising information on viral strains, drug compounds, and their known interactions. As far as we know, this is the first dataset and learning model on combination therapy for viruses. Our proposal includes using a random forest model, an SVM model, and a deep model to train viral combination therapy. The machine learning models showed the highest performance, and the predicted values were validated by a t-test, indicating the effectiveness of the proposed methods. One of the predicted combinations of acyclovir and ribavirin has been experimentally confirmed to have a synergistic antiviral effect against herpes simplex type-1 virus, as described in the literature.
Collapse
Affiliation(s)
- Shayan Majidifar
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Arash Zabihian
- Department of QA, Kimia Zist Parsian Pharmaceutical Company, Zanjan, Iran
| | - Mohsen Hooshmand
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| |
Collapse
|
8
|
Qian J, Yang B, Wang S, Yuan S, Zhu W, Zhou Z, Zhang Y, Hu G. Drug Repurposing for COVID-19 by Constructing a Comorbidity Network with Central Nervous System Disorders. Int J Mol Sci 2024; 25:8917. [PMID: 39201608 PMCID: PMC11354300 DOI: 10.3390/ijms25168917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
In the post-COVID-19 era, treatment options for potential SARS-CoV-2 outbreaks remain limited. An increased incidence of central nervous system (CNS) disorders has been observed in long-term COVID-19 patients. Understanding the shared molecular mechanisms between these conditions may provide new insights for developing effective therapies. This study developed an integrative drug-repurposing framework for COVID-19, leveraging comorbidity data with CNS disorders, network-based modular analysis, and dynamic perturbation analysis to identify potential drug targets and candidates against SARS-CoV-2. We constructed a comorbidity network based on the literature and data collection, including COVID-19-related proteins and genes associated with Alzheimer's disease, Parkinson's disease, multiple sclerosis, and autism spectrum disorder. Functional module detection and annotation identified a module primarily involved in protein synthesis as a key target module, utilizing connectivity map drug perturbation data. Through the construction of a weighted drug-target network and dynamic network-based drug-repurposing analysis, ubiquitin-carboxy-terminal hydrolase L1 emerged as a potential drug target. Molecular dynamics simulations suggested pregnenolone and BRD-K87426499 as two drug candidates for COVID-19. This study introduces a dynamic-perturbation-network-based drug-repurposing approach to identify COVID-19 drug targets and candidates by incorporating the comorbidity conditions of CNS disorders.
Collapse
Affiliation(s)
- Jing Qian
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
| | - Bin Yang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
| | - Shuo Wang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
| | - Su Yuan
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
| | - Wenjing Zhu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
| | - Ziyun Zhou
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
| | - Yujuan Zhang
- Experimental Center of Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Guang Hu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Department of Bioinformatics, Center for Systems Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215213, China; (J.Q.); (S.W.)
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China
- Key Laboratory of Alkene-Carbon Fibres-Based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou 215123, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| |
Collapse
|
9
|
Maier A, Hartung M, Abovsky M, Adamowicz K, Bader G, Baier S, Blumenthal D, Chen J, Elkjaer M, Garcia-Hernandez C, Helmy M, Hoffmann M, Jurisica I, Kotlyar M, Lazareva O, Levi H, List M, Lobentanzer S, Loscalzo J, Malod-Dognin N, Manz Q, Matschinske J, Mee M, Oubounyt M, Pastrello C, Pico A, Pillich R, Poschenrieder J, Pratt D, Pržulj N, Sadegh S, Saez-Rodriguez J, Sarkar S, Shaked G, Shamir R, Trummer N, Turhan U, Wang RS, Zolotareva O, Baumbach J. Drugst.One - a plug-and-play solution for online systems medicine and network-based drug repurposing. Nucleic Acids Res 2024; 52:W481-W488. [PMID: 38783119 PMCID: PMC11223884 DOI: 10.1093/nar/gkae388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
In recent decades, the development of new drugs has become increasingly expensive and inefficient, and the molecular mechanisms of most pharmaceuticals remain poorly understood. In response, computational systems and network medicine tools have emerged to identify potential drug repurposing candidates. However, these tools often require complex installation and lack intuitive visual network mining capabilities. To tackle these challenges, we introduce Drugst.One, a platform that assists specialized computational medicine tools in becoming user-friendly, web-based utilities for drug repurposing. With just three lines of code, Drugst.One turns any systems biology software into an interactive web tool for modeling and analyzing complex protein-drug-disease networks. Demonstrating its broad adaptability, Drugst.One has been successfully integrated with 21 computational systems medicine tools. Available at https://drugst.one, Drugst.One has significant potential for streamlining the drug discovery process, allowing researchers to focus on essential aspects of pharmaceutical treatment research.
Collapse
Affiliation(s)
- Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Michael Hartung
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Mark Abovsky
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto, ON M5T 0S8, Canada
| | - Klaudia Adamowicz
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Sylvie Baier
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - David B Blumenthal
- Department Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Jing Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Maria L Elkjaer
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Mohamed Helmy
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Canada
- School of Public Health, University of Saskatchewan, Canada
- Department of Computer Science, University of Saskatchewan, Canada
- Department of Computer Science, Lakehead University, Canada
- Department of Computer Science, Idaho State University, USA
- Bioinformatics Institute (BII), A*STAR, Singapore
| | - Markus Hoffmann
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Germany
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD 20892, USA
| | - Igor Jurisica
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto, ON M5T 0S8, Canada
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Max Kotlyar
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto, ON M5T 0S8, Canada
| | - Olga Lazareva
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Junior Clinical Cooperation Unit Multiparametric methods for early detection of prostate cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Hagai Levi
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Markus List
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Sebastian Lobentanzer
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Quirin Manz
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Julian Matschinske
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Miles Mee
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Mhaned Oubounyt
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Chiara Pastrello
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto, ON M5T 0S8, Canada
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, 1650 Owens Street, San Francisco, 94158 California, USA
| | - Rudolf T Pillich
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Julian M Poschenrieder
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Dexter Pratt
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Nataša Pržulj
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, University College London, London WC1E 6BT, UK
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Sepideh Sadegh
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Suryadipto Sarkar
- Department Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Gideon Shaked
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Ron Shamir
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Nico Trummer
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Ugur Turhan
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Rui-Sheng Wang
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Olga Zolotareva
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Computational Biomedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
10
|
Chan JFW, Yuan S, Chu H, Sridhar S, Yuen KY. COVID-19 drug discovery and treatment options. Nat Rev Microbiol 2024; 22:391-407. [PMID: 38622352 DOI: 10.1038/s41579-024-01036-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 04/17/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused substantial morbidity and mortality, and serious social and economic disruptions worldwide. Unvaccinated or incompletely vaccinated older individuals with underlying diseases are especially prone to severe disease. In patients with non-fatal disease, long COVID affecting multiple body systems may persist for months. Unlike SARS-CoV and Middle East respiratory syndrome coronavirus, which have either been mitigated or remained geographically restricted, SARS-CoV-2 has disseminated globally and is likely to continue circulating in humans with possible emergence of new variants that may render vaccines less effective. Thus, safe, effective and readily available COVID-19 therapeutics are urgently needed. In this Review, we summarize the major drug discovery approaches, preclinical antiviral evaluation models, representative virus-targeting and host-targeting therapeutic options, and key therapeutics currently in clinical use for COVID-19. Preparedness against future coronavirus pandemics relies not only on effective vaccines but also on broad-spectrum antivirals targeting conserved viral components or universal host targets, and new therapeutics that can precisely modulate the immune response during infection.
Collapse
Affiliation(s)
- Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China.
| |
Collapse
|
11
|
Tang K, Sun Q, Zeng J, Tang J, Cheng P, Qiu Z, Long H, Chen Y, Zhang C, Wei J, Qiu X, Jiang G, Fang Q, Sun L, Sun C, Du X. Network-based approach for drug repurposing against mpox. Int J Biol Macromol 2024; 270:132468. [PMID: 38761900 DOI: 10.1016/j.ijbiomac.2024.132468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 04/28/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
The current outbreak of mpox presents a significant threat to the global community. However, the lack of mpox-specific drugs necessitates the identification of additional candidates for clinical trials. In this study, a network medicine framework was used to investigate poxviruses-human interactions to identify potential drugs effective against the mpox virus (MPXV). The results indicated that poxviruses preferentially target hubs on the human interactome, and that these virally-targeted proteins (VTPs) tend to aggregate together within specific modules. Comorbidity analysis revealed that mpox is closely related to immune system diseases. Based on predicted drug-target interactions, 268 drugs were identified using the network proximity approach, among which 23 drugs displaying the least side-effects and significant proximity to MPXV were selected as the final candidates. Lastly, specific drugs were explored based on VTPs, differentially expressed proteins, and intermediate nodes, corresponding to different categories. These findings provide novel insights that can contribute to a deeper understanding of the pathogenesis of MPXV and development of ready-to-use treatment strategies based on drug repurposing.
Collapse
Affiliation(s)
- Kang Tang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health, Guangdong Medical University, Dongguan 523808, PR China
| | - Qianru Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Preventive health division, Xijing Hospital, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, PR China
| | - Jinfeng Zeng
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jing Tang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Peiwen Cheng
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Zekai Qiu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Department of Molecular and Radiooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Medical Faculty Heidelberg, Heidelberg University, Heidelberg 69047, Germany
| | - Haoyu Long
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yilin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Chi Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jie Wei
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xiaoping Qiu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Shenzhen Key Laboratory of Pathogenic Microbes & Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Qianglin Fang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Shenzhen Key Laboratory of Pathogenic Microbes & Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Litao Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Shenzhen Key Laboratory of Pathogenic Microbes & Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Shenzhen Key Laboratory of Pathogenic Microbes & Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xiangjun Du
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Shenzhen Key Laboratory of Pathogenic Microbes & Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou 510030, PR China.
| |
Collapse
|
12
|
Xie W, Yu J, Huang L, For LS, Zheng Z, Chen X, Wang Y, Liu Z, Peng C, Wong KC. DeepSeq2Drug: An expandable ensemble end-to-end anti-viral drug repurposing benchmark framework by multi-modal embeddings and transfer learning. Comput Biol Med 2024; 175:108487. [PMID: 38653064 DOI: 10.1016/j.compbiomed.2024.108487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Drug repurposing is promising in multiple scenarios, such as emerging viral outbreak controls and cost reductions of drug discovery. Traditional graph-based drug repurposing methods are limited to fast, large-scale virtual screens, as they constrain the counts for drugs and targets and fail to predict novel viruses or drugs. Moreover, though deep learning has been proposed for drug repurposing, only a few methods have been used, including a group of pre-trained deep learning models for embedding generation and transfer learning. Hence, we propose DeepSeq2Drug to tackle the shortcomings of previous methods. We leverage multi-modal embeddings and an ensemble strategy to complement the numbers of drugs and viruses and to guarantee the novel prediction. This framework (including the expanded version) involves four modal types: six NLP models, four CV models, four graph models, and two sequence models. In detail, we first make a pipeline and calculate the predictive performance of each pair of viral and drug embeddings. Then, we select the best embedding pairs and apply an ensemble strategy to conduct anti-viral drug repurposing. To validate the effect of the proposed ensemble model, a monkeypox virus (MPV) case study is conducted to reflect the potential predictive capability. This framework could be a benchmark method for further pre-trained deep learning optimization and anti-viral drug repurposing tasks. We also build software further to make the proposed model easier to reuse. The code and software are freely available at http://deepseq2drug.cs.cityu.edu.hk.
