51
|
Yu W, Bai Y, Raha A, Su Z, Geng F. Integrative In Silico Investigation Reveals the Host-Virus Interactions in Repurposed Drugs Against SARS-CoV-2. FRONTIERS IN BIOINFORMATICS 2022; 1:763540. [PMID: 36303774 PMCID: PMC9580895 DOI: 10.3389/fbinf.2021.763540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
The ongoing COVID-19 outbreak have posed a significant threat to public health worldwide. Recently Toll-like receptor (TLR) has been proposed to be the drug target of SARS-CoV-2 treatment, the specificity and efficacy of such treatments remain unknown. In the present study we performed the investigation of repurposed drugs via a framework comprising of Search Tool for Interacting Chemicals (STITCH), Kyoto Encyclopedia of Genes and Genomes (KEGG), molecular docking, and virus-host-drug interactome mapping. Chloroquine (CQ) and hydroxychloroquine (HCQ) were utilized as probes to explore the interaction network that is linked to SARS-CoV-2. 47 drug targets were shown to be overlapped with SARS-CoV-2 network and were enriched in TLR signaling pathway. Molecular docking analysis and molecular dynamics simulation determined the direct binding affinity of TLR9 to CQ and HCQ. Furthermore, we established SARS-CoV-2-human-drug protein interaction map and identified the axis of TLR9-ERC1-Nsp13 and TLR9-RIPK1-Nsp12. Therefore, the elucidation of the interactions of SARS-CoV-2 with TLR9 axis will not only provide pivotal insights into SARS-CoV-2 infection and pathogenesis but also improve the treatment against COVID-19.
Collapse
Affiliation(s)
- Wenhui Yu
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Yuxin Bai
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Arjun Raha
- Department of Mechanical Engineering, McMaster University, Hamilton, ON, Canada
| | - Zhi Su
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON, Canada
| | - Fei Geng
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON, Canada
- *Correspondence: Fei Geng,
| |
Collapse
|
52
|
Zhang Q, Gao J, Wu JT, Cao Z, Dajun Zeng D. Data science approaches to confronting the COVID-19 pandemic: a narrative review. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2022; 380:20210127. [PMID: 34802267 PMCID: PMC8607150 DOI: 10.1098/rsta.2021.0127] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/22/2021] [Indexed: 05/07/2023]
Abstract
During the COVID-19 pandemic, more than ever, data science has become a powerful weapon in combating an infectious disease epidemic and arguably any future infectious disease epidemic. Computer scientists, data scientists, physicists and mathematicians have joined public health professionals and virologists to confront the largest pandemic in the century by capitalizing on the large-scale 'big data' generated and harnessed for combating the COVID-19 pandemic. In this paper, we review the newly born data science approaches to confronting COVID-19, including the estimation of epidemiological parameters, digital contact tracing, diagnosis, policy-making, resource allocation, risk assessment, mental health surveillance, social media analytics, drug repurposing and drug development. We compare the new approaches with conventional epidemiological studies, discuss lessons we learned from the COVID-19 pandemic, and highlight opportunities and challenges of data science approaches to confronting future infectious disease epidemics. This article is part of the theme issue 'Data science approaches to infectious disease surveillance'.
Collapse
Affiliation(s)
- Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Hong Kong
| | - Jianxi Gao
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Joseph T. Wu
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Zhidong Cao
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, People’s Republic of China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100190, People’s Republic of China
| | - Daniel Dajun Zeng
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, People’s Republic of China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100190, People’s Republic of China
| |
Collapse
|
53
|
Luedemann M, Stadler D, Cheng CC, Protzer U, Knolle PA, Donakonda S. Montelukast is a dual-purpose inhibitor of SARS-CoV-2 infection and virus-induced IL-6 expression identified by structure-based drug repurposing. Comput Struct Biotechnol J 2022; 20:799-811. [PMID: 35126884 PMCID: PMC8800171 DOI: 10.1016/j.csbj.2022.01.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/22/2022] [Accepted: 01/23/2022] [Indexed: 12/13/2022] Open
Abstract
Drug-repurposing has been instrumental to identify drugs preventing SARS-CoV-2 replication or attenuating the disease course of COVID-19. Here, we identify through structure-based drug-repurposing a dual-purpose inhibitor of SARS-CoV-2 infection and of IL-6 production by immune cells. We created a computational structure model of the receptor binding domain (RBD) of the SARS-CoV-2 spike 1 protein, and used this model for insilico screening against a library of 6171 molecularly defined binding-sites from drug molecules. Molecular dynamics simulation of candidate molecules with high RBD binding-scores in docking analysis predicted montelukast, an antagonist of the cysteinyl-leukotriene-receptor, to disturb the RBD structure, and infection experiments demonstrated inhibition of SARS-CoV-2 infection, although montelukast binding was outside the ACE2-binding site. Molecular dynamics simulation of SARS-CoV-2 variant RBDs correctly predicted interference of montelukast with infection by the beta but not the more infectious alpha variant. With distinct binding sites for RBD and the leukotriene receptor, montelukast also prevented SARS-CoV-2-induced IL-6 release from immune cells. The inhibition of SARS-CoV-2 infection through a molecule binding distal to the ACE-binding site of the RBD points towards an allosteric mechanism that is not conserved in the more infectious alpha and delta SARS-CoV-2 variants.
Collapse
|
54
|
Computational approaches for drug repositioning and repurposing to combat SARS-CoV-2 infection. COMPUTATIONAL APPROACHES FOR NOVEL THERAPEUTIC AND DIAGNOSTIC DESIGNING TO MITIGATE SARS-COV-2 INFECTION 2022:247-265. [PMCID: PMC9300474 DOI: 10.1016/b978-0-323-91172-6.00008-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Drug repositioning (also referred to as drug repurposing) is the method of exploring novel therapeutic indications for Food and Drug Administration-approved clinically implemented drugs. The unique strategy of drug repositioning is used to boost the drug development process since drug discovery is an expensive, arduous, cumbersome, and high-risk procedure. Recently, several pharmaceutical firms have used the drug repositioning technique in their drug discovery and development programs to develop new medications based on the identification of new therapeutic targets. This technique is extremely effective, saves time, is comparatively economical, and has a low chance of failure. Developing appropriate treatment measures to inhibit the spread of Coronavirus disease-2019 (COVID-19) is currently a top priority. As a result, several studies were conducted to build novel therapeutic molecules using diverse strategies of drug repurposing to discover drug candidates against COVID-19 infection that can act as substantial inhibitors against virus particles. By implementing virtual screening of drug libraries, it is possible to identify potential drugs through drug repurposing. A molecular docking approach and calculation of binding free energy are used to estimate binding affinity and drug–receptor interactions. Drug-repurposing methodologies can be divided into three categories: target-oriented, drug-oriented, and disease-oriented, based on the gathered data about the various physicochemical, pharmacokinetic and pharmacological features of a drug candidate. Using computational methods such as homology modeling and molecular similarity, this methodology aids in determining the binding interaction of drug molecules with the target protein of the virus. In this book chapter, we explore a typical set of currently utilized computational techniques for identifying repurposable drug molecules for COVID-19, as well as their supporting databases. We also assess promising drugs anticipated by computational approaches to drugs currently being evaluated in clinical trials. Moreover, we also examine the takeaways from the evaluated research efforts, such as how to competently combine bioinformatics tools with experimental work and suggest a fully integrated drug-repurposing approach to combat the deadly COVID-19 infection.
Collapse
|
55
|
Middha SK, David A, Haldar S, Boro H, Panda P, Bajare N, Milesh L, Devaraj V, Usha T. Databases, DrugBank, and virtual screening platforms for therapeutic development. COMPUTATIONAL APPROACHES FOR NOVEL THERAPEUTIC AND DIAGNOSTIC DESIGNING TO MITIGATE SARS-COV-2 INFECTION 2022. [PMCID: PMC9300480 DOI: 10.1016/b978-0-323-91172-6.00021-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The upsurge of the severe acute respiratory syndrome-Coronavirus-2 (SARS-CoV-2) has turned into a global health disaster. Many remodeled medications were suggested for treatment in the early stages of this pandemic, but these dosages afterward came across with distinct offshoots. Thus, these consequences compelled the scientists to develop new drugs using various antiviral, antiinflammatory, antibacterial, and phytochemical compounds. A handful of drugs have been scrutinized in silico, in vitro, plus through human trials such as anti-SARS-CoV-2 agents and made available as various databases by various scientific communities. The SARS-CoV-2 pandemic databases are designed to allay difficulties associated with this scenario. Some of the popular databases are GESS (global evaluation of SARS-CoV-2/HCoV-19 sequences) which gives a thorough study of data based on tenfold of thousands of complete coverage and quality of SARS-CoV-2 genomes, CORona Drug InTERactions (CORDITE) database for SARS-CoV-2 which profoundly combines the understanding of potential drugs and make it available for scientists and medicos. SARSCOVIDB set one’s sights to merge all differential gene expression data, at mRNA and protein levels, helping to accelerate analysis and research on the molecular impact of covid-19. This chapter aims to provide a piece of complete information about the SARS-CoV-2 virus databases, potentially available drugs, and virtual screening methods. And also provides a different webserver to reach out for information related to the COVID-19 pandemic and its future.
Collapse
|
56
|
Law JN, Akers K, Tasnina N, Santina CMD, Deutsch S, Kshirsagar M, Klein-Seetharaman J, Crovella M, Rajagopalan P, Kasif S, Murali TM. Interpretable network propagation with application to expanding the repertoire of human proteins that interact with SARS-CoV-2. Gigascience 2021; 10:giab082. [PMID: 34966926 PMCID: PMC8716363 DOI: 10.1093/gigascience/giab082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/21/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Network propagation has been widely used for nearly 20 years to predict gene functions and phenotypes. Despite the popularity of this approach, little attention has been paid to the question of provenance tracing in this context, e.g., determining how much any experimental observation in the input contributes to the score of every prediction. RESULTS We design a network propagation framework with 2 novel components and apply it to predict human proteins that directly or indirectly interact with SARS-CoV-2 proteins. First, we trace the provenance of each prediction to its experimentally validated sources, which in our case are human proteins experimentally determined to interact with viral proteins. Second, we design a technique that helps to reduce the manual adjustment of parameters by users. We find that for every top-ranking prediction, the highest contribution to its score arises from a direct neighbor in a human protein-protein interaction network. We further analyze these results to develop functional insights on SARS-CoV-2 that expand on known biology such as the connection between endoplasmic reticulum stress, HSPA5, and anti-clotting agents. CONCLUSIONS We examine how our provenance-tracing method can be generalized to a broad class of network-based algorithms. We provide a useful resource for the SARS-CoV-2 community that implicates many previously undocumented proteins with putative functional relationships to viral infection. This resource includes potential drugs that can be opportunistically repositioned to target these proteins. We also discuss how our overall framework can be extended to other, newly emerging viruses.