Collapse
Affiliation(s)
- Weidun Xie
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Jixiang Yu
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Lei Huang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Lek Shyuen For
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Zetian Zheng
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Xingjian Chen
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuchen Wang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Zhichao Liu
- Sir William Dunn School of Pathology, University of Oxford, UK
| | - Chengbin Peng
- College of Information Science and Engineering, Ningbo University, Ningbo, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China; Hong Kong Institute for Data Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China.
| |
Collapse
|
13
|
Yu Z, Wu Z, Wang Z, Wang Y, Zhou M, Li W, Liu G, Tang Y. Network-Based Methods and Their Applications in Drug Discovery. J Chem Inf Model 2024; 64:57-75. [PMID: 38150548 DOI: 10.1021/acs.jcim.3c01613] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Drug discovery is time-consuming, expensive, and predominantly follows the "one drug → one target → one disease" paradigm. With the rapid development of systems biology and network pharmacology, a novel drug discovery paradigm, "multidrug → multitarget → multidisease", has emerged. This new holistic paradigm of drug discovery aligns well with the essence of networks, leading to the emergence of network-based methods in the field of drug discovery. In this Perspective, we initially introduce the concept and data sources of networks and highlight classical methodologies employed in network-based methods. Subsequently, we focus on the practical applications of network-based methods across various areas of drug discovery, such as target prediction, virtual screening, prediction of drug therapeutic effects or adverse drug events, and elucidation of molecular mechanisms. In addition, we provide representative web servers for researchers to use network-based methods in specific applications. Finally, we discuss several challenges of network-based methods and the directions for future development. In a word, network-based methods could serve as powerful tools to accelerate drug discovery.
Collapse
Affiliation(s)
- Zhuohang Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zengrui Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Ze Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yimeng Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Moran Zhou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
14
|
Zhou Y, Zhang Y, Zhao D, Yu X, Shen X, Zhou Y, Wang S, Qiu Y, Chen Y, Zhu F. TTD: Therapeutic Target Database describing target druggability information. Nucleic Acids Res 2024; 52:D1465-D1477. [PMID: 37713619 PMCID: PMC10767903 DOI: 10.1093/nar/gkad751] [Citation(s) in RCA: 189] [Impact Index Per Article: 189.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/31/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023] Open
Abstract
Target discovery is one of the essential steps in modern drug development, and the identification of promising targets is fundamental for developing first-in-class drug. A variety of methods have emerged for target assessment based on druggability analysis, which refers to the likelihood of a target being effectively modulated by drug-like agents. In the therapeutic target database (TTD), nine categories of established druggability characteristics were thus collected for 426 successful, 1014 clinical trial, 212 preclinical/patented, and 1479 literature-reported targets via systematic review. These characteristic categories were classified into three distinct perspectives: molecular interaction/regulation, human system profile and cell-based expression variation. With the rapid progression of technology and concerted effort in drug discovery, TTD and other databases were highly expected to facilitate the explorations of druggability characteristics for the discovery and validation of innovative drug target. TTD is now freely accessible at: https://idrblab.org/ttd/.
Collapse
Affiliation(s)
- Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Diagnosis and Treatment of Severe Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310000, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Donghai Zhao
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyuan Yu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yuan Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shanshan Wang
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Yunqing Qiu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Diagnosis and Treatment of Severe Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Yuzong Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
15
|
Pellegrini M. Advances in Network-Based Drug Repositioning. LECTURE NOTES IN COMPUTER SCIENCE 2024:99-114. [DOI: 10.1007/978-3-031-55248-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
16
|
Barjasteh AH, Al-Asady AM, Latifi H, Al Okla S, Al-Nazwani N, Avan A, Khazaei M, Ryzhikov M, Nadi-Yazdi H, Hassanian SM. Maximizing Treatment Options for IBD through Drug Repurposing. Curr Pharm Des 2024; 30:2538-2549. [PMID: 39039672 DOI: 10.2174/0113816128318032240702045822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/24/2024]
Abstract
Chronic inflammation characterizes Inflammatory Bowel Disease (IBD), encompassing Crohn's Disease (CD) and Ulcerative Colitis (UC). Despite modest activity of disease in most UC patients, exacerbations occur, especially in those with severe symptoms, necessitating interventions, like colectomy. Current treatments for IBD, predominantly small molecule therapies, impose significant economic burdens. Drug repurposing offers a cost-effective alternative, leveraging existing drugs for novel therapeutic applications. This approach capitalizes on shared molecular pathways across diseases, accelerating therapeutic discovery while minimizing costs and risks. This article provides an overview of IBD and explores drug repurposing as a promising avenue for more effective and affordable treatments. Through computational and animal studies, potential drug candidates are categorized, offering insights into IBD pathogenesis and treatment strategies.
Collapse
Affiliation(s)
| | - Abdulridha Mohammed Al-Asady
- Department of Medical Sciences, Faculty of Nursing, Warith Al-Anbiyaa University, Karbala, Iraq
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Karbala, Iraq
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanieh Latifi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Souad Al Okla
- College of Medicine and Health Sciences, National University of Science and Technology, Muscat, Oman
- Department of Animal Biology, Faculty of Sciences, Damascus University, Damascus, Syria
| | - Nasser Al-Nazwani
- Department of Biochemistry, College of Medicine and Health Sciences, National University of Science and Technology, Sohar, Oman
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- School of Medicine, Saint Louis University, St. Louis, MO 63103, USA
| | - Hanieh Nadi-Yazdi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Bharadwaj A, Kaur R, Gupta S. Emerging Treatment Approaches for COVID-19 Infection: A Critical Review. Curr Mol Med 2024; 24:435-448. [PMID: 37070448 DOI: 10.2174/1566524023666230417112543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 04/19/2023]
Abstract
In the present scenario, the SARS-CoV-2 virus has imposed enormous damage on human survival and the global financial system. It has been estimated that around 111 million people all around the world have been infected, and about 2.47 million people died due to this pandemic. The major symptoms were sneezing, coughing, cold, difficulty breathing, pneumonia, and multi-organ failure associated 1with SARS-CoV-2. Currently, two key problems, namely insufficient attempts to develop drugs against SARSCoV-2 and the lack of any biological regulating process, are mostly responsible for the havoc caused by this virus. Henceforth, developing a few novel drugs is urgently required to cure this pandemic. It has been noticed that the pathogenesis of COVID-19 is caused by two main events: infection and immune deficiency, that occur during the pathological process. Antiviral medication can treat both the virus and the host cells. Therefore, in the present review, the major approaches for the treatment have been divided into "target virus" and "target host" groups. These two mechanisms primarily rely on drug repositioning, novel approaches, and possible targets. Initially, we discussed the traditional drugs per the physicians' recommendations. Moreover, such therapeutics have no potential to fight against COVID-19. After that, detailed investigation and analysis were conducted to find some novel vaccines and monoclonal antibodies and conduct a few clinical trials to check their effectiveness against SARSCoV- 2 and mutant strains. Additionally, this study presents the most successful methods for its treatment, including combinatorial therapy. Nanotechnology was studied to build efficient nanocarriers to overcome the traditional constraints of antiviral and biological therapies.
Collapse
Affiliation(s)
- Alok Bharadwaj
- Department of Biotechnology, GLA University, Mathura, 281406, UP, India
| | - Rasanpreet Kaur
- Department of Biotechnology, GLA University, Mathura, 281406, UP, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, 281406, UP, India
| |
Collapse
|
18
|
Tan M, Xia J, Luo H, Meng G, Zhu Z. Applying the digital data and the bioinformatics tools in SARS-CoV-2 research. Comput Struct Biotechnol J 2023; 21:4697-4705. [PMID: 37841328 PMCID: PMC10568291 DOI: 10.1016/j.csbj.2023.09.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023] Open
Abstract
Bioinformatics has been playing a crucial role in the scientific progress to fight against the pandemic of the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The advances in novel algorithms, mega data technology, artificial intelligence and deep learning assisted the development of novel bioinformatics tools to analyze daily increasing SARS-CoV-2 data in the past years. These tools were applied in genomic analyses, evolutionary tracking, epidemiological analyses, protein structure interpretation, studies in virus-host interaction and clinical performance. To promote the in-silico analysis in the future, we conducted a review which summarized the databases, web services and software applied in SARS-CoV-2 research. Those digital resources applied in SARS-CoV-2 research may also potentially contribute to the research in other coronavirus and non-coronavirus viruses.
Collapse
Affiliation(s)
- Meng Tan
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiaxin Xia
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Haitao Luo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Geng Meng
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhenglin Zhu
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
19
|
Häring C, Jungwirth J, Schroeder J, Löffler B, Engert B, Ehrhardt C. The Local Anaesthetic Procaine Prodrugs ProcCluster ® and Procaine Hydrochloride Impair SARS-CoV-2 Replication and Egress In Vitro. Int J Mol Sci 2023; 24:14584. [PMID: 37834031 PMCID: PMC10572566 DOI: 10.3390/ijms241914584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
As vaccination efforts against SARS-CoV-2 progress in many countries, there is still an urgent need for efficient antiviral treatment strategies for those with severer disease courses, and lately, considerable efforts have been undertaken to repurpose existing drugs as antivirals. The local anaesthetic procaine has been investigated for antiviral properties against several viruses over the past decades. Here, we present data on the inhibitory effect of the procaine prodrugs ProcCluster® and procaine hydrochloride on SARS-CoV-2 infection in vitro. Both procaine prodrugs limit SARS-CoV-2 progeny virus titres as well as reduce interferon and cytokine responses in a proportional manner to the virus load. The addition of procaine during the early stages of the SARS-CoV-2 replication cycle in a cell culture first limits the production of subgenomic RNA transcripts, and later affects the replication of the viral genomic RNA. Interestingly, procaine additionally exerts a prominent effect on SARS-CoV-2 progeny virus release when added late during the replication cycle, when viral RNA production and protein production are already largely completed.