Collapse
Affiliation(s)
- Jeffrey N Law
- Interdisciplinary Ph.D. Program in Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Kyle Akers
- Interdisciplinary Ph.D. Program in Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Nure Tasnina
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, USA
| | | | - Shay Deutsch
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
| | | | | | - Mark Crovella
- Department of Computer Science, Boston University, Boston, MA 02215, USA
| | | | - Simon Kasif
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
57
|
Jukič M, Kores K, Janežič D, Bren U. Repurposing of Drugs for SARS-CoV-2 Using Inverse Docking Fingerprints. Front Chem 2021; 9:757826. [PMID: 35028304 PMCID: PMC8748264 DOI: 10.3389/fchem.2021.757826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 or SARS-CoV-2 is a virus that belongs to the Coronaviridae family. This group of viruses commonly causes colds but possesses a tremendous pathogenic potential. In humans, an outbreak of SARS caused by the SARS-CoV virus was first reported in 2003, followed by 2012 when the Middle East respiratory syndrome coronavirus (MERS-CoV) led to an outbreak of Middle East respiratory syndrome (MERS). Moreover, COVID-19 represents a serious socioeconomic and global health problem that has already claimed more than four million lives. To date, there are only a handful of therapeutic options to combat this disease, and only a single direct-acting antiviral, the conditionally approved remdesivir. Since there is an urgent need for active drugs against SARS-CoV-2, the strategy of drug repurposing represents one of the fastest ways to achieve this goal. An in silico drug repurposing study using two methods was conducted. A structure-based virtual screening of the FDA-approved drug database on SARS-CoV-2 main protease was performed, and the 11 highest-scoring compounds with known 3CLpro activity were identified while the methodology was used to report further 11 potential and completely novel 3CLpro inhibitors. Then, inverse molecular docking was performed on the entire viral protein database as well as on the Coronaviridae family protein subset to examine the hit compounds in detail. Instead of target fishing, inverse docking fingerprints were generated for each hit compound as well as for the five most frequently reported and direct-acting repurposed drugs that served as controls. In this way, the target-hitting space was examined and compared and we can support the further biological evaluation of all 11 newly reported hits on SARS-CoV-2 3CLpro as well as recommend further in-depth studies on antihelminthic class member compounds. The authors acknowledge the general usefulness of this approach for a full-fledged inverse docking fingerprint screening in the future.
Collapse
Affiliation(s)
- Marko Jukič
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Koper, Slovenia
| | - Katarina Kores
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Dušanka Janežič
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Koper, Slovenia
| | - Urban Bren
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Koper, Slovenia
| |
Collapse
|
58
|
Bradfute SB. The discovery and development of novel treatment strategies for filoviruses. Expert Opin Drug Discov 2021; 17:139-149. [PMID: 34962451 DOI: 10.1080/17460441.2022.2013800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Filoviruses are negative-stranded, enveloped RNA viruses that can cause hemorrhagic fever in humans and include Ebola and Marburg viruses. Lethality rates can reach 90% in isolated outbreaks. The 2013-2016 Ebola virus epidemic demonstrated the global threat of filoviruses and hastened development of vaccines and therapeutics. There are six known filoviruses that cause disease in humans, but still few therapeutics are available for treatment. AREAS COVERED This review summarizes identification, testing, and development of therapeutics based on the peer-reviewed scientific literature beginning with the discovery of filoviruses in 1967. Small molecules, antibodies, cytokines, antisense, post-exposure vaccination, and host-targeted therapeutic approaches are discussed. An emphasis is placed on therapeutics that have shown promise in in vivo studies. EXPERT OPINION Two monoclonal antibody regimens are approved for use in humans for one filovirus (Ebola virus), and preclinical nonhuman primate studies suggest that other monoclonal-based therapies are likely to be effective against other filoviruses. Significant progress has been made in small-molecule antivirals and host-targeted approaches. An important consideration is the necessity of pan-filovirus therapeutics via broadly effective small molecules, antibody cocktails, and cross-reactive antibodies. The use of filovirus therapeutics as prophylactic treatment or in chronically infected individuals should be considered.
Collapse
Affiliation(s)
- Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, USA
| |
Collapse
|
59
|
Rando HM, Wellhausen N, Ghosh S, Lee AJ, Dattoli AA, Hu F, Byrd JB, Rafizadeh DN, Lordan R, Qi Y, Sun Y, Brueffer C, Field JM, Ben Guebila M, Jadavji NM, Skelly AN, Ramsundar B, Wang J, Goel RR, Park Y, COVID-19 Review Consortium
BansalVikasBartonJohn P.BocaSimina M.BoerckelJoel D.BruefferChristianByrdJames BrianCaponeStephenDasShiktaDattoliAnna AdaDziakJohn J.FieldJeffrey M.GhoshSoumitaGitterAnthonyGoelRishi RajGreeneCasey S.GuebilaMarouen BenHimmelsteinDaniel S.HuFenglingJadavjiNafisa M.KamilJeremy P.KnyazevSergeyKollaLikhithaLeeAlexandra J.LordanRonanLubianaTiagoLukanTemitayoMacLeanAdam L.MaiDavidMangulSergheiManheimDavidMcGowanLucy D’AgostinoNaikAmrutaParkYoSonPerrinDimitriQiYanjunRafizadehDiane N.RamsundarBharathRandoHalie M.RaySandipanRobsonMichael P.RubinettiVincentSellElizabethShinholsterLamonicaSkellyAshwin N.SunYuchenSunYushaSzetoGregory L.VelazquezRyanWangJinhuiWellhausenNils, Boca SM, Gitter A, Greene CS. Identification and Development of Therapeutics for COVID-19. mSystems 2021; 6:e0023321. [PMID: 34726496 PMCID: PMC8562484 DOI: 10.1128/msystems.00233-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
After emerging in China in late 2019, the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spread worldwide, and as of mid-2021, it remains a significant threat globally. Only a few coronaviruses are known to infect humans, and only two cause infections similar in severity to SARS-CoV-2: Severe acute respiratory syndrome-related coronavirus, a species closely related to SARS-CoV-2 that emerged in 2002, and Middle East respiratory syndrome-related coronavirus, which emerged in 2012. Unlike the current pandemic, previous epidemics were controlled rapidly through public health measures, but the body of research investigating severe acute respiratory syndrome and Middle East respiratory syndrome has proven valuable for identifying approaches to treating and preventing novel coronavirus disease 2019 (COVID-19). Building on this research, the medical and scientific communities have responded rapidly to the COVID-19 crisis and identified many candidate therapeutics. The approaches used to identify candidates fall into four main categories: adaptation of clinical approaches to diseases with related pathologies, adaptation based on virological properties, adaptation based on host response, and data-driven identification (ID) of candidates based on physical properties or on pharmacological compendia. To date, a small number of therapeutics have already been authorized by regulatory agencies such as the Food and Drug Administration (FDA), while most remain under investigation. The scale of the COVID-19 crisis offers a rare opportunity to collect data on the effects of candidate therapeutics. This information provides insight not only into the management of coronavirus diseases but also into the relative success of different approaches to identifying candidate therapeutics against an emerging disease. IMPORTANCE The COVID-19 pandemic is a rapidly evolving crisis. With the worldwide scientific community shifting focus onto the SARS-CoV-2 virus and COVID-19, a large number of possible pharmaceutical approaches for treatment and prevention have been proposed. What was known about each of these potential interventions evolved rapidly throughout 2020 and 2021. This fast-paced area of research provides important insight into how the ongoing pandemic can be managed and also demonstrates the power of interdisciplinary collaboration to rapidly understand a virus and match its characteristics with existing or novel pharmaceuticals. As illustrated by the continued threat of viral epidemics during the current millennium, a rapid and strategic response to emerging viral threats can save lives. In this review, we explore how different modes of identifying candidate therapeutics have borne out during COVID-19.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Soumita Ghosh
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Ada Dattoli
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fengling Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Brian Byrd
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Diane N. Rafizadeh
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yanjun Qi
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | - Yuchen Sun
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | | | - Jeffrey M. Field
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Nafisa M. Jadavji
- Biomedical Science, Midwestern University, Glendale, Arizona, USA
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Ashwin N. Skelly
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Jinhui Wang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rishi Raj Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - YoSon Park
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - COVID-19 Review Consortium
BansalVikasBartonJohn P.BocaSimina M.BoerckelJoel D.BruefferChristianByrdJames BrianCaponeStephenDasShiktaDattoliAnna AdaDziakJohn J.FieldJeffrey M.GhoshSoumitaGitterAnthonyGoelRishi RajGreeneCasey S.GuebilaMarouen BenHimmelsteinDaniel S.HuFenglingJadavjiNafisa M.KamilJeremy P.KnyazevSergeyKollaLikhithaLeeAlexandra J.LordanRonanLubianaTiagoLukanTemitayoMacLeanAdam L.MaiDavidMangulSergheiManheimDavidMcGowanLucy D’AgostinoNaikAmrutaParkYoSonPerrinDimitriQiYanjunRafizadehDiane N.RamsundarBharathRandoHalie M.RaySandipanRobsonMichael P.RubinettiVincentSellElizabethShinholsterLamonicaSkellyAshwin N.SunYuchenSunYushaSzetoGregory L.VelazquezRyanWangJinhuiWellhausenNils
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
- Biomedical Science, Midwestern University, Glendale, Arizona, USA
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- The DeepChem Project
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, R & D, AstraZeneca, Gaithersburg, Maryland, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Simina M. Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, R & D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
60
|
Mediterranean Diet a Potential Strategy against SARS-CoV-2 Infection: A Narrative Review. Medicina (B Aires) 2021; 57:medicina57121389. [PMID: 34946334 PMCID: PMC8704657 DOI: 10.3390/medicina57121389] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 01/08/2023] Open
Abstract
Mediterranean Diet represents the traditional eating habits of populations living around the Mediterranean Sea, and it is associated with a lower risk of overall mortality and cancer incidence and cardiovascular diseases. Severe acute respiratory syndrome coronavirus 2 is a new pandemic, and represents a significant and critical threat to global human health. In this study, we aimed to review the possible effects of Mediterranean Diet against the risk of the coronavirus disease 2019. Several vitamins, minerals, fatty acids, and phytochemicals with their potential anti-COVID-19 activity are presented. Different risk factors may increase or reduce the probability of contracting the disease. Mediterranean Diet has also a positive action on inflammation and immune system and could have a protective effect against severe acute respiratory syndrome coronavirus 2. Further studies are needed to corroborate the benefits of the Mediterranean Diet protective role on infection with SARS-CoV-2.
Collapse
|
61
|
Mukherjee A, Verma A, Bihani S, Burli A, Mantri K, Srivastava S. Proteomics advances towards developing SARS-CoV-2 therapeutics using in silico drug repurposing approaches. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 39:1-12. [PMID: 34906319 PMCID: PMC8222565 DOI: 10.1016/j.ddtec.2021.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/21/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022]
Abstract
Standing amidst the COVID-19 pandemic, we have faced major medical and economic crisis in recent times which remains to be an unresolved issue till date. Although the scientific community has made significant progress towards diagnosis and understanding the disease; however, effective therapeutics are still lacking. Several omics-based studies, especially proteomics and interactomics, have contributed significantly in terms of identifying biomarker panels that can potentially be used for the disease prognosis. This has also paved the way to identify the targets for drug repurposing as a therapeutic alternative. US Food and Drug Administration (FDA) has set in motion more than 500 drug development programs on an emergency basis, most of them are focusing on repurposed drugs. Remdesivir is one such success of a robust and quick drug repurposing approach. The advancements in omics-based technologies has allowed to explore altered host proteins, which were earlier restricted to only SARS-CoV-2 protein signatures. In this article, we have reviewed major contributions of proteomics and interactomics techniques towards identifying therapeutic targets for COVID-19. Furthermore, in-silico molecular docking approaches to streamline potential drug candidates are also discussed.
Collapse
Affiliation(s)
- Amrita Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Ayushi Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Surbhi Bihani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Ananya Burli
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Krishi Mantri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
62
|
Abstract
The development of effective antiviral therapy for COVID-19 is critical for those awaiting vaccination, as well as for those who do not respond robustly to vaccination. This review summarizes 1 year of progress in the race to develop antiviral therapies for COVID-19, including research spanning preclinical and clinical drug development efforts, with an emphasis on antiviral compounds that are in clinical development or that are high priorities for clinical development. The review is divided into sections on compounds that inhibit SARS-CoV-2 enzymes, including its polymerase and proteases; compounds that inhibit virus entry, including monoclonal antibodies; interferons; and repurposed drugs that inhibit host processes required for SARS-CoV-2 replication. The review concludes with a summary of the lessons to be learned from SARS-CoV-2 drug development efforts and the challenges to continued progress.
Collapse
Affiliation(s)
- Kaiming Tao
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Philip L. Tzou
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Janin Nouhin
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hector Bonilla
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Prasanna Jagannathan
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Robert W. Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
63
|
Identification of a dual acting SARS-CoV-2 proteases inhibitor through in silico design and step-by-step biological characterization. Eur J Med Chem 2021; 226:113863. [PMID: 34571172 PMCID: PMC8457654 DOI: 10.1016/j.ejmech.2021.113863] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/12/2022]
Abstract
COVID-19 pandemic, starting from the latest 2019, and caused by SARS-CoV-2 pathogen, led to the hardest health-socio-economic disaster in the last century. Despite the tremendous scientific efforts, mainly focused on the development of vaccines, identification of potent and efficient anti-SARS-CoV-2 therapeutics still represents an unmet need. Remdesivir, an anti-Ebola drug selected from a repurposing campaign, is the only drug approved, so far, for the treatment of the infection. Nevertheless, WHO in later 2020 has recommended against its use in COVID-19. In the present paper, we describe a step-by-step in silico design of a small library of compounds as main protease (Mpro) inhibitors. All the molecules were screened by an enzymatic assay on Mpro and, then, cellular activity was evaluated using Vero cells viral infection model. The cellular screening disclosed compounds 29 and 34 as in-vitro SARS-CoV-2 replication inhibitors at non-toxic concentrations (0.32 < EC50 < 5.98 μM). To rationalize these results, additional in-vitro assays were performed, focusing on papain like protease (PLpro) and spike protein (SP) as potential targets for the synthesized molecules. This pharmacological workflow allowed the identification of compound 29, as a dual acting SARS-CoV-2 proteases inhibitor featuring micromolar inhibitory potency versus Mpro (IC50 = 1.72 μM) and submicromolar potency versus PLpro (IC50 = 0.67 μM), and of compound 34 as a selective SP inhibitor (IC50 = 3.26 μM).
Collapse
|
64
|
Network pharmacology: curing causal mechanisms instead of treating symptoms. Trends Pharmacol Sci 2021; 43:136-150. [PMID: 34895945 DOI: 10.1016/j.tips.2021.11.004] [Citation(s) in RCA: 523] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/05/2021] [Accepted: 11/04/2021] [Indexed: 12/15/2022]
Abstract
For complex diseases, most drugs are highly ineffective, and the success rate of drug discovery is in constant decline. While low quality, reproducibility issues, and translational irrelevance of most basic and preclinical research have contributed to this, the current organ-centricity of medicine and the 'one disease-one target-one drug' dogma obstruct innovation in the most profound manner. Systems and network medicine and their therapeutic arm, network pharmacology, revolutionize how we define, diagnose, treat, and, ideally, cure diseases. Descriptive disease phenotypes are replaced by endotypes defined by causal, multitarget signaling modules that also explain respective comorbidities. Precise and effective therapeutic intervention is achieved by synergistic multicompound network pharmacology and drug repurposing, obviating the need for drug discovery and speeding up clinical translation.
Collapse
|
65
|
Tripathi D, Sodani M, Gupta PK, Kulkarni S. Host directed therapies: COVID-19 and beyond. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100058. [PMID: 34870156 PMCID: PMC8464038 DOI: 10.1016/j.crphar.2021.100058] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/14/2021] [Accepted: 09/19/2021] [Indexed: 12/15/2022] Open
Abstract
The global spread of SARS-CoV-2 has necessitated the development of novel, safe and effective therapeutic agents against this virus to stop the pandemic, however the development of novel antivirals may take years, hence, the best alternative available, is to repurpose the existing antiviral drugs with known safety profile in humans. After more than one year into this pandemic, global efforts have yielded the fruits and with the launch of many vaccines in the market, the world is inching towards the end of this pandemic, nonetheless, future pandemics of this magnitude or even greater cannot be denied. The preparedness against viruses of unknown origin should be maintained and the broad-spectrum antivirals with activity against range of viruses should be developed to curb future viral pandemics. The majority of antivirals developed till date are pathogen specific agents, which target critical viral pathways and lack broad spectrum activity required to target wide range of viruses. The surge in drug resistance among pathogens has rendered a compelling need to shift our focus towards host directed factors in the treatment of infectious diseases. This gains special relevance in the case of viral infections, where the pathogen encodes a handful of genes and predominantly depends on host factors for their propagation and persistence. Therefore, future antiviral drug development should focus more on targeting molecules of host pathways that are often hijacked by many viruses. Such cellular proteins of host pathways offer attractive targets for the development of broad-spectrum anticipatory antivirals. In the present article, we have reviewed the host directed therapies (HDTs) effective against viral infections with a special focus on COVID-19. This article also discusses the strategies involved in identifying novel host targets and subsequent development of broad spectrum HDTs.
Collapse
Affiliation(s)
- Devavrat Tripathi
- Radiation Medicine Centre, Bhabha Atomic Research Centre, C/O Tata Memorial Hospital Annexe, Parel, Mumbai, 400012, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Megha Sodani
- Radiation Medicine Centre, Bhabha Atomic Research Centre, C/O Tata Memorial Hospital Annexe, Parel, Mumbai, 400012, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Pramod Kumar Gupta
- Radiation Medicine Centre, Bhabha Atomic Research Centre, C/O Tata Memorial Hospital Annexe, Parel, Mumbai, 400012, India
- Corresponding author.
| | - Savita Kulkarni
- Radiation Medicine Centre, Bhabha Atomic Research Centre, C/O Tata Memorial Hospital Annexe, Parel, Mumbai, 400012, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Corresponding author. Radiation Medicine Centre, Bhabha Atomic Research Centre, C/O Tata Memorial Hospital Annexe, Parel, Mumbai, 400012, India.
| |
Collapse
|
66
|
Siminea N, Popescu V, Sanchez Martin JA, Florea D, Gavril G, Gheorghe AM, Iţcuş C, Kanhaiya K, Pacioglu O, Popa IL, Trandafir R, Tusa MI, Sidoroff M, Păun M, Czeizler E, Păun A, Petre I. Network analytics for drug repurposing in COVID-19. Brief Bioinform 2021; 23:6447433. [PMID: 34864885 PMCID: PMC8690228 DOI: 10.1093/bib/bbab490] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 01/08/2023] Open
Abstract
To better understand the potential of drug repurposing in COVID-19, we analyzed control strategies over essential host factors for SARS-CoV-2 infection. We constructed comprehensive directed protein–protein interaction (PPI) networks integrating the top-ranked host factors, the drug target proteins and directed PPI data. We analyzed the networks to identify drug targets and combinations thereof that offer efficient control over the host factors. We validated our findings against clinical studies data and bioinformatics studies. Our method offers a new insight into the molecular details of the disease and into potentially new therapy targets for it. Our approach for drug repurposing is significant beyond COVID-19 and may be applied also to other diseases.
Collapse
Affiliation(s)
- Nicoleta Siminea
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania.,Faculty of Mathematics and Computer Science, University of Bucharest, 14 Academiei, 010014, Romania
| | - Victor Popescu
- Department of Information Technologies, Åbo Akademi University, 3 Tuomiokirkontori, 20500, Finland
| | - Jose Angel Sanchez Martin
- Department of Computer Science, Technical University of Madrid, 7 Calle Ramiro de Maeztu, 28040, Spain
| | - Daniela Florea
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Georgiana Gavril
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Ana-Maria Gheorghe
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Corina Iţcuş
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Krishna Kanhaiya
- Department of Information Technologies, Åbo Akademi University, 3 Tuomiokirkontori, 20500, Finland
| | - Octavian Pacioglu
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Ioana Laura Popa
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Romica Trandafir
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Maria Iris Tusa
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Manuela Sidoroff
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Mihaela Păun
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania.,Faculty of Administration and Business, University of Bucharest, 4-12 Regina Elisabeta Boulevard, 030018, Romania
| | - Eugen Czeizler
- Department of Information Technologies, Åbo Akademi University, 3 Tuomiokirkontori, 20500, Finland.,Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Andrei Păun
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania.,Faculty of Mathematics and Computer Science, University of Bucharest, 14 Academiei, 010014, Romania
| | - Ion Petre
- Department of Mathematics and Statistics, University of Turku, 5 Vesilinnantie, 20014, Finland.,Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| |
Collapse
|
67
|
Aronskyy I, Masoudi-Sobhanzadeh Y, Cappuccio A, Zaslavsky E. Advances in the computational landscape for repurposed drugs against COVID-19. Drug Discov Today 2021; 26:2800-2815. [PMID: 34339864 PMCID: PMC8323501 DOI: 10.1016/j.drudis.2021.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic has caused millions of deaths and massive societal distress worldwide. Therapeutic solutions are urgently needed, but de novo drug development remains a lengthy process. One promising alternative is computational drug repurposing, which enables the prioritization of existing compounds through fast in silico analyses. Recent efforts based on molecular docking, machine learning, and network analysis have produced actionable predictions. Some predicted drugs, targeting viral proteins and pathological host pathways are undergoing clinical trials. Here, we review this work, highlight drugs with high predicted efficacy and classify their mechanisms of action. We discuss the strengths and limitations of the published methodologies and outline possible future directions. Finally, we curate a list of COVID-19 data portals and other repositories that could be used to accelerate future research.
Collapse
Affiliation(s)
- Illya Aronskyy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yosef Masoudi-Sobhanzadeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Antonio Cappuccio
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Elena Zaslavsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
68
|
Dong TN, Brogden G, Gerold G, Khosla M. A multitask transfer learning framework for the prediction of virus-human protein-protein interactions. BMC Bioinformatics 2021; 22:572. [PMID: 34837942 PMCID: PMC8626732 DOI: 10.1186/s12859-021-04484-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Viral infections are causing significant morbidity and mortality worldwide. Understanding the interaction patterns between a particular virus and human proteins plays a crucial role in unveiling the underlying mechanism of viral infection and pathogenesis. This could further help in prevention and treatment of virus-related diseases. However, the task of predicting protein-protein interactions between a new virus and human cells is extremely challenging due to scarce data on virus-human interactions and fast mutation rates of most viruses. RESULTS We developed a multitask transfer learning approach that exploits the information of around 24 million protein sequences and the interaction patterns from the human interactome to counter the problem of small training datasets. Instead of using hand-crafted protein features, we utilize statistically rich protein representations learned by a deep language modeling approach from a massive source of protein sequences. Additionally, we employ an additional objective which aims to maximize the probability of observing human protein-protein interactions. This additional task objective acts as a regularizer and also allows to incorporate domain knowledge to inform the virus-human protein-protein interaction prediction model. CONCLUSIONS Our approach achieved competitive results on 13 benchmark datasets and the case study for the SARS-COV-2 virus receptor. Experimental results show that our proposed model works effectively for both virus-human and bacteria-human protein-protein interaction prediction tasks. We share our code for reproducibility and future research at https://git.l3s.uni-hannover.de/dong/multitask-transfer .
Collapse
Affiliation(s)
- Thi Ngan Dong
- L3S Research Center, Leibniz University Hannover, Hannover, Germany.
| | - Graham Brogden
- Institute for Biochemistry, University of Veterinary Medicine, Hannover, Germany.,Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Gisa Gerold
- Institute for Biochemistry, University of Veterinary Medicine, Hannover, Germany.,Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany.,Department of Clinical Microbiology, Umeå University, Umeå, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Megha Khosla
- L3S Research Center, Leibniz University Hannover, Hannover, Germany
| |
Collapse
|
69
|
Network medicine for disease module identification and drug repurposing with the NeDRex platform. Nat Commun 2021; 12:6848. [PMID: 34824199 PMCID: PMC8617287 DOI: 10.1038/s41467-021-27138-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/04/2021] [Indexed: 12/17/2022] Open
Abstract
Traditional drug discovery faces a severe efficacy crisis. Repurposing of registered drugs provides an alternative with lower costs and faster drug development timelines. However, the data necessary for the identification of disease modules, i.e. pathways and sub-networks describing the mechanisms of complex diseases which contain potential drug targets, are scattered across independent databases. Moreover, existing studies are limited to predictions for specific diseases or non-translational algorithmic approaches. There is an unmet need for adaptable tools allowing biomedical researchers to employ network-based drug repurposing approaches for their individual use cases. We close this gap with NeDRex, an integrative and interactive platform for network-based drug repurposing and disease module discovery. NeDRex integrates ten different data sources covering genes, drugs, drug targets, disease annotations, and their relationships. NeDRex allows for constructing heterogeneous biological networks, mining them for disease modules, prioritizing drugs targeting disease mechanisms, and statistical validation. We demonstrate the utility of NeDRex in five specific use-cases.