Collapse
Affiliation(s)
- Clio Häring
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany; (C.H.); (J.J.); (J.S.)
| | - Johannes Jungwirth
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany; (C.H.); (J.J.); (J.S.)
| | - Josefine Schroeder
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany; (C.H.); (J.J.); (J.S.)
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany;
| | | | - Christina Ehrhardt
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany; (C.H.); (J.J.); (J.S.)
| |
Collapse
|
20
|
Wei X, Pan C, Zhang X, Zhang W. Total network controllability analysis discovers explainable drugs for Covid-19 treatment. Biol Direct 2023; 18:55. [PMID: 37670359 PMCID: PMC10478273 DOI: 10.1186/s13062-023-00410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND The active pursuit of network medicine for drug repurposing, particularly for combating Covid-19, has stimulated interest in the concept of structural controllability in cellular networks. We sought to extend this theory, focusing on the defense rather than control of the cell against viral infections. Accordingly, we extended structural controllability to total structural controllability and introduced the concept of control hubs. Perturbing any control hub may render the cell uncontrollable by exogenous stimuli like viral infections, so control hubs are ideal drug targets. RESULTS We developed an efficient algorithm to identify all control hubs, applying it to a largest homogeneous network of human protein interactions, including interactions between human and SARS-CoV-2 proteins. Our method recognized 65 druggable control hubs with enriched antiviral functions. Utilizing these hubs, we categorized potential drugs into four groups: antiviral and anti-inflammatory agents, drugs acting on the central nervous system, dietary supplements, and compounds enhancing immunity. An exemplification of our approach's effectiveness, Fostamatinib, a drug initially developed for chronic immune thrombocytopenia, is now in clinical trials for treating Covid-19. Preclinical trial data demonstrated that Fostamatinib could reduce mortality rates, ICU stay length, and disease severity in Covid-19 patients. CONCLUSIONS Our findings confirm the efficacy of our novel strategy that leverages control hubs as drug targets. This approach provides insights into the molecular mechanisms of potential therapeutics for Covid-19, making it a valuable tool for interpretable drug discovery. Our new approach is general and applicable to repurposing drugs for other diseases.
Collapse
Affiliation(s)
- Xinru Wei
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, 210001, China
| | - Chunyu Pan
- School of Computer Science and Engineering, Northeastern University, Shenyang, Liaoning, 110167, China
| | - Xizhe Zhang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, 210001, China.
| | - Weixiong Zhang
- Department of Health Technology and Informatics, Department of Computing, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
21
|
Wei X, Pan C, Zhang X, Zhang W. Total network controllability analysis discovers explainable drugs for Covid-19 treatment. RESEARCH SQUARE 2023:rs.3.rs-3147521. [PMID: 37503262 PMCID: PMC10371104 DOI: 10.21203/rs.3.rs-3147521/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background The active pursuit of network medicine for drug repurposing, particularly for combating Covid-19, has stimulated interest in the concept of structural control capability in cellular networks. We sought to extend this theory, focusing on the defense rather than control of the cell against viral infections. Accordingly, we extended structural controllability to total structural controllability and introduced the concept of control hubs. Perturbing any control hub may render the cell uncontrollable by exogenous stimuli like viral infections, so control hubs are ideal drug targets. Results We developed an efficient algorithm to identify all control hubs, applying it to the largest homogeneous network of human protein interactions, including interactions between human and SARS-CoV-2 proteins. Our method recognized 65 druggable control hubs with enriched antiviral functions. Utilizing these hubs, we categorized potential drugs into four groups: antiviral and anti-inflammatory agents, drugs acting on the central nervous system, dietary supplements, and compounds enhancing immunity. An exemplification of our approach's effectiveness, Fostamatinib, a drug initially developed for chronic immune thrombocytopenia, is now in clinical trials for treating Covid-19. Preclinical trial data demonstrated that Fostamatinib could reduce mortality rates, ICU stay length, and disease severity in Covid-19 patients. Conclusions Our findings confirm the efficacy of our novel strategy that leverages control hubs as drug targets. This approach provides insights into the molecular mechanisms of potential therapeutics for Covid-19, making it a valuable tool for interpretable drug discovery.
Collapse
Affiliation(s)
- Xinru Wei
- The Affiliated Brain Hospital of Nanjing Medical University
| | | | - Xizhe Zhang
- The Affiliated Brain Hospital of Nanjing Medical University
| | | |
Collapse
|
22
|
Maier A, Hartung M, Abovsky M, Adamowicz K, Bader GD, Baier S, Blumenthal DB, Chen J, Elkjaer ML, Garcia-Hernandez C, Helmy M, Hoffmann M, Jurisica I, Kotlyar M, Lazareva O, Levi H, List M, Lobentanzer S, Loscalzo J, Malod-Dognin N, Manz Q, Matschinske J, Mee M, Oubounyt M, Pico AR, Pillich RT, Poschenrieder JM, Pratt D, Pržulj N, Sadegh S, Saez-Rodriguez J, Sarkar S, Shaked G, Shamir R, Trummer N, Turhan U, Wang R, Zolotareva O, Baumbach J. Drugst.One - A plug-and-play solution for online systems medicine and network-based drug repurposing. ARXIV 2023:arXiv:2305.15453v2. [PMID: 37332567 PMCID: PMC10274948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
In recent decades, the development of new drugs has become increasingly expensive and inefficient, and the molecular mechanisms of most pharmaceuticals remain poorly understood. In response, computational systems and network medicine tools have emerged to identify potential drug repurposing candidates. However, these tools often require complex installation and lack intuitive visual network mining capabilities. To tackle these challenges, we introduce Drugst.One, a platform that assists specialized computational medicine tools in becoming user-friendly, web-based utilities for drug repurposing. With just three lines of code, Drugst.One turns any systems biology software into an interactive web tool for modeling and analyzing complex protein-drug-disease networks. Demonstrating its broad adaptability, Drugst.One has been successfully integrated with 21 computational systems medicine tools. Available at https://drugst.one, Drugst.One has significant potential for streamlining the drug discovery process, allowing researchers to focus on essential aspects of pharmaceutical treatment research.
Collapse
Affiliation(s)
- Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Michael Hartung
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Mark Abovsky
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, and Data Science Discovery Centre, Osteoarthritis Research Program, Krembil Research Institute, UHN, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, 60 Leonard Avenue, 5KD-407, Toronto, ON, M5T 0S8, Canada
| | - Klaudia Adamowicz
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Sylvie Baier
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - David B Blumenthal
- Department Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Jing Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Maria L Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Mohamed Helmy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Markus Hoffmann
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Advanced Study (Lichtenbergstrasse 2a, D-85748 Garching, Germany), Technical University of Munich, Germany
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD 20892, United States of America
| | - Igor Jurisica
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, and Data Science Discovery Centre, Osteoarthritis Research Program, Krembil Research Institute, UHN, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, 60 Leonard Avenue, 5KD-407, Toronto, ON, M5T 0S8, Canada
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Max Kotlyar
- Division of Orthopaedic Surgery, Schroeder Arthritis Institute, and Data Science Discovery Centre, Osteoarthritis Research Program, Krembil Research Institute, UHN, Toronto, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, 60 Leonard Avenue, 5KD-407, Toronto, ON, M5T 0S8, Canada
| | - Olga Lazareva
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Junior Clinical Cooperation Unit Multiparametric methods for early detection of prostate cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Hagai Levi
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Sebastian Lobentanzer
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Quirin Manz
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Julian Matschinske
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Miles Mee
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Mhaned Oubounyt
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, 1650 Owens Street, San Francisco, 94158, California, USA
| | - Rudolf T Pillich
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Julian M Poschenrieder
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Dexter Pratt
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Nataša Pržulj
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, University College London, London WC1E 6BT, UK
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Sepideh Sadegh
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Suryadipto Sarkar
- Department Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Gideon Shaked
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Ron Shamir
- Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel
| | - Nico Trummer
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Ugur Turhan
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Ruisheng Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Olga Zolotareva
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Computational Biomedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
23
|
Majumder R, Ghosh S, Singh MK, Das A, Roy Chowdhury S, Saha A, Saha RP. Revisiting the COVID-19 Pandemic: An Insight into Long-Term Post-COVID Complications and Repurposing of Drugs. COVID 2023; 3:494-519. [DOI: 10.3390/covid3040037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
SARS-CoV-2 is a highly contagious and dangerous coronavirus that has been spreading around the world since late December 2019. Severe COVID-19 has been observed to induce severe damage to the alveoli, and the slow loss of lung function led to the deaths of many patients. Scientists from all over the world are now saying that SARS-CoV-2 can spread through the air, which is a very frightening prospect for humans. Many scientists thought that this virus would evolve during the first wave of the pandemic and that the second wave of reinfection with the coronavirus would also be very dangerous. In late 2020 and early 2021, researchers found different genetic versions of the SARS-CoV-2 virus in many places around the world. Patients with different types of viruses had different symptoms. It is now evident from numerous case studies that many COVID-19 patients who are released from nursing homes or hospitals are more prone to developing multi-organ dysfunction than the general population. Understanding the pathophysiology of COVID-19 and its impact on various organ systems is crucial for developing effective treatment strategies and managing long-term health consequences. The case studies highlighted in this review provide valuable insights into the ongoing health concerns of individuals affected by COVID-19.
Collapse
Affiliation(s)
- Rajib Majumder
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Sanmitra Ghosh
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Manoj K. Singh
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Swagata Roy Chowdhury
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Abinit Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| | - Rudra P. Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata 700126, India
| |
Collapse
|
24
|
Systematic Guidelines for Effective Utilization of COVID-19 Databases in Genomic, Epidemiologic, and Clinical Research. Viruses 2023; 15:v15030692. [PMID: 36992400 PMCID: PMC10059256 DOI: 10.3390/v15030692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/27/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
The pandemic has led to the production and accumulation of various types of data related to coronavirus disease 2019 (COVID-19). To understand the features and characteristics of COVID-19 data, we summarized representative databases and determined the data types, purpose, and utilization details of each database. In addition, we categorized COVID-19 associated databases into epidemiological data, genome and protein data, and drug and target data. We found that the data present in each of these databases have nine separate purposes (clade/variant/lineage, genome browser, protein structure, epidemiological data, visualization, data analysis tool, treatment, literature, and immunity) according to the types of data. Utilizing the databases we investigated, we created four queries as integrative analysis methods that aimed to answer important scientific questions related to COVID-19. Our queries can make effective use of multiple databases to produce valuable results that can reveal novel findings through comprehensive analysis. This allows clinical researchers, epidemiologists, and clinicians to have easy access to COVID-19 data without requiring expert knowledge in computing or data science. We expect that users will be able to reference our examples to construct their own integrative analysis methods, which will act as a basis for further scientific inquiry and data searching.