Collapse
|
70
|
Mayburd A. Cross-testing of direct-action antivirals, universal vaccines, or search for host-level antivirals: what will sooner lead to a generic capability to combat the emerging viral pandemics? Expert Rev Anti Infect Ther 2021; 20:507-511. [PMID: 34719314 DOI: 10.1080/14787210.2022.2000859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mitigation of future viral pandemics is an enormous technical problem, but its solution is essential for preservation of life, economic well-being, and social stability. The author examined host-level, direct action antiviral, and universal vaccine approaches while presenting a specific screening proposal. AREAS COVERED The author examined the most recent biomedical literature publicly available in the databases and identified the publications supporting the principle of cross-applicability of direct-action antivirals (DAA) within similar viral families and at greater phylogenetic distances. EXPERT OPINION Comparing different approaches, the author showed that the cocktails of DAAs, including parent compounds that passed Phase I trials need to be preemptively tested for all major viral families, approved, and stockpiled (or dual-use production facilities designated). The quick distribution of the pre-approved and pre-positioned antiviral cocktails (even of moderate efficiency) reduces mortality and economic damage many-fold, resulting in the trillion-scale savings in a pandemic context. This pre-positioning approach is only one in the combinatorial toolkit that needs to be included in the plan for all viral families of importance. A dedicated international public-private initiative can achieve savings in these proactive preparedness efforts, as well as to keep the focus of politicians and public on the problem.
Collapse
Affiliation(s)
- Anatoly Mayburd
- School of System's Biology, George Mason University, Fairfax, VA, USA
| |
Collapse
|
71
|
Kotlyar M, Pastrello C, Ahmed Z, Chee J, Varyova Z, Jurisica I. IID 2021: towards context-specific protein interaction analyses by increased coverage, enhanced annotation and enrichment analysis. Nucleic Acids Res 2021; 50:D640-D647. [PMID: 34755877 PMCID: PMC8728267 DOI: 10.1093/nar/gkab1034] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/13/2021] [Accepted: 11/03/2021] [Indexed: 01/02/2023] Open
Abstract
Improved bioassays have significantly increased the rate of identifying new protein-protein interactions (PPIs), and the number of detected human PPIs has greatly exceeded early estimates of human interactome size. These new PPIs provide a more complete view of disease mechanisms but precise understanding of how PPIs affect phenotype remains a challenge. It requires knowledge of PPI context (e.g. tissues, subcellular localizations), and functional roles, especially within pathways and protein complexes. The previous IID release focused on PPI context, providing networks with comprehensive tissue, disease, cellular localization, and druggability annotations. The current update adds developmental stages to the available contexts, and provides a way of assigning context to PPIs that could not be previously annotated due to insufficient data or incompatibility with available context categories (e.g. interactions between membrane and cytoplasmic proteins). This update also annotates PPIs with conservation across species, directionality in pathways, membership in large complexes, interaction stability (i.e. stable or transient), and mutation effects. Enrichment analysis is now available for all annotations, and includes multiple options; for example, context annotations can be analyzed with respect to PPIs or network proteins. In addition to tabular view or download, IID provides online network visualization. This update is available at http://ophid.utoronto.ca/iid.
Collapse
Affiliation(s)
- Max Kotlyar
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Zuhaib Ahmed
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Justin Chee
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Zofia Varyova
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada.,Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON M5S 1A4, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
72
|
MotieGhader H, Safavi E, Rezapour A, Amoodizaj FF, Iranifam RA. Drug repurposing for coronavirus (SARS-CoV-2) based on gene co-expression network analysis. Sci Rep 2021; 11:21872. [PMID: 34750486 PMCID: PMC8576023 DOI: 10.1038/s41598-021-01410-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is a highly contagious viral respiratory illness. This illness is spurred on by a coronavirus known as SARS-associated coronavirus (SARS-CoV). SARS was first detected in Asia in late February 2003. The genome of this virus is very similar to the SARS-CoV-2. Therefore, the study of SARS-CoV disease and the identification of effective drugs to treat this disease can be new clues for the treatment of SARS-Cov-2. This study aimed to discover novel potential drugs for SARS-CoV disease in order to treating SARS-Cov-2 disease based on a novel systems biology approach. To this end, gene co-expression network analysis was applied. First, the gene co-expression network was reconstructed for 1441 genes, and then two gene modules were discovered as significant modules. Next, a list of miRNAs and transcription factors that target gene co-expression modules' genes were gathered from the valid databases, and two sub-networks formed of transcription factors and miRNAs were established. Afterward, the list of the drugs targeting obtained sub-networks' genes was retrieved from the DGIDb database, and two drug-gene and drug-TF interaction networks were reconstructed. Finally, after conducting different network analyses, we proposed five drugs, including FLUOROURACIL, CISPLATIN, SIROLIMUS, CYCLOPHOSPHAMIDE, and METHYLDOPA, as candidate drugs for SARS-CoV-2 coronavirus treatment. Moreover, ten miRNAs including miR-193b, miR-192, miR-215, miR-34a, miR-16, miR-16, miR-92a, miR-30a, miR-7, and miR-26b were found to be significant miRNAs in treating SARS-CoV-2 coronavirus.
Collapse
Affiliation(s)
- Habib MotieGhader
- Department of Basic Sciences, Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | - Esmaeil Safavi
- Department of Basic Sciences, Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Department of Basic Sciences, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Ali Rezapour
- Department of Animal Science, Faculty of Agriculture, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Fatemeh Firouzi Amoodizaj
- Department of Basic Sciences, Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Roya Asl Iranifam
- Department of Basic Sciences, Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
73
|
Cakir M, Obernier K, Forget A, Krogan NJ. Target Discovery for Host-Directed Antiviral Therapies: Application of Proteomics Approaches. mSystems 2021; 6:e0038821. [PMID: 34519533 PMCID: PMC8547474 DOI: 10.1128/msystems.00388-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Current epidemics, such as AIDS or flu, and the emergence of new threatening pathogens, such as the one causing the current coronavirus disease 2019 (COVID-19) pandemic, represent major global health challenges. While vaccination is an important part of the arsenal to counter the spread of viral diseases, it presents limitations and needs to be complemented by efficient therapeutic solutions. Intricate knowledge of host-pathogen interactions is a powerful tool to identify host-dependent vulnerabilities that can be exploited to dampen viral replication. Such host-directed antiviral therapies are promising and are less prone to the development of drug-resistant viral strains. Here, we first describe proteomics-based strategies that allow the rapid characterization of host-pathogen interactions. We then discuss how such data can be exploited to help prioritize compounds with potential host-directed antiviral activity that can be tested in preclinical models.
Collapse
Affiliation(s)
- Merve Cakir
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, USA
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, California, USA
| | - Kirsten Obernier
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, USA
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, California, USA
| | - Antoine Forget
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, USA
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, California, USA
| | - Nevan J. Krogan
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, USA
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, California, USA
| |
Collapse
|
74
|
Menestrina L, Cabrelle C, Recanatini M. COVIDrugNet: a network-based web tool to investigate the drugs currently in clinical trial to contrast COVID-19. Sci Rep 2021; 11:19426. [PMID: 34593915 PMCID: PMC8484553 DOI: 10.1038/s41598-021-98812-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/13/2021] [Indexed: 02/08/2023] Open
Abstract
The COVID-19 pandemic poses a huge problem of public health that requires the implementation of all available means to contrast it, and drugs are one of them. In this context, we observed an unmet need of depicting the continuously evolving scenario of the ongoing drug clinical trials through an easy-to-use, freely accessible online tool. Starting from this consideration, we developed COVIDrugNet ( http://compmedchem.unibo.it/covidrugnet ), a web application that allows users to capture a holistic view and keep up to date on how the clinical drug research is responding to the SARS-CoV-2 infection. Here, we describe the web app and show through some examples how one can explore the whole landscape of medicines in clinical trial for the treatment of COVID-19 and try to probe the consistency of the current approaches with the available biological and pharmacological evidence. We conclude that careful analyses of the COVID-19 drug-target system based on COVIDrugNet can help to understand the biological implications of the proposed drug options, and eventually improve the search for more effective therapies.
Collapse
Affiliation(s)
- Luca Menestrina
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
| | - Chiara Cabrelle
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
| | - Maurizio Recanatini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy.
| |
Collapse
|
75
|
Zambrana C, Xenos A, Böttcher R, Malod-Dognin N, Pržulj N. Network neighbors of viral targets and differentially expressed genes in COVID-19 are drug target candidates. Sci Rep 2021; 11:18985. [PMID: 34556735 PMCID: PMC8460804 DOI: 10.1038/s41598-021-98289-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic is raging. It revealed the importance of rapid scientific advancement towards understanding and treating new diseases. To address this challenge, we adapt an explainable artificial intelligence algorithm for data fusion and utilize it on new omics data on viral-host interactions, human protein interactions, and drugs to better understand SARS-CoV-2 infection mechanisms and predict new drug-target interactions for COVID-19. We discover that in the human interactome, the human proteins targeted by SARS-CoV-2 proteins and the genes that are differentially expressed after the infection have common neighbors central in the interactome that may be key to the disease mechanisms. We uncover 185 new drug-target interactions targeting 49 of these key genes and suggest re-purposing of 149 FDA-approved drugs, including drugs targeting VEGF and nitric oxide signaling, whose pathways coincide with the observed COVID-19 symptoms. Our integrative methodology is universal and can enable insight into this and other serious diseases.
Collapse
Affiliation(s)
| | | | | | - Noël Malod-Dognin
- Barcelona Supercomputing Center, Barcelona, Spain
- Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Nataša Pržulj
- Barcelona Supercomputing Center, Barcelona, Spain.
- Department of Computer Science, University College London, London, WC1E 6BT, UK.
- ICREA, Pg. Lluís Companys 23, Barcelona, Spain.
| |
Collapse
|
76
|
A Vision of Future Healthcare: Potential Opportunities and Risks of Systems Medicine from a Citizen and Patient Perspective-Results of a Qualitative Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18189879. [PMID: 34574802 PMCID: PMC8465522 DOI: 10.3390/ijerph18189879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022]
Abstract
Advances in (bio)medicine and technological innovations make it possible to combine high-dimensional, heterogeneous health data to better understand causes of diseases and make them usable for predictive, preventive, and precision medicine. This study aimed to determine views on and expectations of “systems medicine” from the perspective of citizens and patients in six focus group interviews, all transcribed verbatim and content analyzed. A future vision of the use of systems medicine in healthcare served as a stimulus for the discussion. The results show that although certain aspects of systems medicine were seen positive (e.g., use of smart technology, digitalization, and networking in healthcare), the perceived risks dominated. The high degree of technification was perceived as emotionally burdensome (e.g., reduction of people to their data, loss of control, dehumanization). The risk-benefit balance for the use of risk-prediction models for disease events and trajectories was rated as rather negative. There were normative and ethical concerns about unwanted data use, discrimination, and restriction of fundamental rights. These concerns and needs of citizens and patients must be addressed in policy frameworks and health policy implementation strategies to reduce negative emotions and attitudes toward systems medicine and to take advantage of its opportunities.
Collapse
|
77
|
Wang G, Zeng L, Huang Q, Lu Z, Sui R, Liu D, Zeng H, Liu X, Chu S, Kou X, Li H. Exploring the Molecular Mechanism of Liuwei Dihuang Pills for Treating Diabetic Nephropathy by Combined Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:7262208. [PMID: 34552655 PMCID: PMC8452392 DOI: 10.1155/2021/7262208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/20/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a common and serious complication of diabetes, but without a satisfactory treatment strategy till now. Liuwei Dihuang pills (LDP), an effective Chinese medicinal formula, has been used to treat DN for more than 1000 years. However, its underlying mechanism of action is still vague. METHODS Active compounds and corresponding targets of LDP were predicted from the TCMSP database. DN disease targets were extracted from the OMIM, GeneCards, TTD, DisGeNET, and DrugBank databases. Subsequently, the "herbal-compound-target" network and protein-protein interaction (PPI) network were constructed and analyzed via the STRING web platform and Cytoscape software. GO functional and KEGG pathway enrichment analyses were carried out on the Metascape web platform. Molecular docking utilized AutoDock Vina and PyMOL software. RESULTS 41 active components and 186 corresponding targets of LDP were screened out. 131 common targets of LDP and DN were acquired. Quercetin, kaempferol, beta-sitosterol, diosgenin, and stigmasterol could be defined as five crucial compounds. JUN, MAPK8, AKT1, EGF, TP53, VEGFA, MMP9, MAPK1, and TNF might be the nine key targets. The enrichment analysis showed that common targets were mainly associated with inflammation reaction, oxidative stress, immune regulation, and cell apoptosis. AGE-RAGE and IL-17 were the suggested two significant signal pathways. Molecular docking revealed that the nine key targets could closely bind to their corresponding active compounds. CONCLUSION The present study fully reveals the multicompound's and multitarget's characteristics of LDP in DN treatment. Furthermore, this study provides valuable evidence for further scientific research of the pharmacological mechanisms and broader clinical application.
Collapse
Affiliation(s)
- Gaoxiang Wang
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen 518033, Guangdong, China
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Lin Zeng
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong, China
| | - Qian Huang
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong, China
| | - Zhaoqi Lu
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong, China
| | - Ruiqing Sui
- Department of Oncology, Lishui District Traditional Chinese Medicine Hospital, Nanjing 211200, Jiangsu, China
| | - Deliang Liu
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Hua Zeng
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen 518033, Guangdong, China
| | - Xuemei Liu
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Shufang Chu
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Xinhui Kou
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Huilin Li
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| |
Collapse
|
78
|
Azad A, Fatima S, Capraro A, Waters SA, Vafaee F. Integrative resource for network-based investigation of COVID-19 combinatorial drug repositioning and mechanism of action. PATTERNS (NEW YORK, N.Y.) 2021; 2:100325. [PMID: 34278363 PMCID: PMC8277549 DOI: 10.1016/j.patter.2021.100325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/12/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022]
Abstract
An effective monotherapy to target the complex and multifactorial pathology of SARS-CoV-2 infection poses a challenge to drug repositioning, which can be improved by combination therapy. We developed an online network pharmacology-based drug repositioning platform, COVID-CDR (http://vafaeelab.com/COVID19repositioning.html), that enables a visual and quantitative investigation of the interplay between the primary drug targets and the SARS-CoV-2-host interactome in the human protein-protein interaction network. COVID-CDR prioritizes drug combinations with potential to act synergistically through different, yet potentially complementary, pathways. It provides the options for understanding multi-evidence drug-pair similarity scores along with several other relevant information on individual drugs or drug pairs. Overall, COVID-CDR is a first-of-its-kind online platform that provides a systematic approach for pre-clinical in silico investigation of combination therapies for treating COVID-19 at the fingertips of the clinicians and researchers.
Collapse
Affiliation(s)
- A.K.M. Azad
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Shadma Fatima
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
- Department of Medical Oncology, Ingham Institute of Applied Research, Sydney, Australia
| | - Alexander Capraro
- School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre, UNSW Sydney and Sydney Children's Hospital, Sydney, Australia
| | - Shafagh A. Waters
- School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre, UNSW Sydney and Sydney Children's Hospital, Sydney, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
- Data Science Hub, University of New South Wales, Kensington, NSW, Australia
| |
Collapse
|
79
|
Hollander M, Do T, Will T, Helms V. Detecting Rewiring Events in Protein-Protein Interaction Networks Based on Transcriptomic Data. FRONTIERS IN BIOINFORMATICS 2021; 1:724297. [PMID: 36303788 PMCID: PMC9581068 DOI: 10.3389/fbinf.2021.724297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/23/2021] [Indexed: 12/25/2022] Open
Abstract
Proteins rarely carry out their cellular functions in isolation. Instead, eukaryotic proteins engage in about six interactions with other proteins on average. The aggregated protein interactome of an organism forms a “hairy ball”-type protein-protein interaction (PPI) network. Yet, in a typical human cell, only about half of all proteins are expressed at a particular time. Hence, it has become common practice to prune the full PPI network to the subset of expressed proteins. If RNAseq data is available, one can further resolve the specific protein isoforms present in a cell or tissue. Here, we review various approaches, software tools and webservices that enable users to construct context-specific or tissue-specific PPI networks and how these are rewired between two cellular conditions. We illustrate their different functionalities on the example of the interactions involving the human TNR6 protein. In an outlook, we describe how PPI networks may be integrated with epigenetic data or with data on the activity of splicing factors.
Collapse
|
80
|
Luo L, Qiu Q, Huang F, Liu K, Lan Y, Li X, Huang Y, Cui L, Luo H. Drug repurposing against coronavirus disease 2019 (COVID-19): A review. J Pharm Anal 2021; 11:683-690. [PMID: 34513115 PMCID: PMC8416689 DOI: 10.1016/j.jpha.2021.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/16/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Since December 2019, severe acute respiratory syndrome coronavirus 2 has been found to be the culprit in the coronavirus disease 2019 (COVID-19), causing a global pandemic. Despite the existence of many vaccine programs, the number of confirmed cases and fatalities due to COVID-19 is still increasing. Furthermore, a number of variants have been reported. Because of the absence of approved anti-coronavirus drugs, the treatment and management of COVID-19 has become a global challenge. Under these circumstances, drug repurposing is an effective method to identify candidate drugs with a shorter cycle of clinical trials. Here, we summarize the current status of the application of drug repurposing in COVID-19, including drug repurposing based on virtual computer screening, network pharmacology, and bioactivity, which may be a beneficial COVID-19 treatment. Mechanism of SARS-CoV-2 infection and drug targets were reviewed. Drug repurposing against COVID-19 based on computer virtual screening, network pharmacology, bioactivity were summarized. The use of drug repurposing in COVID-19 was addressed.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.,Marine Medical Research Institute of Zhanjiang, Zhanjiang, 524023, Guangdong, China
| | - Qin Qiu
- Graduate School, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Fangfang Huang
- Graduate School, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Kaifeng Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yongqi Lan
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xiaoling Li
- Animal Experiment Center, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yuge Huang
- Department of Pediatrics, the Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| |
Collapse
|
81
|
Mok DZL, Chan CYY, Ooi EE, Chan KR. The effects of aging on host resistance and disease tolerance to SARS-CoV-2 infection. FEBS J 2021; 288:5055-5070. [PMID: 33124149 PMCID: PMC8518758 DOI: 10.1111/febs.15613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 01/08/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) crisis caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has triggered a large-scale pandemic that is afflicting millions of individuals in over 200 countries. The clinical spectrum caused by SARS-CoV-2 infections can range from asymptomatic infection to mild undifferentiated febrile illness to severe respiratory disease with multiple complications. Elderly patients (aged 60 and above) with comorbidities such as cardiovascular diseases and diabetes mellitus appear to be at highest risk of a severe disease outcome. To protect against pulmonary immunopathology caused by SARS-CoV-2 infection, the host primarily depends on two distinct defense strategies: resistance and disease tolerance. Resistance is the ability of the host to suppress and eliminate incoming viruses. By contrast, disease tolerance refers to host responses that promote host health regardless of their impact on viral replication. Disruption of either resistance or disease tolerance mechanisms or both could underpin predisposition to elevated risk of severe disease during viral infection. Aging can disrupt host resistance and disease tolerance by compromising immune functions, weakening of the unfolded protein response, progressive mitochondrial dysfunction, and altering metabolic processes. A comprehensive understanding of the molecular mechanisms underlying declining host defense in elderly individuals could thus pave the way to provide new opportunities and approaches for the treatment of severe COVID-19.
Collapse
Affiliation(s)
- Darren Z. L. Mok
- Emerging Infectious Diseases ProgramDuke‐NUS Medical SchoolSingaporeSingapore
| | | | - Eng Eong Ooi
- Emerging Infectious Diseases ProgramDuke‐NUS Medical SchoolSingaporeSingapore
- Viral Research & Experimental Medicine Center @ SingHealth/Duke‐NUS (ViREMiCS)SingaporeSingapore
- Singapore‐MIT Alliance in Research and TechnologyAntimicrobial Resistance Interdisciplinary Research GroupSingaporeSingapore
- Saw Swee Hock School of Public HealthNational University of SingaporeSingapore
- Department of Microbiology and ImmunologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Kuan Rong Chan
- Emerging Infectious Diseases ProgramDuke‐NUS Medical SchoolSingaporeSingapore
| |
Collapse
|
82
|
In silico analysis of the aggregation propensity of the SARS-CoV-2 proteome: Insight into possible cellular pathologies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140693. [PMID: 34237472 PMCID: PMC8256665 DOI: 10.1016/j.bbapap.2021.140693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022]
Abstract
The SARS-CoV-2 virus causes the coronavirus disease 19 emerged in 2020. The pandemic triggered a turmoil in public health and is having a tremendous social and economic impact around the globe. Upon entry into host cells, the SARS-CoV-2 virus hijacks cellular machineries to produce and maintain its own proteins, spreading the infection. Although the disease is known for prominent respiratory symptoms, accumulating evidence is also demonstrating the involvement of the central nervous system, with possible mid- and long-term neurological consequences. In this study, we conducted a detailed bioinformatic analysis of the SARS-CoV-2 proteome aggregation propensity by using several complementary computational tools. Our study identified 10 aggregation prone proteins in the reference SARS-CoV-2 strain: the non-structural proteins Nsp4, Nsp6 and Nsp7 as well as ORF3a, ORF6, ORF7a, ORF7b, ORF10, CovE and CovM. By searching for the available mutants of each protein, we have found that most proteins are conserved, while ORF3a and ORF7b are variable and characterized by the occurrence of a large number of mutants with increased aggregation propensity. The geographical distribution of the mutants revealed interesting differences in the localization of aggregation-prone mutants of each protein. Aggregation-prone mutants of ORF7b were found in 7 European countries, whereas those of ORF3a in only 2. Aggregation-prone sequences of ORF7b, but not of ORF3a, were identified in Australia, India, Nepal, China, and Thailand. Our results are important for future analysis of a possible correlation between higher transmissibility and infection, as well as the presence of neurological symptoms with aggregation propensity of SARS-CoV-2 proteins.
Collapse
|
83
|
Unal MA, Bayrakdar F, Nazir H, Besbinar O, Gurcan C, Lozano N, Arellano LM, Yalcin S, Panatli O, Celik D, Alkaya D, Agan A, Fusco L, Suzuk Yildiz S, Delogu LG, Akcali KC, Kostarelos K, Yilmazer A. Graphene Oxide Nanosheets Interact and Interfere with SARS-CoV-2 Surface Proteins and Cell Receptors to Inhibit Infectivity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101483. [PMID: 33988903 PMCID: PMC8236978 DOI: 10.1002/smll.202101483] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Indexed: 05/19/2023]
Abstract
Nanotechnology can offer a number of options against coronavirus disease 2019 (COVID-19) acting both extracellularly and intracellularly to the host cells. Here, the aim is to explore graphene oxide (GO), the most studied 2D nanomaterial in biomedical applications, as a nanoscale platform for interaction with SARS-CoV-2. Molecular docking analyses of GO sheets on interaction with three different structures: SARS-CoV-2 viral spike (open state - 6VYB or closed state - 6VXX), ACE2 (1R42), and the ACE2-bound spike complex (6M0J) are performed. GO shows high affinity for the surface of all three structures (6M0J, 6VYB and 6VXX). When binding affinities and involved bonding types are compared, GO interacts more strongly with the spike or ACE2, compared to 6M0J. Infection experiments using infectious viral particles from four different clades as classified by Global Initiative on Sharing all Influenza Data (GISAID), are performed for validation purposes. Thin, biological-grade GO nanoscale (few hundred nanometers in lateral dimension) sheets are able to significantly reduce copies for three different viral clades. This data has demonstrated that GO sheets have the capacity to interact with SARS-CoV-2 surface components and disrupt infectivity even in the presence of any mutations on the viral spike. GO nanosheets are proposed to be further explored as a nanoscale platform for development of antiviral strategies against COVID-19.