Collapse
|
25
|
DRaW: prediction of COVID-19 antivirals by deep learning-an objection on using matrix factorization. BMC Bioinformatics 2023; 24:52. [PMID: 36793010 PMCID: PMC9931173 DOI: 10.1186/s12859-023-05181-8] [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: 11/04/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Due to the high resource consumption of introducing a new drug, drug repurposing plays an essential role in drug discovery. To do this, researchers examine the current drug-target interaction (DTI) to predict new interactions for the approved drugs. Matrix factorization methods have much attention and utilization in DTIs. However, they suffer from some drawbacks. METHODS We explain why matrix factorization is not the best for DTI prediction. Then, we propose a deep learning model (DRaW) to predict DTIs without having input data leakage. We compare our model with several matrix factorization methods and a deep model on three COVID-19 datasets. In addition, to ensure the validation of DRaW, we evaluate it on benchmark datasets. Furthermore, as an external validation, we conduct a docking study on the COVID-19 recommended drugs. RESULTS In all cases, the results confirm that DRaW outperforms matrix factorization and deep models. The docking results approve the top-ranked recommended drugs for COVID-19. CONCLUSIONS In this paper, we show that it may not be the best choice to use matrix factorization in the DTI prediction. Matrix factorization methods suffer from some intrinsic issues, e.g., sparsity in the domain of bioinformatics applications and fixed-unchanged size of the matrix-related paradigm. Therefore, we propose an alternative method (DRaW) that uses feature vectors rather than matrix factorization and demonstrates better performance than other famous methods on three COVID-19 and four benchmark datasets.
Collapse
|
26
|
Xenos A, Malod-Dognin N, Zambrana C, Pržulj N. Integrated Data Analysis Uncovers New COVID-19 Related Genes and Potential Drug Re-Purposing Candidates. Int J Mol Sci 2023; 24:1431. [PMID: 36674947 PMCID: PMC9863794 DOI: 10.3390/ijms24021431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
The COVID-19 pandemic is an acute and rapidly evolving global health crisis. To better understand this disease's molecular basis and design therapeutic strategies, we built upon the recently proposed concept of an integrated cell, iCell, fusing three omics, tissue-specific human molecular interaction networks. We applied this methodology to construct infected and control iCells using gene expression data from patient samples and three cell lines. We found large differences between patient-based and cell line-based iCells (both infected and control), suggesting that cell lines are ill-suited to studying this disease. We compared patient-based infected and control iCells and uncovered genes whose functioning (wiring patterns in iCells) is altered by the disease. We validated in the literature that 18 out of the top 20 of the most rewired genes are indeed COVID-19-related. Since only three of these genes are targets of approved drugs, we applied another data fusion step to predict drugs for re-purposing. We confirmed with molecular docking that the predicted drugs can bind to their predicted targets. Our most interesting prediction is artenimol, an antimalarial agent targeting ZFP62, one of our newly identified COVID-19-related genes. This drug is a derivative of artemisinin drugs that are already under clinical investigation for their potential role in the treatment of COVID-19. Our results demonstrate further applicability of the iCell framework for integrative comparative studies of human diseases.
Collapse
Affiliation(s)
- Alexandros Xenos
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, Universitat Politecnica de Catalunya (UPC), 08034 Barcelona, Spain
| | - Noël Malod-Dognin
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Carme Zambrana
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, Universitat Politecnica de Catalunya (UPC), 08034 Barcelona, Spain
| | - Nataša Pržulj
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, University College London, London WC1E 6BT, UK
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
27
|
Ray S, Lall S, Mukhopadhyay A, Bandyopadhyay S, Schönhuth A. Deep variational graph autoencoders for novel host-directed therapy options against COVID-19. Artif Intell Med 2022; 134:102418. [PMID: 36462892 PMCID: PMC9556806 DOI: 10.1016/j.artmed.2022.102418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/22/2022] [Accepted: 10/02/2022] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic has been keeping asking urgent questions with respect to therapeutic options. Existing drugs that can be repurposed promise rapid implementation in practice because of their prior approval. Conceivably, there is still room for substantial improvement, because most advanced artificial intelligence techniques for screening drug repositories have not been exploited so far. We construct a comprehensive network by combining year-long curated drug-protein/protein-protein interaction data on the one hand, and most recent SARS-CoV-2 protein interaction data on the other hand. We learn the structure of the resulting encompassing molecular interaction network and predict missing links using variational graph autoencoders (VGAEs), as a most advanced deep learning technique that has not been explored so far. We focus on hitherto unknown links between drugs and human proteins that play key roles in the replication cycle of SARS-CoV-2. Thereby, we establish novel host-directed therapy (HDT) options whose utmost plausibility is confirmed by realistic simulations. As a consequence, many of the predicted links are likely to be crucial for the virus to thrive on the one hand, and can be targeted with existing drugs on the other hand.
Collapse
Affiliation(s)
- Sumanta Ray
- Department of Computer Science and Engineering, Aliah University, New Town, Kolkata, India; Health Analytics Network, PA, USA.
| | - Snehalika Lall
- Machine Intelligence Unit, Indian Statistical Institute, Kolkata, India
| | - Anirban Mukhopadhyay
- Department of Computer Science and Engineering, University of Kalyani, Kalyani, India
| | | | | |
Collapse
|
28
|
Li J, Xue Y, Wang X, Smith LS, He B, Liu S, Zhu H. Tissue- and cell-expression of druggable host proteins provide insights into repurposing drugs for COVID-19. Clin Transl Sci 2022; 15:2796-2811. [PMID: 36259251 PMCID: PMC9747131 DOI: 10.1111/cts.13400] [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: 03/12/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 01/26/2023] Open
Abstract
Several human host proteins play important roles in the lifecycle of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Many drugs targeting these host proteins have been investigated as potential therapeutics for coronavirus disease 2019 (COVID-19). The tissue-specific expressions of selected host proteins were summarized using proteomics data retrieved from the Human Protein Atlas, ProteomicsDB, Human Proteome Map databases, and a clinical COVID-19 study. Protein expression features in different cell lines were summarized based on recent proteomics studies. The half-maximal effective concentration or half-maximal inhibitory concentration values were collected from in vitro studies. The pharmacokinetic data were mainly from studies in healthy subjects or non-COVID-19 patients. Considerable tissue-specific expression patterns were observed for several host proteins. ACE2 expression in the lungs was significantly lower than in many other tissues (e.g., the kidneys and intestines); TMPRSS2 expression in the lungs was significantly lower than in other tissues (e.g., the prostate and intestines). The expression levels of endocytosis-associated proteins CTSL, CLTC, NPC1, and PIKfyve in the lungs were comparable to or higher than most other tissues. TMPRSS2 expression was markedly different between cell lines, which could be associated with the cell-dependent antiviral activities of several drugs. Drug delivery receptor ICAM1 and CTSB were expressed at a higher level in the lungs than in other tissues. In conclusion, the cell- and tissue-specific proteomics data could help interpret the in vitro antiviral activities of host-directed drugs in various cells and aid the transition of the in vitro findings to clinical research to develop safe and effective therapeutics for COVID-19.
Collapse
Affiliation(s)
- Jiapeng Li
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Yanling Xue
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Xinwen Wang
- Department of Pharmaceutical SciencesNortheast Ohio Medical University College of PharmacyRootstownOhioUSA
| | - Logan S. Smith
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Bing He
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
| | - Shuhan Liu
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| | - Hao‐Jie Zhu
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| |
Collapse
|
29
|
Buzzao D, Castresana-Aguirre M, Guala D, Sonnhammer ELL. TOPAS, a network-based approach to detect disease modules in a top-down fashion. NAR Genom Bioinform 2022; 4:lqac093. [PMCID: PMC9706483 DOI: 10.1093/nargab/lqac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/14/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022] Open
Abstract
A vast scenario of potential disease mechanisms and remedies is yet to be discovered. The field of Network Medicine has grown thanks to the massive amount of high-throughput data and the emerging evidence that disease-related proteins form ‘disease modules’. Relying on prior disease knowledge, network-based disease module detection algorithms aim at connecting the list of known disease associated genes by exploiting interaction networks. Most existing methods extend disease modules by iteratively adding connector genes in a bottom-up fashion, while top-down approaches remain largely unexplored. We have created TOPAS, an iterative approach that aims at connecting the largest number of seed nodes in a top-down fashion through connectors that guarantee the highest flow of a Random Walk with Restart in a network of functional associations. We used a corpus of 382 manually selected functional gene sets to benchmark our algorithm against SCA, DIAMOnD, MaxLink and ROBUST across four interactomes. We demonstrate that TOPAS outperforms competing methods in terms of Seed Recovery Rate, Seed to Connector Ratio and consistency during module detection. We also show that TOPAS achieves competitive performance in terms of biological relevance of detected modules and scalability.
Collapse
Affiliation(s)
- Davide Buzzao
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Box 1031, 171 21 Solna, Sweden
| | | | - Dimitri Guala
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Box 1031, 171 21 Solna, Sweden
| | | |
Collapse
|
30
|
Ravindran V, Wagoner J, Athanasiadis P, Den Hartigh AB, Sidorova JM, Ianevski A, Fink SL, Frigessi A, White J, Polyak SJ, Aittokallio T. Discovery of host-directed modulators of virus infection by probing the SARS-CoV-2-host protein-protein interaction network. Brief Bioinform 2022; 23:bbac456. [PMID: 36305426 PMCID: PMC9677461 DOI: 10.1093/bib/bbac456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/05/2022] [Accepted: 09/23/2022] [Indexed: 12/14/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has highlighted the need to better understand virus-host interactions. We developed a network-based method that expands the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-host protein interaction network and identifies host targets that modulate viral infection. To disrupt the SARS-CoV-2 interactome, we systematically probed for potent compounds that selectively target the identified host proteins with high expression in cells relevant to COVID-19. We experimentally tested seven chemical inhibitors of the identified host proteins for modulation of SARS-CoV-2 infection in human cells that express ACE2 and TMPRSS2. Inhibition of the epigenetic regulators bromodomain-containing protein 4 (BRD4) and histone deacetylase 2 (HDAC2), along with ubiquitin-specific peptidase (USP10), enhanced SARS-CoV-2 infection. Such proviral effect was observed upon treatment with compounds JQ1, vorinostat, romidepsin and spautin-1, when measured by cytopathic effect and validated by viral RNA assays, suggesting that the host proteins HDAC2, BRD4 and USP10 have antiviral functions. We observed marked differences in antiviral effects across cell lines, which may have consequences for identification of selective modulators of viral infection or potential antiviral therapeutics. While network-based approaches enable systematic identification of host targets and selective compounds that may modulate the SARS-CoV-2 interactome, further developments are warranted to increase their accuracy and cell-context specificity.
Collapse
Affiliation(s)
- Vandana Ravindran
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
| | - Jessica Wagoner
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Paschalis Athanasiadis
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
| | - Andreas B Den Hartigh
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Julia M Sidorova
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Susan L Fink
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Arnoldo Frigessi
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Judith White
- Department of Cell Biology and Department of Microbiology, University of Virginia, Charlottesville, VA, USA
| | - Stephen J Polyak
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Tero Aittokallio
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Lu L, Qin J, Chen J, Yu N, Miyano S, Deng Z, Li C. Recent computational drug repositioning strategies against SARS-CoV-2. Comput Struct Biotechnol J 2022; 20:5713-5728. [PMID: 36277237 PMCID: PMC9575573 DOI: 10.1016/j.csbj.2022.10.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/08/2022] Open
Abstract
We performed a comprehensive review of computational drug repositioning methods applied to COVID-19 based on differing data types including sequence data, expression data, structure data and interaction data. We found that graph theory and neural network were the most used strategies for drug repositioning in the case of COVID-19. Integrating different levels of data may improve the success rate for drug repositioning.
Since COVID-19 emerged in 2019, significant levels of suffering and disruption have been caused on a global scale. Although vaccines have become widely used, the virus has shown its potential for evading immunities or acquiring other novel characteristics. Whether current drug treatments are still effective for people infected with Omicron remains unclear. Due to the long development cycles and high expense requirements of de novo drug development, many researchers have turned to consider drug repositioning in the search to find effective treatments for COVID-19. Here, we review such drug repositioning and combination efforts towards providing better handling. For potential drugs under consideration, aspects of both structure and function require attention, with specific categories of sequence, expression, structure, and interaction, the key parameters for investigation. For different data types, we show the corresponding differing drug repositioning methods that have been exploited. As incorporating drug combinations can increase therapeutic efficacy and reduce toxicity, we also review computational strategies to reveal drug combination potential. Taken together, we found that graph theory and neural network were the most used strategy with high potential towards drug repositioning for COVID-19. Integrating different levels of data may further improve the success rate of drug repositioning.
Collapse
Affiliation(s)
- Lu Lu
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiale Qin
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, China
| | - Jiandong Chen
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,School of Public Health, Undergraduate School of Zhejiang University, Hangzhou, China
| | - Na Yu
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Zhenzhong Deng
- Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China,Corresponding authors at: Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (C. Li).
| | - Chen Li
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China,Corresponding authors at: Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (C. Li).
| |
Collapse
|
32
|
Yue R, Dutta A. Computational systems biology in disease modeling and control, review and perspectives. NPJ Syst Biol Appl 2022; 8:37. [PMID: 36192551 PMCID: PMC9528884 DOI: 10.1038/s41540-022-00247-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/05/2022] [Indexed: 02/02/2023] Open
Abstract
Omics-based approaches have become increasingly influential in identifying disease mechanisms and drug responses. Considering that diseases and drug responses are co-expressed and regulated in the relevant omics data interactions, the traditional way of grabbing omics data from single isolated layers cannot always obtain valuable inference. Also, drugs have adverse effects that may impair patients, and launching new medicines for diseases is costly. To resolve the above difficulties, systems biology is applied to predict potential molecular interactions by integrating omics data from genomic, proteomic, transcriptional, and metabolic layers. Combined with known drug reactions, the resulting models improve medicines' therapeutical performance by re-purposing the existing drugs and combining drug molecules without off-target effects. Based on the identified computational models, drug administration control laws are designed to balance toxicity and efficacy. This review introduces biomedical applications and analyses of interactions among gene, protein and drug molecules for modeling disease mechanisms and drug responses. The therapeutical performance can be improved by combining the predictive and computational models with drug administration designed by control laws. The challenges are also discussed for its clinical uses in this work.
Collapse
Affiliation(s)
- Rongting Yue
- Department of Electrical and Computer Engineering, University of Connecticut, 371 Fairfield Way, Storrs, CT, 06269, USA.
| | - Abhishek Dutta
- Department of Electrical and Computer Engineering, University of Connecticut, 371 Fairfield Way, Storrs, CT, 06269, USA
| |
Collapse
|
33
|
Niranjan V, Setlur AS, Karunakaran C, Uttarkar A, Kumar KM, Skariyachan S. Scope of repurposed drugs against the potential targets of the latest variants of SARS-CoV-2. Struct Chem 2022; 33:1585-1608. [PMID: 35938064 PMCID: PMC9346052 DOI: 10.1007/s11224-022-02020-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022]
Abstract
The unprecedented outbreak of the severe acute respiratory syndrome (SARS) Coronavirus-2, across the globe, triggered a worldwide uproar in the search for immediate treatment strategies. With no specific drug and not much data available, alternative approaches such as drug repurposing came to the limelight. To date, extensive research on the repositioning of drugs has led to the identification of numerous drugs against various important protein targets of the coronavirus strains, with hopes of the drugs working against the major variants of concerns (alpha, beta, gamma, delta, omicron) of the virus. Advancements in computational sciences have led to improved scope of repurposing via techniques such as structure-based approaches including molecular docking, molecular dynamic simulations and quantitative structure activity relationships, network-based approaches, and artificial intelligence-based approaches with other core machine and deep learning algorithms. This review highlights the various approaches to repurposing drugs from a computational biological perspective, with various mechanisms of action of the drugs against some of the major protein targets of SARS-CoV-2. Additionally, clinical trials data on potential COVID-19 repurposed drugs are also highlighted with stress on the major SARS-CoV-2 targets and the structural effect of variants on these targets. The interaction modelling of some important repurposed drugs has also been elucidated. Furthermore, the merits and demerits of drug repurposing are also discussed, with a focus on the scope and applications of the latest advancements in repurposing.
Collapse
Affiliation(s)
- Vidya Niranjan
- Department of Biotechnology, RV College of Engineering, Bengaluru, Karnataka India
| | | | | | - Akshay Uttarkar
- Department of Biotechnology, RV College of Engineering, Bengaluru, Karnataka India
| | - Kalavathi Murugan Kumar
- Department of Bioinformatics, Pondicherry University, Chinna Kalapet, Kalapet, Puducherry, Tamil Nadu India
| | - Sinosh Skariyachan
- Department of Microbiology, St. Pius X College, Rajapuram, Kasaragod, Kerala India
| |
Collapse
|
34
|
Gao K, Wang R, Chen J, Cheng L, Frishcosy J, Huzumi Y, Qiu Y, Schluckbier T, Wei X, Wei GW. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chem Rev 2022; 122:11287-11368. [PMID: 35594413 PMCID: PMC9159519 DOI: 10.1021/acs.chemrev.1c00965] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite tremendous efforts in the past two years, our understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), virus-host interactions, immune response, virulence, transmission, and evolution is still very limited. This limitation calls for further in-depth investigation. Computational studies have become an indispensable component in combating coronavirus disease 2019 (COVID-19) due to their low cost, their efficiency, and the fact that they are free from safety and ethical constraints. Additionally, the mechanism that governs the global evolution and transmission of SARS-CoV-2 cannot be revealed from individual experiments and was discovered by integrating genotyping of massive viral sequences, biophysical modeling of protein-protein interactions, deep mutational data, deep learning, and advanced mathematics. There exists a tsunami of literature on the molecular modeling, simulations, and predictions of SARS-CoV-2 and related developments of drugs, vaccines, antibodies, and diagnostics. To provide readers with a quick update about this literature, we present a comprehensive and systematic methodology-centered review. Aspects such as molecular biophysics, bioinformatics, cheminformatics, machine learning, and mathematics are discussed. This review will be beneficial to researchers who are looking for ways to contribute to SARS-CoV-2 studies and those who are interested in the status of the field.
Collapse
Affiliation(s)
- Kaifu Gao
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jiahui Chen
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Limei Cheng
- Clinical
Pharmacology and Pharmacometrics, Bristol
Myers Squibb, Princeton, New Jersey 08536, United States
| | - Jaclyn Frishcosy
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuta Huzumi
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuchi Qiu
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tom Schluckbier
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xiaoqi Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
35
|
Pan X, Lin X, Cao D, Zeng X, Yu PS, He L, Nussinov R, Cheng F. Deep learning for drug repurposing: Methods, databases, and applications. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2022. [DOI: 10.1002/wcms.1597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xiaoqin Pan
- School of Computer Science and Engineering Hunan University Changsha Hunan China
| | - Xuan Lin
- School of Computer Science Xiangtan University Xiangtan China
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education Xiangtan University Xiangtan China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha China
| | - Xiangxiang Zeng
- School of Computer Science and Engineering Hunan University Changsha Hunan China
| | - Philip S. Yu
- Department of Computer Science University of Illinois at Chicago Chicago Illinois USA
| | - Lifang He
- Department of Computer Science and Engineering Lehigh University Bethlehem Pennsylvania USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research National Cancer Institute at Frederick Frederick Maryland USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Cleveland Ohio USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine Case Western Reserve University Cleveland Ohio USA
- Case Comprehensive Cancer Center Case Western Reserve University School of Medicine Cleveland Ohio USA
| |
Collapse
|
36
|
Zou L, Moch C, Graille M, Chapat C. The SARS-CoV-2 protein NSP2 impairs the silencing capacity of the human 4EHP-GIGYF2 complex. iScience 2022; 25:104646. [PMID: 35756894 PMCID: PMC9213009 DOI: 10.1016/j.isci.2022.104646] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/12/2022] [Accepted: 06/15/2022] [Indexed: 01/20/2023] Open
Abstract
There is an urgent need for a molecular understanding of how SARS-CoV-2 influences the machineries of the host cell. Herein, we focused our attention on the capacity of the SARS-CoV-2 protein NSP2 to bind the human 4EHP-GIGYF2 complex, a key factor involved in microRNA-mediated silencing of gene expression. Using in vitro interaction assays, our data demonstrate that NSP2 physically associates with both 4EHP and a central segment in GIGYF2 in the cytoplasm. We also provide functional evidence showing that NSP2 impairs the function of GIGYF2 in mediating translation repression using reporter-based assays. Collectively, these data reveal the potential impact of NSP2 on the post-transcriptional silencing of gene expression in human cells, pointing out 4EHP-GIGYF2 targeting as a possible strategy of SARS-CoV-2 to take over the silencing machinery and to suppress host defenses.