Collapse
Affiliation(s)
| | - Fatma Bayrakdar
- Ministry of Health General Directorate of Public HealthMicrobiology References LaboratorySihhiyeAnkara06430Turkey
| | - Hasan Nazir
- Department of ChemistryAnkara UniversityTandoganAnkara06100Turkey
| | - Omur Besbinar
- Stem Cell InstituteAnkara UniversityBalgatAnkara06520Turkey
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| | - Cansu Gurcan
- Stem Cell InstituteAnkara UniversityBalgatAnkara06520Turkey
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| | - Neus Lozano
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)UAB Campus BellaterraBarcelona08193Spain
| | - Luis M. Arellano
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)UAB Campus BellaterraBarcelona08193Spain
| | - Süleyman Yalcin
- Ministry of Health General Directorate of Public HealthMicrobiology References LaboratorySihhiyeAnkara06430Turkey
| | - Oguzhan Panatli
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| | - Dogantan Celik
- Stem Cell InstituteAnkara UniversityBalgatAnkara06520Turkey
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| | - Damla Alkaya
- Stem Cell InstituteAnkara UniversityBalgatAnkara06520Turkey
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| | - Aydan Agan
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| | - Laura Fusco
- Department of Biomedical SciencesUniversity of PaduaPadua35122Italy
| | - Serap Suzuk Yildiz
- Ministry of Health General Directorate of Public HealthMicrobiology References LaboratorySihhiyeAnkara06430Turkey
| | | | - Kamil Can Akcali
- Stem Cell InstituteAnkara UniversityBalgatAnkara06520Turkey
- Department of BiophysicsFaculty of MedicineAnkara UniversitySihhiyeAnkara06230Turkey
| | - Kostas Kostarelos
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)UAB Campus BellaterraBarcelona08193Spain
- Nanomedicine Lab National Graphene Institute and Faculty of Biology Medicine & HealthThe University of ManchesterAV Hill BuildingManchesterM13 9PTUnited Kingdom
| | - Açelya Yilmazer
- Stem Cell InstituteAnkara UniversityBalgatAnkara06520Turkey
- Department of Biomedical EngineeringAnkara UniversityGolbasiAnkara06830Turkey
| |
Collapse
|
84
|
Han N, Hwang W, Tzelepis K, Schmerer P, Yankova E, MacMahon M, Lei W, M Katritsis N, Liu A, Felgenhauer U, Schuldt A, Harris R, Chapman K, McCaughan F, Weber F, Kouzarides T. Identification of SARS-CoV-2-induced pathways reveals drug repurposing strategies. SCIENCE ADVANCES 2021; 7:eabh3032. [PMID: 34193418 PMCID: PMC8245040 DOI: 10.1126/sciadv.abh3032] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/14/2021] [Indexed: 05/02/2023]
Abstract
The global outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) necessitates the rapid development of new therapies against coronavirus disease 2019 (COVID-19) infection. Here, we present the identification of 200 approved drugs, appropriate for repurposing against COVID-19. We constructed a SARS-CoV-2-induced protein network, based on disease signatures defined by COVID-19 multiomics datasets, and cross-examined these pathways against approved drugs. This analysis identified 200 drugs predicted to target SARS-CoV-2-induced pathways, 40 of which are already in COVID-19 clinical trials, testifying to the validity of the approach. Using artificial neural network analysis, we classified these 200 drugs into nine distinct pathways, within two overarching mechanisms of action (MoAs): viral replication (126) and immune response (74). Two drugs (proguanil and sulfasalazine) implicated in viral replication were shown to inhibit replication in cell assays. This unbiased and validated analysis opens new avenues for the rapid repurposing of approved drugs into clinical trials.
Collapse
Affiliation(s)
- Namshik Han
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK.
| | - Woochang Hwang
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | | | - Patrick Schmerer
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Gießen 35392, Germany
| | - Eliza Yankova
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Méabh MacMahon
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
- Centre for Therapeutics Discovery, LifeArc, Stevenage, UK
| | - Winnie Lei
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Nicholas M Katritsis
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Anika Liu
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Data and Computational Sciences, GSK, London, UK
| | - Ulrike Felgenhauer
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Gießen 35392, Germany
| | - Alison Schuldt
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Rebecca Harris
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Kathryn Chapman
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Frank McCaughan
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Gießen 35392, Germany
| | - Tony Kouzarides
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK.
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
85
|
Salgado-Albarrán M, Navarro-Delgado EI, Del Moral-Morales A, Alcaraz N, Baumbach J, González-Barrios R, Soto-Reyes E. Comparative transcriptome analysis reveals key epigenetic targets in SARS-CoV-2 infection. NPJ Syst Biol Appl 2021; 7:21. [PMID: 34031419 PMCID: PMC8144203 DOI: 10.1038/s41540-021-00181-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/08/2021] [Indexed: 02/04/2023] Open
Abstract
COVID-19 is an infection caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome coronavirus 2), which has caused a global outbreak. Current research efforts are focused on the understanding of the molecular mechanisms involved in SARS-CoV-2 infection in order to propose drug-based therapeutic options. Transcriptional changes due to epigenetic regulation are key host cell responses to viral infection and have been studied in SARS-CoV and MERS-CoV; however, such changes are not fully described for SARS-CoV-2. In this study, we analyzed multiple transcriptomes obtained from cell lines infected with MERS-CoV, SARS-CoV, and SARS-CoV-2, and from COVID-19 patient-derived samples. Using integrative analyses of gene co-expression networks and de-novo pathway enrichment, we characterize different gene modules and protein pathways enriched with Transcription Factors or Epifactors relevant for SARS-CoV-2 infection. We identified EP300, MOV10, RELA, and TRIM25 as top candidates, and more than 60 additional proteins involved in the epigenetic response during viral infection that has therapeutic potential. Our results show that targeting the epigenetic machinery could be a feasible alternative to treat COVID-19.
Collapse
Affiliation(s)
- Marisol Salgado-Albarrán
- grid.7220.70000 0001 2157 0393Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa (UAM-C), Mexico City, Mexico ,grid.6936.a0000000123222966Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Erick I. Navarro-Delgado
- grid.419167.c0000 0004 1777 1207Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Aylin Del Moral-Morales
- grid.7220.70000 0001 2157 0393Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa (UAM-C), Mexico City, Mexico
| | - Nicolas Alcaraz
- grid.5254.60000 0001 0674 042XThe Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jan Baumbach
- grid.9026.d0000 0001 2287 2617Chair of Computational Systems Biology, University of Hamburg, Hamburg, Germany ,grid.10825.3e0000 0001 0728 0170Computational BioMedicine Lab, Institute of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Rodrigo González-Barrios
- grid.419167.c0000 0004 1777 1207Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Ernesto Soto-Reyes
- grid.7220.70000 0001 2157 0393Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa (UAM-C), Mexico City, Mexico
| |
Collapse
|
86
|
Can the COVID-19 Pandemic Disrupt the Current Drug Development Practices? Int J Mol Sci 2021; 22:ijms22115457. [PMID: 34064287 PMCID: PMC8196831 DOI: 10.3390/ijms22115457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/24/2022] Open
Abstract
Therapeutics and vaccines against the COVID-19 pandemic need to be developed rapidly and efficiently, given its severity. To maximize the efficiency and productivity of drug development, the world has adopted disruptive technologies and approaches in various drug development areas. Telehealth, characterized by the heavy use of digital technologies; drug repositioning strategies, aided by computational breakthroughs; and data tracking tool hubs, enabling real-time information sharing, have received much attention. Moreover, drug developers have engaged in open innovation by establishing various types of collaborations, many of which have been carried out across nations and enterprises. Finally, regulatory agencies have attempted to operate on a more flexible review basis than before. Although such disruptive approaches have partly reshaped drug development practices, issues and challenges remain before the completion of this paradigm shift in conventional drug development practices for the post-pandemic era. In this review, we have highlighted the role of a collaborative community of experts in order to figure out how disruptive technologies can be fully integrated into the current drug development practices and improve drug development efficiency for the post-pandemic era.
Collapse
|
87
|
Exploring antibody repurposing for COVID-19: beyond presumed roles of therapeutic antibodies. Sci Rep 2021; 11:10220. [PMID: 33986382 PMCID: PMC8119408 DOI: 10.1038/s41598-021-89621-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/29/2021] [Indexed: 12/24/2022] Open
Abstract
The urgent need for a treatment of COVID-19 has left researchers with limited choice of either developing an effective vaccine or identifying approved/investigational drugs developed for other medical conditions for potential repurposing, thus bypassing long clinical trials. In this work, we compared the sequences of experimentally verified SARS-CoV-2 neutralizing antibodies and sequentially/structurally similar commercialized therapeutic monoclonal antibodies. We have identified three therapeutic antibodies, Tremelimumab, Ipilimumab and Afasevikumab. Interestingly, these antibodies target CTLA4 and IL17A, levels of which have been shown to be elevated during severe SARS-CoV-2 infection. The candidate antibodies were evaluated further for epitope restriction, interaction energy and interaction surface to gauge their repurposability to tackle SARS-CoV-2 infection. Our work provides candidate antibody scaffolds with dual activities of plausible viral neutralization and immunosuppression. Further, these candidate antibodies can also be explored in diagnostic test kits for SARS-CoV-2 infection. We opine that this in silico workflow to screen and analyze antibodies for repurposing would have widespread applications.
Collapse
|
88
|
Morselli Gysi D, do Valle Í, Zitnik M, Ameli A, Gan X, Varol O, Ghiassian SD, Patten JJ, Davey RA, Loscalzo J, Barabási AL. Network medicine framework for identifying drug-repurposing opportunities for COVID-19. Proc Natl Acad Sci U S A 2021; 118:e2025581118. [PMID: 33906951 PMCID: PMC8126852 DOI: 10.1073/pnas.2025581118] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The COVID-19 pandemic has highlighted the need to quickly and reliably prioritize clinically approved compounds for their potential effectiveness for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Here, we deployed algorithms relying on artificial intelligence, network diffusion, and network proximity, tasking each of them to rank 6,340 drugs for their expected efficacy against SARS-CoV-2. To test the predictions, we used as ground truth 918 drugs experimentally screened in VeroE6 cells, as well as the list of drugs in clinical trials that capture the medical community's assessment of drugs with potential COVID-19 efficacy. We find that no single predictive algorithm offers consistently reliable outcomes across all datasets and metrics. This outcome prompted us to develop a multimodal technology that fuses the predictions of all algorithms, finding that a consensus among the different predictive methods consistently exceeds the performance of the best individual pipelines. We screened in human cells the top-ranked drugs, obtaining a 62% success rate, in contrast to the 0.8% hit rate of nonguided screenings. Of the six drugs that reduced viral infection, four could be directly repurposed to treat COVID-19, proposing novel treatments for COVID-19. We also found that 76 of the 77 drugs that successfully reduced viral infection do not bind the proteins targeted by SARS-CoV-2, indicating that these network drugs rely on network-based mechanisms that cannot be identified using docking-based strategies. These advances offer a methodological pathway to identify repurposable drugs for future pathogens and neglected diseases underserved by the costs and extended timeline of de novo drug development.