Collapse
Affiliation(s)
- Limei Zou
- Laboratoire de Biologie Structurale de la Cellule (BIOC), CNRS, Ecole polytechnique, IP Paris. F-91128 Palaiseau, France
| | - Clara Moch
- Laboratoire de Biologie Structurale de la Cellule (BIOC), CNRS, Ecole polytechnique, IP Paris. F-91128 Palaiseau, France
| | - Marc Graille
- Laboratoire de Biologie Structurale de la Cellule (BIOC), CNRS, Ecole polytechnique, IP Paris. F-91128 Palaiseau, France
| | - Clément Chapat
- Laboratoire de Biologie Structurale de la Cellule (BIOC), CNRS, Ecole polytechnique, IP Paris. F-91128 Palaiseau, France
| |
Collapse
|
37
|
Rintala TJ, Ghosh A, Fortino V. Network approaches for modeling the effect of drugs and diseases. Brief Bioinform 2022; 23:6608969. [PMID: 35704883 PMCID: PMC9294412 DOI: 10.1093/bib/bbac229] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/29/2022] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
The network approach is quickly becoming a fundamental building block of computational methods aiming at elucidating the mechanism of action (MoA) and therapeutic effect of drugs. By modeling the effect of drugs and diseases on different biological networks, it is possible to better explain the interplay between disease perturbations and drug targets as well as how drug compounds induce favorable biological responses and/or adverse effects. Omics technologies have been extensively used to generate the data needed to study the mechanisms of action of drugs and diseases. These data are often exploited to define condition-specific networks and to study whether drugs can reverse disease perturbations. In this review, we describe network data mining algorithms that are commonly used to study drug’s MoA and to improve our understanding of the basis of chronic diseases. These methods can support fundamental stages of the drug development process, including the identification of putative drug targets, the in silico screening of drug compounds and drug combinations for the treatment of diseases. We also discuss recent studies using biological and omics-driven networks to search for possible repurposed FDA-approved drug treatments for SARS-CoV-2 infections (COVID-19).
Collapse
Affiliation(s)
- T J Rintala
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Arindam Ghosh
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - V Fortino
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
38
|
Zong N, Li N, Wen A, Ngo V, Yu Y, Huang M, Chowdhury S, Jiang C, Fu S, Weinshilboum R, Jiang G, Hunter L, Liu H. BETA: a comprehensive benchmark for computational drug-target prediction. Brief Bioinform 2022; 23:6596989. [PMID: 35649342 PMCID: PMC9294420 DOI: 10.1093/bib/bbac199] [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: 01/14/2022] [Revised: 04/10/2022] [Accepted: 04/29/2022] [Indexed: 11/14/2022] Open
Abstract
Internal validation is the most popular evaluation strategy used for drug-target predictive models. The simple random shuffling in the cross-validation, however, is not always ideal to handle large, diverse and copious datasets as it could potentially introduce bias. Hence, these predictive models cannot be comprehensively evaluated to provide insight into their general performance on a variety of use-cases (e.g. permutations of different levels of connectiveness and categories in drug and target space, as well as validations based on different data sources). In this work, we introduce a benchmark, BETA, that aims to address this gap by (i) providing an extensive multipartite network consisting of 0.97 million biomedical concepts and 8.5 million associations, in addition to 62 million drug-drug and protein-protein similarities and (ii) presenting evaluation strategies that reflect seven cases (i.e. general, screening with different connectivity, target and drug screening based on categories, searching for specific drugs and targets and drug repurposing for specific diseases), a total of seven Tests (consisting of 344 Tasks in total) across multiple sampling and validation strategies. Six state-of-the-art methods covering two broad input data types (chemical structure- and gene sequence-based and network-based) were tested across all the developed Tasks. The best-worst performing cases have been analyzed to demonstrate the ability of the proposed benchmark to identify limitations of the tested methods for running over the benchmark tasks. The results highlight BETA as a benchmark in the selection of computational strategies for drug repurposing and target discovery.
Collapse
Affiliation(s)
- Nansu Zong
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Ning Li
- Center for Structure Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD
| | - Andrew Wen
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Victoria Ngo
- Betty Irene Moore School of Nursing, University of California Davis Health, Sacramento, CA.,Stanford Health Policy, Stanford School of Medicine and Freeman Spogli Institute for International Studies, Palo Alto, CA
| | - Yue Yu
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Ming Huang
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Shaika Chowdhury
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Chao Jiang
- Department of Computer Science and Software Engineering, Auburn University, Auburn, AL
| | - Sunyang Fu
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Guoqian Jiang
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| | - Lawrence Hunter
- Department of Pharmacology, University of Colorado Denver, Aurora, CO
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN
| |
Collapse
|
39
|
Hartung M, Anastasi E, Mamdouh ZM, Nogales C, Schmidt HHHW, Baumbach J, Zolotareva O, List M. Cancer driver drug interaction explorer. Nucleic Acids Res 2022; 50:W138-W144. [PMID: 35580047 PMCID: PMC9252786 DOI: 10.1093/nar/gkac384] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is a heterogeneous disease characterized by unregulated cell growth and promoted by mutations in cancer driver genes some of which encode suitable drug targets. Since the distinct set of cancer driver genes can vary between and within cancer types, evidence-based selection of drugs is crucial for targeted therapy following the precision medicine paradigm. However, many putative cancer driver genes can not be targeted directly, suggesting an indirect approach that considers alternative functionally related targets in the gene interaction network. Once potential drug targets have been identified, it is essential to consider all available drugs. Since tools that offer support for systematic discovery of drug repurposing candidates in oncology are lacking, we developed CADDIE, a web application integrating six human gene-gene and four drug-gene interaction databases, information regarding cancer driver genes, cancer-type specific mutation frequencies, gene expression information, genetically related diseases, and anticancer drugs. CADDIE offers access to various network algorithms for identifying drug targets and drug repurposing candidates. It guides users from the selection of seed genes to the identification of therapeutic targets or drug candidates, making network medicine algorithms accessible for clinical research. CADDIE is available at https://exbio.wzw.tum.de/caddie/ and programmatically via a python package at https://pypi.org/project/caddiepy/.
Collapse
Affiliation(s)
- Michael Hartung
- Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany
| | - Elisa Anastasi
- School of Computing, Newcastle University, 2308 Newcastle upon Tyne, UK
| | - Zeinab M Mamdouh
- Department of Pharmacology and Personalised Medicine, Maastricht University, 6229 Maastricht, Netherlands.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt
| | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, Maastricht University, 6229 Maastricht, Netherlands
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, Maastricht University, 6229 Maastricht, Netherlands
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany.,Computational Biomedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, 5230 Odense, Denmark
| | - Olga Zolotareva
- Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany.,Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
40
|
Zhou H, Ni WJ, Huang W, Wang Z, Cai M, Sun YC. Advances in Pathogenesis, Progression, Potential Targets and Targeted Therapeutic Strategies in SARS-CoV-2-Induced COVID-19. Front Immunol 2022; 13:834942. [PMID: 35450063 PMCID: PMC9016159 DOI: 10.3389/fimmu.2022.834942] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/07/2022] [Indexed: 01/18/2023] Open
Abstract
As the new year of 2020 approaches, an acute respiratory disease quietly caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as coronavirus disease 2019 (COVID-19) was reported in Wuhan, China. Subsequently, COVID-19 broke out on a global scale and formed a global public health emergency. To date, the destruction that has lasted for more than two years has not stopped and has caused the virus to continuously evolve new mutant strains. SARS-CoV-2 infection has been shown to cause multiple complications and lead to severe disability and death, which has dealt a heavy blow to global development, not only in the medical field but also in social security, economic development, global cooperation and communication. To date, studies on the epidemiology, pathogenic mechanism and pathological characteristics of SARS-CoV-2-induced COVID-19, as well as target confirmation, drug screening, and clinical intervention have achieved remarkable effects. With the continuous efforts of the WHO, governments of various countries, and scientific research and medical personnel, the public's awareness of COVID-19 is gradually deepening, a variety of prevention methods and detection methods have been implemented, and multiple vaccines and drugs have been developed and urgently marketed. However, these do not appear to have completely stopped the pandemic and ravages of this virus. Meanwhile, research on SARS-CoV-2-induced COVID-19 has also seen some twists and controversies, such as potential drugs and the role of vaccines. In view of the fact that research on SARS-CoV-2 and COVID-19 has been extensive and in depth, this review will systematically update the current understanding of the epidemiology, transmission mechanism, pathological features, potential targets, promising drugs and ongoing clinical trials, which will provide important references and new directions for SARS-CoV-2 and COVID-19 research.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei-Jian Ni
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Huang
- The Third People’s Hospital of Hefei, The Third Clinical College of Anhui Medical University, Hefei, China
| | - Zhen Wang
- Anhui Provincial Children’s Hospital, Children’s Hospital of Fudan University-Anhui Campus, Hefei, China
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yan-Cai Sun
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
41
|
Unmodified methodologies in target discovery for small molecule drugs: A rising star. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Barsi S, Papp H, Valdeolivas A, Tóth DJ, Kuczmog A, Madai M, Hunyady L, Várnai P, Saez-Rodriguez J, Jakab F, Szalai B. Computational drug repurposing against SARS-CoV-2 reveals plasma membrane cholesterol depletion as key factor of antiviral drug activity. PLoS Comput Biol 2022; 18:e1010021. [PMID: 35404937 PMCID: PMC9022874 DOI: 10.1371/journal.pcbi.1010021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/21/2022] [Accepted: 03/15/2022] [Indexed: 01/09/2023] Open
Abstract
Comparing SARS-CoV-2 infection-induced gene expression signatures to drug treatment-induced gene expression signatures is a promising bioinformatic tool to repurpose existing drugs against SARS-CoV-2. The general hypothesis of signature-based drug repurposing is that drugs with inverse similarity to a disease signature can reverse disease phenotype and thus be effective against it. However, in the case of viral infection diseases, like SARS-CoV-2, infected cells also activate adaptive, antiviral pathways, so that the relationship between effective drug and disease signature can be more ambiguous. To address this question, we analysed gene expression data from in vitro SARS-CoV-2 infected cell lines, and gene expression signatures of drugs showing anti-SARS-CoV-2 activity. Our extensive functional genomic analysis showed that both infection and treatment with in vitro effective drugs leads to activation of antiviral pathways like NFkB and JAK-STAT. Based on the similarity-and not inverse similarity-between drug and infection-induced gene expression signatures, we were able to predict the in vitro antiviral activity of drugs. We also identified SREBF1/2, key regulators of lipid metabolising enzymes, as the most activated transcription factors by several in vitro effective antiviral drugs. Using a fluorescently labeled cholesterol sensor, we showed that these drugs decrease the cholesterol levels of plasma-membrane. Supplementing drug-treated cells with cholesterol reversed the in vitro antiviral effect, suggesting the depleting plasma-membrane cholesterol plays a key role in virus inhibitory mechanism. Our results can help to more effectively repurpose approved drugs against SARS-CoV-2, and also highlights key mechanisms behind their antiviral effect.