Collapse
Affiliation(s)
- Deisy Morselli Gysi
- Network Science Institute, Northeastern University, Boston, MA 02115
- Department of Physics, Northeastern University, Boston, MA 02115
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Ítalo do Valle
- Network Science Institute, Northeastern University, Boston, MA 02115
- Department of Physics, Northeastern University, Boston, MA 02115
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard University, Boston, MA 02115
- Harvard Data Science Initiative, Harvard University, Cambridge, MA 02138
| | - Asher Ameli
- Department of Physics, Northeastern University, Boston, MA 02115
- Data Science Department, Scipher Medicine, Waltham, MA 02453
| | - Xiao Gan
- Network Science Institute, Northeastern University, Boston, MA 02115
- Department of Physics, Northeastern University, Boston, MA 02115
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Onur Varol
- Network Science Institute, Northeastern University, Boston, MA 02115
- Department of Physics, Northeastern University, Boston, MA 02115
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | | | - J J Patten
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Robert A Davey
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Albert-László Barabási
- Network Science Institute, Northeastern University, Boston, MA 02115;
- Department of Physics, Northeastern University, Boston, MA 02115
- Department of Network and Data Science, Central European University, Budapest 1051, Hungary
| |
Collapse
|
89
|
Brief review on repurposed drugs and vaccines for possible treatment of COVID-19. Eur J Pharmacol 2021; 898:173977. [PMID: 33639193 PMCID: PMC7905377 DOI: 10.1016/j.ejphar.2021.173977] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/06/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the pandemic coronavirus disease 2019 (Covid-19) has claimed more than a million lives. Various in silico, in vitro, and in vivo studies are being conducted to understand the effect of SARS-CoV-2 on the cellular metabolism of humans and the various drugs and drug-targets that may be used. In this review, we discuss protein-protein interactions (PPIs) between viral and human proteins as well as viral targets like proteases. We try to understand the molecular mechanism of various repurposed antiviral drugs against SARS-CoV-2, their combination therapies, drug dosage regimens, and their adverse effects along with possible alternatives like non-toxic antiviral phytochemicals. Ultimately, randomized controlled trials are needed to identify which of these compounds has the required balance of efficacy and safety. We also focus on the recent advancements in diagnostic methods and vaccine candidates developed around the world to fight against Covid-19.
Collapse
|
90
|
Krämer A, Billaud JN, Tugendreich S, Shiffman D, Jones M, Green J. The Coronavirus Network Explorer: mining a large-scale knowledge graph for effects of SARS-CoV-2 on host cell function. BMC Bioinformatics 2021; 22:229. [PMID: 33941085 PMCID: PMC8091149 DOI: 10.1186/s12859-021-04148-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Leveraging previously identified viral interactions with human host proteins, we apply a machine learning-based approach to connect SARS-CoV-2 viral proteins to relevant host biological functions, diseases, and pathways in a large-scale knowledge graph derived from the biomedical literature. Our goal is to explore how SARS-CoV-2 could interfere with various host cell functions, and to identify drug targets amongst the host genes that could potentially be modulated against COVID-19 by repurposing existing drugs. The machine learning model employed here involves gene embeddings that leverage causal gene expression signatures curated from literature. In contrast to other network-based approaches for drug repurposing, our approach explicitly takes the direction of effects into account, distinguishing between activation and inhibition. RESULTS We have constructed 70 networks connecting SARS-CoV-2 viral proteins to various biological functions, diseases, and pathways reflecting viral biology, clinical observations, and co-morbidities in the context of COVID-19. Results are presented in the form of interactive network visualizations through a web interface, the Coronavirus Network Explorer (CNE), that allows exploration of underlying experimental evidence. We find that existing drugs targeting genes in those networks are strongly enriched in the set of drugs that are already in clinical trials against COVID-19. CONCLUSIONS The approach presented here can identify biologically plausible hypotheses for COVID-19 pathogenesis, explicitly connected to the immunological, virological and pathological observations seen in SARS-CoV-2 infected patients. The discovery of repurposable drugs is driven by prior knowledge of relevant functional endpoints that reflect known viral biology or clinical observations, therefore suggesting potential mechanisms of action. We believe that the CNE offers relevant insights that go beyond more conventional network approaches, and can be a valuable tool for drug repurposing. The CNE is available at https://digitalinsights.qiagen.com/coronavirus-network-explorer .
Collapse
Affiliation(s)
| | | | | | | | | | - Jeff Green
- Digital Insights, QIAGEN, Redwood City, USA
| |
Collapse
|
91
|
Dotolo S, Marabotti A, Facchiano A, Tagliaferri R. A review on drug repurposing applicable to COVID-19. Brief Bioinform 2021; 22:726-741. [PMID: 33147623 PMCID: PMC7665348 DOI: 10.1093/bib/bbaa288] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Drug repurposing involves the identification of new applications for existing drugs at a lower cost and in a shorter time. There are different computational drug-repurposing strategies and some of these approaches have been applied to the coronavirus disease 2019 (COVID-19) pandemic. Computational drug-repositioning approaches applied to COVID-19 can be broadly categorized into (i) network-based models, (ii) structure-based approaches and (iii) artificial intelligence (AI) approaches. Network-based approaches are divided into two categories: network-based clustering approaches and network-based propagation approaches. Both of them allowed to annotate some important patterns, to identify proteins that are functionally associated with COVID-19 and to discover novel drug–disease or drug–target relationships useful for new therapies. Structure-based approaches allowed to identify small chemical compounds able to bind macromolecular targets to evaluate how a chemical compound can interact with the biological counterpart, trying to find new applications for existing drugs. AI-based networks appear, at the moment, less relevant since they need more data for their application.
Collapse
Affiliation(s)
| | | | | | - Roberto Tagliaferri
- Artificial Intelligence, Statistical Pattern Recognition, Clustering, Biomedical imaging and Bioinformatics
| |
Collapse
|
92
|
Lazareva O, Baumbach J, List M, Blumenthal DB. On the limits of active module identification. Brief Bioinform 2021; 22:6189770. [PMID: 33782690 DOI: 10.1093/bib/bbab066] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
In network and systems medicine, active module identification methods (AMIMs) are widely used for discovering candidate molecular disease mechanisms. To this end, AMIMs combine network analysis algorithms with molecular profiling data, most commonly, by projecting gene expression data onto generic protein-protein interaction (PPI) networks. Although active module identification has led to various novel insights into complex diseases, there is increasing awareness in the field that the combination of gene expression data and PPI network is problematic because up-to-date PPI networks have a very small diameter and are subject to both technical and literature bias. In this paper, we report the results of an extensive study where we analyzed for the first time whether widely used AMIMs really benefit from using PPI networks. Our results clearly show that, except for the recently proposed AMIM DOMINO, the tested AMIMs do not produce biologically more meaningful candidate disease modules on widely used PPI networks than on random networks with the same node degrees. AMIMs hence mainly learn from the node degrees and mostly fail to exploit the biological knowledge encoded in the edges of the PPI networks. This has far-reaching consequences for the field of active module identification. In particular, we suggest that novel algorithms are needed which overcome the degree bias of most existing AMIMs and/or work with customized, context-specific networks instead of generic PPI networks.
Collapse
Affiliation(s)
- Olga Lazareva
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany.,Chair of Computational Systems Biology, University of Hamburg, Hamburg, Germany.,Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Markus List
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| | - David B Blumenthal
- Chair of Experimental Bioinformatics, Technical University of Munich, Freising, Germany
| |
Collapse
|
93
|
Structure insights of SARS-CoV-2 open state envelope protein and inhibiting through active phytochemical of ayurvedic medicinal plants from Withania somnifera. Saudi J Biol Sci 2021; 28:3594-3601. [PMID: 33758570 PMCID: PMC7970802 DOI: 10.1016/j.sjbs.2021.03.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Coronaviruses have been causing pandemic situations across the globe for the past two decades and the focus is on identifying suitable novel targets for antivirals and vaccine development. SARS-CoV-2 encodes a small hydrophobic envelope (E) protein that mediates envelope formation, budding, replication, and release of progeny viruses from the host. Through this study, the SARS-CoV-2 E protein is studied for its open and closed state and focused in identifying antiviral herbs used in traditional medicine practices for COVID-19 infections. In this study using computational tools, we docked the shortlisted phytochemicals with the envelope protein of the SARS-CoV-2 virus and the results hint that these compounds interact with the pore-lining residues. The molecular level understanding of the open state is considered and the active inhibitors from the phytochemicals of Ayurvedic medicinal plants from Withania somnifera. We have thus identified a potential phytochemical compound that directly binds with the pore region of the E protein and thereby blocks its channel activity. Blocking the ion channel activity of E protein is directly related to the inhibition of virus replication. The study shows encouraging results on the usage of these phytochemicals in the treatment/management of SARS-CoV-2 infection.
Collapse
|
94
|
Mishra CB, Pandey P, Sharma RD, Malik MZ, Mongre RK, Lynn AM, Prasad R, Jeon R, Prakash A. Identifying the natural polyphenol catechin as a multi-targeted agent against SARS-CoV-2 for the plausible therapy of COVID-19: an integrated computational approach. Brief Bioinform 2021; 22:1346-1360. [PMID: 33386025 PMCID: PMC7799228 DOI: 10.1093/bib/bbaa378] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/03/2020] [Accepted: 11/26/2020] [Indexed: 01/18/2023] Open
Abstract
The global pandemic crisis, coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has claimed the lives of millions of people across the world. Development and testing of anti-SARS-CoV-2 drugs or vaccines have not turned to be realistic within the timeframe needed to combat this pandemic. Here, we report a comprehensive computational approach to identify the multi-targeted drug molecules against the SARS-CoV-2 proteins, whichare crucially involved in the viral-host interaction, replication of the virus inside the host, disease progression and transmission of coronavirus infection. Virtual screening of 75 FDA-approved potential antiviral drugs against the target proteins, spike (S) glycoprotein, human angiotensin-converting enzyme 2 (hACE2), 3-chymotrypsin-like cysteine protease (3CLpro), cathepsin L (CTSL), nucleocapsid protein, RNA-dependent RNA polymerase (RdRp) and non-structural protein 6 (NSP6), resulted in the selection of seven drugs which preferentially bind to the target proteins. Further, the molecular interactions determined by molecular dynamics simulation revealed that among the 75 drug molecules, catechin can effectively bind to 3CLpro, CTSL, RBD of S protein, NSP6 and nucleocapsid protein. It is more conveniently involved in key molecular interactions, showing binding free energy (ΔGbind) in the range of -5.09 kcal/mol (CTSL) to -26.09 kcal/mol (NSP6). At the binding pocket, catechin is majorly stabilized by the hydrophobic interactions, displays ΔEvdW values: -7.59 to -37.39 kcal/mol. Thus, the structural insights of better binding affinity and favorable molecular interaction of catechin toward multiple target proteins signify that catechin can be potentially explored as a multi-targeted agent against COVID-19.
Collapse
Affiliation(s)
| | - Preeti Pandey
- Department of Chemistry & Biochemistry, University of Oklahoma, OK, USA
| | | | - Md Zubbair Malik
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Raj Kumar Mongre
- College of Pharmacy, Sookmyung Women’s University, Seoul, South Korea
| | - Andrew M Lynn
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology and is the dean of Faculty of Science Engineering and Technology, Amity University Haryana, Haryana 122413, India
| | - Raok Jeon
- College of Pharmacy, Sookmyung Women’s University, Seoul, South Korea
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity Institute of Integrative Sciences and Health, Amity University, Haryana
| |
Collapse
|
95
|
Wang LL, Lo K. Text mining approaches for dealing with the rapidly expanding literature on COVID-19. Brief Bioinform 2021; 22:781-799. [PMID: 33279995 PMCID: PMC7799291 DOI: 10.1093/bib/bbaa296] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
More than 50 000 papers have been published about COVID-19 since the beginning of 2020 and several hundred new papers continue to be published every day. This incredible rate of scientific productivity leads to information overload, making it difficult for researchers, clinicians and public health officials to keep up with the latest findings. Automated text mining techniques for searching, reading and summarizing papers are helpful for addressing information overload. In this review, we describe the many resources that have been introduced to support text mining applications over the COVID-19 literature; specifically, we discuss the corpora, modeling resources, systems and shared tasks that have been introduced for COVID-19. We compile a list of 39 systems that provide functionality such as search, discovery, visualization and summarization over the COVID-19 literature. For each system, we provide a qualitative description and assessment of the system's performance, unique data or user interface features and modeling decisions. Many systems focus on search and discovery, though several systems provide novel features, such as the ability to summarize findings over multiple documents or linking between scientific articles and clinical trials. We also describe the public corpora, models and shared tasks that have been introduced to help reduce repeated effort among community members; some of these resources (especially shared tasks) can provide a basis for comparing the performance of different systems. Finally, we summarize promising results and open challenges for text mining the COVID-19 literature.