Collapse
Affiliation(s)
- Szilvia Barsi
- Semmelweis University, Faculty of Medicine, Department of Physiology, Budapest, Hungary
| | - Henrietta Papp
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Alberto Valdeolivas
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Dániel J. Tóth
- Semmelweis University, Faculty of Medicine, Department of Physiology, Budapest, Hungary
| | - Anett Kuczmog
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Mónika Madai
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - László Hunyady
- Semmelweis University, Faculty of Medicine, Department of Physiology, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Péter Várnai
- Semmelweis University, Faculty of Medicine, Department of Physiology, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Budapest, Hungary
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Ferenc Jakab
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Bence Szalai
- Semmelweis University, Faculty of Medicine, Department of Physiology, Budapest, Hungary
| |
Collapse
|
43
|
Erdogan F, Radu TB, Orlova A, Qadree AK, de Araujo ED, Israelian J, Valent P, Mustjoki SM, Herling M, Moriggl R, Gunning PT. JAK-STAT core cancer pathway: An integrative cancer interactome analysis. J Cell Mol Med 2022; 26:2049-2062. [PMID: 35229974 PMCID: PMC8980946 DOI: 10.1111/jcmm.17228] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 12/25/2022] Open
Abstract
Through a comprehensive review and in silico analysis of reported data on STAT-linked diseases, we analysed the communication pathways and interactome of the seven STATs in major cancer categories and proposed rational targeting approaches for therapeutic intervention to disrupt critical pathways and addictions to hyperactive JAK/STAT in neoplastic states. Although all STATs follow a similar molecular activation pathway, STAT1, STAT2, STAT4 and STAT6 exert specific biological profiles associated with a more restricted pattern of activation by cytokines. STAT3 and STAT5A as well as STAT5B have pleiotropic roles in the body and can act as critical oncogenes that promote many processes involved in cancer development. STAT1, STAT3 and STAT5 also possess tumour suppressive action in certain mutational and cancer type context. Here, we demonstrated member-specific STAT activity in major cancer types. Through systems biology approaches, we found surprising roles for EGFR family members, sex steroid hormone receptor ESR1 interplay with oncogenic STAT function and proposed new drug targeting approaches of oncogenic STAT pathway addiction.
Collapse
Affiliation(s)
- Fettah Erdogan
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaOntarioCanada
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| | - Tudor Bogdan Radu
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaOntarioCanada
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| | - Anna Orlova
- Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Abdul Khawazak Qadree
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaOntarioCanada
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| | - Elvin Dominic de Araujo
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaOntarioCanada
| | - Johan Israelian
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaOntarioCanada
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| | - Peter Valent
- Division of Hematology and HemostaseologyDepartment of Internal Medicine IMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Satu M. Mustjoki
- Translational Immunology Research Program and Department of Clinical Chemistry and HematologyUniversity of HelsinkiHelsinkiFinland
- Hematology Research UnitHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Marco Herling
- Department of Hematology, Cellular Therapy, and HemostaseologyUniversity of LeipzigLeipzigGermany
| | - Richard Moriggl
- Institute of Animal Breeding and GeneticsUniversity of Veterinary MedicineViennaAustria
| | - Patrick Thomas Gunning
- Department of Chemical and Physical SciencesUniversity of Toronto MississaugaMississaugaOntarioCanada
- Department of ChemistryUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
44
|
Koliński M, Kałużna E, Piwecka M. RNA–protein interactomes as invaluable resources to study RNA viruses: Insights from SARS CoV‐2 studies. WIRES RNA 2022; 13:e1727. [PMID: 35343064 PMCID: PMC9111084 DOI: 10.1002/wrna.1727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/27/2022]
Abstract
Understanding the molecular mechanisms of severe respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection is essential for the successful development of therapeutic strategies against the COVID‐19 pandemic. Numerous studies have focused on the identification of host factors and cellular pathways involved in the viral replication cycle. The speed and magnitude of hijacking the translation machinery of host mRNA, and shutting down host transcription are still not well understood. Since SARS‐CoV‐2 relies on host RNA‐binding proteins for the infection progression, several efforts have been made to define the SARS‐CoV‐2 RNA‐bound proteomes (RNA–protein interactomes). Methodologies that enable the systemic capture of protein interactors of given RNA in vivo have been adapted for the identification of the SARS‐CoV‐2 RNA interactome. The obtained proteomic data aided by genome‐wide and targeted CRISPR perturbation screens, revealed host factors with either pro‐ or anti‐viral activity and highlighted cellular processes and factors involved in host response. We focus here on the recent studies on SARS‐CoV‐2 RNA–protein interactomes, with regard to both the technological aspects of RNA interactome capture methods and the obtained results. We also summarize several related studies, which were used in the interpretation of the SARS‐CoV‐2 RNA–protein interactomes. These studies provided the selection of host factors that are potentially suitable candidates for antiviral therapy. Finally, we underscore the importance of RNA–protein interactome studies in regard to the effective development of antiviral strategies against current and future threats. This article is categorized under:RNA Interactions with Proteins and Other Molecules > Protein‐RNA Interactions: Functional Implications RNA in Disease and Development > RNA in Disease RNA Methods > RNA Analyses in Cells
Collapse
Affiliation(s)
- Marcin Koliński
- Department of Non‐Coding RNAs Institute of Bioorganic Chemistry, Polish Academy of Sciences Poznan Poland
| | - Ewelina Kałużna
- Department of Non‐Coding RNAs Institute of Bioorganic Chemistry, Polish Academy of Sciences Poznan Poland
| | - Monika Piwecka
- Department of Non‐Coding RNAs Institute of Bioorganic Chemistry, Polish Academy of Sciences Poznan Poland
| |
Collapse
|
45
|
Bernett J, Krupke D, Sadegh S, Baumbach J, Fekete SP, Kacprowski T, List M, Blumenthal DB. Robust disease module mining via enumeration of diverse prize-collecting Steiner trees. Bioinformatics 2022; 38:1600-1606. [PMID: 34984440 DOI: 10.1093/bioinformatics/btab876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/29/2021] [Accepted: 12/31/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Disease module mining methods (DMMMs) extract subgraphs that constitute candidate disease mechanisms from molecular interaction networks such as protein-protein interaction (PPI) networks. Irrespective of the employed models, DMMMs typically include non-robust steps in their workflows, i.e. the computed subnetworks vary when running the DMMMs multiple times on equivalent input. This lack of robustness has a negative effect on the trustworthiness of the obtained subnetworks and is hence detrimental for the widespread adoption of DMMMs in the biomedical sciences. RESULTS To overcome this problem, we present a new DMMM called ROBUST (robust disease module mining via enumeration of diverse prize-collecting Steiner trees). In a large-scale empirical evaluation, we show that ROBUST outperforms competing methods in terms of robustness, scalability and, in most settings, functional relevance of the produced modules, measured via KEGG (Kyoto Encyclopedia of Genes and Genomes) gene set enrichment scores and overlap with DisGeNET disease genes. AVAILABILITY AND IMPLEMENTATION A Python 3 implementation and scripts to reproduce the results reported in this article are available on GitHub: https://github.com/bionetslab/robust, https://github.com/bionetslab/robust-eval. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Judith Bernett
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Dominik Krupke
- Department of Computer Science, TU Braunschweig, 38106 Braunschweig, Germany
| | - Sepideh Sadegh
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany.,Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany.,Department of Mathematics and Computer Science, University of Southern Denmark, 5230 Odense, Denmark
| | - Sándor P Fekete
- Department of Computer Science, TU Braunschweig, 38106 Braunschweig, Germany.,Braunschweig Integrated Centre of Systems Biology (BRICS), 38106 Braunschweig, Germany
| | - Tim Kacprowski
- Braunschweig Integrated Centre of Systems Biology (BRICS), 38106 Braunschweig, Germany.,Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technical University of Braunschweig and Hannover Medical School, 38106 Braunschweig, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - David B Blumenthal
- Department Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| |
Collapse
|
46
|
Tian X, Shen L, Gao P, Huang L, Liu G, Zhou L, Peng L. Discovery of Potential Therapeutic Drugs for COVID-19 Through Logistic Matrix Factorization With Kernel Diffusion. Front Microbiol 2022; 13:740382. [PMID: 35295301 PMCID: PMC8919055 DOI: 10.3389/fmicb.2022.740382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 02/01/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is rapidly spreading. Researchers around the world are dedicated to finding the treatment clues for COVID-19. Drug repositioning, as a rapid and cost-effective way for finding therapeutic options from available FDA-approved drugs, has been applied to drug discovery for COVID-19. In this study, we develop a novel drug repositioning method (VDA-KLMF) to prioritize possible anti-SARS-CoV-2 drugs integrating virus sequences, drug chemical structures, known Virus-Drug Associations, and Logistic Matrix Factorization with Kernel diffusion. First, Gaussian kernels of viruses and drugs are built based on known VDAs and nearest neighbors. Second, sequence similarity kernel of viruses and chemical structure similarity kernel of drugs are constructed based on biological features and an identity matrix. Third, Gaussian kernel and similarity kernel are diffused. Forth, a logistic matrix factorization model with kernel diffusion is proposed to identify potential anti-SARS-CoV-2 drugs. Finally, molecular dockings between the inferred antiviral drugs and the junction of SARS-CoV-2 spike protein-ACE2 interface are implemented to investigate the binding abilities between them. VDA-KLMF is compared with two state-of-the-art VDA prediction models (VDA-KATZ and VDA-RWR) and three classical association prediction methods (NGRHMDA, LRLSHMDA, and NRLMF) based on 5-fold cross validations on viruses, drugs, and VDAs on three datasets. It obtains the best recalls, AUCs, and AUPRs, significantly outperforming other five methods under the three different cross validations. We observe that four chemical agents coming together on any two datasets, that is, remdesivir, ribavirin, nitazoxanide, and emetine, may be the clues of treatment for COVID-19. The docking results suggest that the key residues K353 and G496 may affect the binding energies and dynamics between the inferred anti-SARS-CoV-2 chemical agents and the junction of the spike protein-ACE2 interface. Integrating various biological data, Gaussian kernel, similarity kernel, and logistic matrix factorization with kernel diffusion, this work demonstrates that a few chemical agents may assist in drug discovery for COVID-19.