Collapse
Affiliation(s)
- Lucy Lu Wang
- The Allen Institute for Artificial Intelligence, Seattle, WA 98112, USA
| | - Kyle Lo
- The Allen Institute for Artificial Intelligence, Seattle, WA 98112, USA
| |
Collapse
|
96
|
Mercatelli D, Holding AN, Giorgi FM. Web tools to fight pandemics: the COVID-19 experience. Brief Bioinform 2021; 22:690-700. [PMID: 33057582 PMCID: PMC7665357 DOI: 10.1093/bib/bbaa261] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 01/30/2023] Open
Abstract
The current outbreak of COVID-19 has generated an unprecedented scientific response worldwide, with the generation of vast amounts of publicly available epidemiological, biological and clinical data. Bioinformatics scientists have quickly produced online methods to provide non-computational users with the opportunity of analyzing such data. In this review, we report the results of this effort, by cataloguing the currently most popular web tools for COVID-19 research and analysis. Our focus was driven on tools drawing data from the fields of epidemiology, genomics, interactomics and pharmacology, in order to provide a meaningful depiction of the current state of the art of COVID-19 online resources.
Collapse
|
97
|
Ahsan MA, Liu Y, Feng C, Zhou Y, Ma G, Bai Y, Chen M. Bioinformatics resources facilitate understanding and harnessing clinical research of SARS-CoV-2. Brief Bioinform 2021; 22:714-725. [PMID: 33432321 PMCID: PMC7929412 DOI: 10.1093/bib/bbaa416] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/18/2020] [Accepted: 12/19/2020] [Indexed: 12/17/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by the coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has created an unprecedented threat to public health. The pandemic has been sweeping the globe, impacting more than 200 countries, with more outbreaks still lurking on the horizon. At the time of the writing, no approved drugs or vaccines are available to treat COVID-19 patients, prompting an urgent need to decipher mechanisms underlying the pathogenesis and develop curative treatments. To fight COVID-19, researchers around the world have provided specific tools and molecular information for SARS-CoV-2. These pieces of information can be integrated to aid computational investigations and facilitate clinical research. This paper reviews current knowledge, the current status of drug development and various resources for key steps toward effective treatment of COVID-19, including the phylogenetic characteristics, genomic conservation and interaction data. The final goal of this paper is to provide information that may be utilized in bioinformatics approaches and aid target prioritization and drug repurposing. Several SARS-CoV-2-related tools/databases were reviewed, and a web-portal named OverCOVID (http://bis.zju.edu.cn/overcovid/) is constructed to provide a detailed interpretation of SARS-CoV-2 basics and share a collection of resources that may contribute to therapeutic advances. These information could improve researchers' understanding of SARS-CoV-2 and help to accelerate the development of new antiviral treatments.
Collapse
Affiliation(s)
- Md Asif Ahsan
- Department of Bioinformatics, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yongjing Liu
- Department of Bioinformatics, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Cong Feng
- Department of Bioinformatics, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yincong Zhou
- Department of Bioinformatics, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Guangyuan Ma
- Department of Bioinformatics, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Youhuang Bai
- Department of Bioinformatics, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences; The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
98
|
Kumar Das J, Tradigo G, Veltri P, H Guzzi P, Roy S. Data science in unveiling COVID-19 pathogenesis and diagnosis: evolutionary origin to drug repurposing. Brief Bioinform 2021; 22:855-872. [PMID: 33592108 PMCID: PMC7929414 DOI: 10.1093/bib/bbaa420] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/09/2020] [Accepted: 12/19/2020] [Indexed: 12/20/2022] Open
Abstract
MOTIVATION The outbreak of novel severe acute respiratory syndrome coronavirus (SARS-CoV-2, also known as COVID-19) in Wuhan has attracted worldwide attention. SARS-CoV-2 causes severe inflammation, which can be fatal. Consequently, there has been a massive and rapid growth in research aimed at throwing light on the mechanisms of infection and the progression of the disease. With regard to this data science is playing a pivotal role in in silico analysis to gain insights into SARS-CoV-2 and the outbreak of COVID-19 in order to forecast, diagnose and come up with a drug to tackle the virus. The availability of large multiomics, radiological, bio-molecular and medical datasets requires the development of novel exploratory and predictive models, or the customisation of existing ones in order to fit the current problem. The high number of approaches generates the need for surveys to guide data scientists and medical practitioners in selecting the right tools to manage their clinical data. RESULTS Focusing on data science methodologies, we conduct a detailed study on the state-of-the-art of works tackling the current pandemic scenario. We consider various current COVID-19 data analytic domains such as phylogenetic analysis, SARS-CoV-2 genome identification, protein structure prediction, host-viral protein interactomics, clinical imaging, epidemiological research and drug discovery. We highlight data types and instances, their generation pipelines and the data science models currently in use. The current study should give a detailed sketch of the road map towards handling COVID-19 like situations by leveraging data science experts in choosing the right tools. We also summarise our review focusing on prime challenges and possible future research directions. CONTACT hguzzi@unicz.it, sroy01@cus.ac.in.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Department of Pediatrics, School of Medicine, Johns Hopkins University, Maryland, USA
| | - Giuseppe Tradigo
- eCampus University, Via Isimbardi 10, 22060 Novedrate, CO, Italy
| | - Pierangelo Veltri
- Department of Surgical and Medical Sciences, Magna Graecia University, Catanzaro, 88100, Italy
| | - Pietro H Guzzi
- Department of Surgical and Medical Sciences, Magna Graecia University, Catanzaro, 88100, Italy
| | - Swarup Roy
- Network Reconstruction & Analysis (NetRA) Lab, Department of Computer Applications, Sikkim University, Gangtok, India
| |
Collapse
|
99
|
Hufsky F, Lamkiewicz K, Almeida A, Aouacheria A, Arighi C, Bateman A, Baumbach J, Beerenwinkel N, Brandt C, Cacciabue M, Chuguransky S, Drechsel O, Finn RD, Fritz A, Fuchs S, Hattab G, Hauschild AC, Heider D, Hoffmann M, Hölzer M, Hoops S, Kaderali L, Kalvari I, von Kleist M, Kmiecinski R, Kühnert D, Lasso G, Libin P, List M, Löchel HF, Martin MJ, Martin R, Matschinske J, McHardy AC, Mendes P, Mistry J, Navratil V, Nawrocki EP, O’Toole ÁN, Ontiveros-Palacios N, Petrov AI, Rangel-Pineros G, Redaschi N, Reimering S, Reinert K, Reyes A, Richardson L, Robertson DL, Sadegh S, Singer JB, Theys K, Upton C, Welzel M, Williams L, Marz M. Computational strategies to combat COVID-19: useful tools to accelerate SARS-CoV-2 and coronavirus research. Brief Bioinform 2021; 22:642-663. [PMID: 33147627 PMCID: PMC7665365 DOI: 10.1093/bib/bbaa232] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/28/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is a novel virus of the family Coronaviridae. The virus causes the infectious disease COVID-19. The biology of coronaviruses has been studied for many years. However, bioinformatics tools designed explicitly for SARS-CoV-2 have only recently been developed as a rapid reaction to the need for fast detection, understanding and treatment of COVID-19. To control the ongoing COVID-19 pandemic, it is of utmost importance to get insight into the evolution and pathogenesis of the virus. In this review, we cover bioinformatics workflows and tools for the routine detection of SARS-CoV-2 infection, the reliable analysis of sequencing data, the tracking of the COVID-19 pandemic and evaluation of containment measures, the study of coronavirus evolution, the discovery of potential drug targets and development of therapeutic strategies. For each tool, we briefly describe its use case and how it advances research specifically for SARS-CoV-2. All tools are free to use and available online, either through web applications or public code repositories. Contact:evbc@unj-jena.de.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Christian Brandt
- Institute of Infectious Disease and Infection Control at Jena University Hospital, Germany
| | - Marco Cacciabue
- Consejo Nacional de Investigaciones Científicas y Tócnicas (CONICET) working on FMDV virology at the Instituto de Agrobiotecnología y Biología Molecular (IABiMo, INTA-CONICET) and at the Departamento de Ciencias Básicas, Universidad Nacional de Luján (UNLu), Argentina
| | | | - Oliver Drechsel
- bioinformatics department at the Robert Koch-Institute, Germany
| | | | - Adrian Fritz
- Computational Biology of Infection Research group of Alice C. McHardy at the Helmholtz Centre for Infection Research, Germany
| | - Stephan Fuchs
- bioinformatics department at the Robert Koch-Institute, Germany
| | - Georges Hattab
- Bioinformatics Division at Philipps-University Marburg, Germany
| | | | - Dominik Heider
- Data Science in Biomedicine at the Philipps-University of Marburg, Germany
| | | | | | - Stefan Hoops
- Biocomplexity Institute and Initiative at the University of Virginia, USA
| | - Lars Kaderali
- Bioinformatics and head of the Institute of Bioinformatics at University Medicine Greifswald, Germany
| | | | - Max von Kleist
- bioinformatics department at the Robert Koch-Institute, Germany
| | - Renó Kmiecinski
- bioinformatics department at the Robert Koch-Institute, Germany
| | | | - Gorka Lasso
- Chandran Lab, Albert Einstein College of Medicine, USA
| | | | | | | | | | | | | | - Alice C McHardy
- Computational Biology of Infection Research Lab at the Helmholtz Centre for Infection Research in Braunschweig, Germany
| | - Pedro Mendes
- Center for Quantitative Medicine of the University of Connecticut School of Medicine, USA
| | | | - Vincent Navratil
- Bioinformatics and Systems Biology at the Rhône Alpes Bioinformatics core facility, Universitó de Lyon, France
| | | | | | | | | | | | - Nicole Redaschi
- Development of the Swiss-Prot group at the SIB for UniProt and SIB resources that cover viral biology (ViralZone)
| | - Susanne Reimering
- Computational Biology of Infection Research group of Alice C. McHardy at the Helmholtz Centre for Infection Research
| | | | | | | | | | - Sepideh Sadegh
- Chair of Experimental Bioinformatics at Technical University of Munich, Germany
| | - Joshua B Singer
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, UK
| | | | - Chris Upton
- Department of Biochemistry and Microbiology, University of Victoria, Canada
| | | | | | - Manja Marz
- Friedrich Schiller University Jena, Germany
| |
Collapse
|
100
|
Caruso FP, Scala G, Cerulo L, Ceccarelli M. A review of COVID-19 biomarkers and drug targets: resources and tools. Brief Bioinform 2021; 22:701-713. [PMID: 33279954 PMCID: PMC7799271 DOI: 10.1093/bib/bbaa328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/05/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023] Open
Abstract
The stratification of patients at risk of progression of COVID-19 and their molecular characterization is of extreme importance to optimize treatment and to identify therapeutic options. The bioinformatics community has responded to the outbreak emergency with a set of tools and resource to identify biomarkers and drug targets that we review here. Starting from a consolidated corpus of 27 570 papers, we adopt latent Dirichlet analysis to extract relevant topics and select those associated with computational methods for biomarker identification and drug repurposing. The selected topics span from machine learning and artificial intelligence for disease characterization to vaccine development and to therapeutic target identification. Although the way to go for the ultimate defeat of the pandemic is still long, the amount of knowledge, data and tools generated so far constitutes an unprecedented example of global cooperation to this threat.
Collapse
|