Collapse
Affiliation(s)
- Xiongfei Tian
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Ling Shen
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Pengfei Gao
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, China
| | - Li Huang
- Academy of Arts and Design, Tsinghua University, Beijing, China
- The Future Laboratory, Tsinghua University, Beijing, China
| | - Guangyi Liu
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
- *Correspondence: Liqian Zhou,
| | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, China
- Lihong Peng,
| |
Collapse
|
47
|
CTCFL regulates the PI3K-Akt pathway and it is a target for personalized ovarian cancer therapy. NPJ Syst Biol Appl 2022; 8:5. [PMID: 35132075 PMCID: PMC8821627 DOI: 10.1038/s41540-022-00214-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 01/05/2022] [Indexed: 12/04/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSC) is the most lethal gynecologic malignancy due to the lack of reliable biomarkers, effective treatment, and chemoresistance. Improving the diagnosis and the development of targeted therapies is still needed. The molecular pathomechanisms driving HGSC progression are not fully understood though crucial for effective diagnosis and identification of novel targeted therapy options. The oncogene CTCFL (BORIS), the paralog of CTCF, is a transcriptional factor highly expressed in ovarian cancer (but in rarely any other tissue in females) with cancer-specific characteristics and therapeutic potential. In this work, we seek to understand the regulatory functions of CTCFL to unravel new target genes with clinical relevance. We used in vitro models to evaluate the transcriptional changes due to the presence of CTCFL, followed by a selection of gene candidates using de novo network enrichment analysis. The resulting mechanistic candidates were further assessed regarding their prognostic potential and druggability. We show that CTCFL-driven genes are involved in cytoplasmic membrane functions; in particular, the PI3K-Akt initiators EGFR1 and VEGFA, as well as ITGB3 and ITGB6 are potential drug targets. Finally, we identified the CTCFL targets ACTBL2, MALT1 and PCDH7 as mechanistic biomarkers to predict survival in HGSC. Finally, we elucidated the value of CTCFL in combination with its targets as a prognostic marker profile for HGSC progression and as putative drug targets.
Collapse
|
48
|
Guo Y, Esfahani F, Shao X, Srinivasan V, Thomo A, Xing L, Zhang X. Integrative COVID-19 biological network inference with probabilistic core decomposition. Brief Bioinform 2022; 23:6425808. [PMID: 34791019 PMCID: PMC8689992 DOI: 10.1093/bib/bbab455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/15/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for millions of deaths around the world. To help contribute to the understanding of crucial knowledge and to further generate new hypotheses relevant to SARS-CoV-2 and human protein interactions, we make use of the information abundant Biomine probabilistic database and extend the experimentally identified SARS-CoV-2-human protein-protein interaction (PPI) network in silico. We generate an extended network by integrating information from the Biomine database, the PPI network and other experimentally validated results. To generate novel hypotheses, we focus on the high-connectivity sub-communities that overlap most with the integrated experimentally validated results in the extended network. Therefore, we propose a new data analysis pipeline that can efficiently compute core decomposition on the extended network and identify dense subgraphs. We then evaluate the identified dense subgraph and the generated hypotheses in three contexts: literature validation for uncovered virus targeting genes and proteins, gene function enrichment analysis on subgraphs and literature support on drug repurposing for identified tissues and diseases related to COVID-19. The major types of the generated hypotheses are proteins with their encoding genes and we rank them by sorting their connections to the integrated experimentally validated nodes. In addition, we compile a comprehensive list of novel genes, and proteins potentially related to COVID-19, as well as novel diseases which might be comorbidities. Together with the generated hypotheses, our results provide novel knowledge relevant to COVID-19 for further validation.
Collapse
Affiliation(s)
- Yang Guo
- Department of Mathematics and Statistics, University of Victoria, 3800 Finnerty Road, V8P 5C2, Victoria, BC, Canada
| | - Fatemeh Esfahani
- Department of Computer Science, University of Victoria, 3800 Finnerty Road, V8P 5C2, Victoria, BC, Canada
| | - Xiaojian Shao
- Digital Technologies Research Centre, National Research Council Canada, 1200 Montreal Road, K1A 0R6, Ottawa, ON, Canada
| | - Venkatesh Srinivasan
- Department of Computer Science, University of Victoria, 3800 Finnerty Road, V8P 5C2, Victoria, BC, Canada
| | - Alex Thomo
- Department of Computer Science, University of Victoria, 3800 Finnerty Road, V8P 5C2, Victoria, BC, Canada
| | - Li Xing
- Department of Mathematics and Statistics, University of Saskatchewan, 110 Science Place, S7N 5A2, Saskatoon, SK, Canada
| | - Xuekui Zhang
- Corresponding author: Xuekui Zhang, Department of Mathematics and Statistics, University of Victoria, 3800 Finnerty Road, V8P 5C2, Victoria, BC, Canada.
| |
Collapse
|
49
|
Wang H, Zhang J, Lu Z, Dai W, Ma C, Xiang Y, Zhang Y. Identification of potential therapeutic targets and mechanisms of COVID-19 through network analysis and screening of chemicals and herbal ingredients. Brief Bioinform 2022; 23:bbab373. [PMID: 34505138 PMCID: PMC8499921 DOI: 10.1093/bib/bbab373] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
After experiencing the COVID-19 pandemic, it is widely acknowledged that a rapid drug repurposing method is highly needed. A series of useful drug repurposing tools have been developed based on data-driven modeling and network pharmacology. Based on the disease module, we identified several hub proteins that play important roles in the onset and development of the COVID-19, which are potential targets for repositioning approved drugs. Moreover, different network distance metrics were applied to quantify the relationship between drug targets and COVID-19 disease targets in the protein-protein-interaction (PPI) network and predict COVID-19 therapeutic effects of bioactive herbal ingredients and chemicals. Furthermore, the tentative mechanisms of candidates were illustrated through molecular docking and gene enrichment analysis. We obtained 15 chemical and 15 herbal ingredient candidates and found that different drugs may play different roles in the process of virus invasion and the onset and development of the COVID-19 disease. Given pandemic outbreaks, our method has an undeniable immense advantage in the feasibility analysis of drug repurposing or drug screening, especially in the analysis of herbal ingredients.
Collapse
Affiliation(s)
- Hong Wang
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Medical Data Science Academy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Engineering Research Center for Clinical Big-data and Drug Evaluation, Chongqing Medical University, Chongqing, 401331, China
| | - Jingqing Zhang
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Zhigang Lu
- Department of Neurology, The First People's Hospital of Jingmen affiliated to Hubei Minzu University, Jingmen, 448000, China
| | - Weina Dai
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Chuanjiang Ma
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yun Xiang
- Gynaecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
| | - Yonghong Zhang
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Medical Data Science Academy, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Engineering Research Center for Clinical Big-data and Drug Evaluation, Chongqing Medical University, Chongqing, 401331, China
| |
Collapse
|
50
|
Santos SDS, Torres M, Galeano D, Sánchez MDM, Cernuzzi L, Paccanaro A. Machine learning and network medicine approaches for drug repositioning for COVID-19. PATTERNS (NEW YORK, N.Y.) 2022; 3:100396. [PMID: 34778851 PMCID: PMC8576113 DOI: 10.1016/j.patter.2021.100396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/21/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022]
Abstract
We present two machine learning approaches for drug repurposing. While we have developed them for COVID-19, they are disease-agnostic. The two methodologies are complementary, targeting SARS-CoV-2 and host factors, respectively. Our first approach consists of a matrix factorization algorithm to rank broad-spectrum antivirals. Our second approach, based on network medicine, uses graph kernels to rank drugs according to the perturbation they induce on a subnetwork of the human interactome that is crucial for SARS-CoV-2 infection/replication. Our experiments show that our top predicted broad-spectrum antivirals include drugs indicated for compassionate use in COVID-19 patients; and that the ranking obtained by our kernel-based approach aligns with experimental data. Finally, we present the COVID-19 repositioning explorer (CoREx), an interactive online tool to explore the interplay between drugs and SARS-CoV-2 host proteins in the context of biological networks, protein function, drug clinical use, and Connectivity Map. CoREx is freely available at: https://paccanarolab.org/corex/.
Collapse
Affiliation(s)
- Suzana de Siqueira Santos
- Escola de Matemática Aplicada, Fundação Getulio Vargas, Rio de Janeiro 22250-900, Brazil
- COVID-19 International Research Team
| | - Mateo Torres
- Escola de Matemática Aplicada, Fundação Getulio Vargas, Rio de Janeiro 22250-900, Brazil
- COVID-19 International Research Team
| | - Diego Galeano
- Escola de Matemática Aplicada, Fundação Getulio Vargas, Rio de Janeiro 22250-900, Brazil
- Facultad de Ingenieria, Universidad Nacional de Asunción, Luque 110948, Paraguay
- COVID-19 International Research Team
| | | | - Luca Cernuzzi
- Universidad Católica “Nuestra Señora de la Asunción”, Asunción C.C. 1683, Paraguay
| | - Alberto Paccanaro
- Escola de Matemática Aplicada, Fundação Getulio Vargas, Rio de Janeiro 22250-900, Brazil
- Department of Computer Science, Centre for Systems and Synthetic Biology, Royal Holloway, University of London, Egham Hill, Egham TW20 0EX, UK
- COVID-19 International Research Team
| |
Collapse
|