1
|
Hu S, Zhong Q, Xie X, Zhang S, Wang J, Liu H, Dai W. Research progress on critical viral protease inhibitors for coronaviruses and enteroviruses. Bioorg Med Chem Lett 2025; 122:130168. [PMID: 40074013 DOI: 10.1016/j.bmcl.2025.130168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Viral infectious diseases have been seriously affecting human life and health. SARS-CoV-2 was the pathogen that caused Coronavirus Disease 2019 (COVID-19), and the impact of COVID-19 is still existing. Enterovirus 71 (EV71) is the primary pathogen of hand, foot, and mouth disease (HFMD), and no effective direct-acting antiviral drugs targeting EV71 has been approved yet. Innate antiviral strategies play an important role in preventing virus infections depending on the powerful immune regulatory system of body, while viruses have evolved to exploit diverse methods to overcome immune response. Viral proteases, which are known in cleaving viral polyproteins, have also been found to modulate the innate immunity of host cells, thereby promoting viral proliferation. Herein, we reviewed the current development of SARS-CoV-2 3CLpro, PLpro, and EV71 3Cpro and 2Apro, mainly including structure, function, modulation of immune response, and inhibitors of these four proteases, to further deepen the understanding of viral pathogenesis and provide a new perspective for subsequent corresponding drug development.
Collapse
Affiliation(s)
- Shulei Hu
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Qiuyu Zhong
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Xiong Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Shurui Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jinlin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Hong Liu
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| | - Wenhao Dai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| |
Collapse
|
2
|
Hoque I, Singh N, Ghosh Dastidar U, Martin AK, Joshi A, Sardana Y, Singh Chawla R, Das N, Patra B, Devi R, Das S, Das D, Kumar S, Ringe RP, Bokara KK, Thakur KG, Talukdar A. Strategic Design and Optimization of Umifenovir Analogues: Balancing Antiviral Efficacy and hERG Toxicity against SARS-CoV-2. J Med Chem 2025; 68:9371-9406. [PMID: 40263709 DOI: 10.1021/acs.jmedchem.4c03093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Arbidol (ARB, Umifenovir), a broad-spectrum antiviral from Russia, lacks Food and Drug Administration (FDA) approval due to insufficient clinical data and undocumented toxicity concerns. Its indole scaffold, with six unique substitutions, enables optimization for improved efficacy. This study optimized ARB's antiviral potency and safety by modifying the N1, C2, C3, and C4 positions. Antiviral efficacy was evaluated in SARS-CoV-2-infected VERO E6 cells, while optimization was guided by absorption, distribution, metabolism, and excretion (ADME), in vivo pharmacokinetic (PK) and hERG. Early modifications at N1 and C2 produced compounds 10 and 14 (IC50 = 1.5 μM), surpassing ARB (IC50 = 9.0 μM). Further refinements yielded compounds 42 (IC50 = 1.1 μM) and 56 (IC50 = 0.24 μM), resolving hERG toxicity (>30 μM). C3 modifications led to lead compounds 77, 79, and 81 (IC50 = 0.67-0.7 μM), achieving superior potency while eliminating hERG toxicity. Mechanism of entry inhibition and immunofluorescence confirmed compound 77 significantly reduced SARS-CoV-2 within Vero cells, supporting their preclinical potential.
Collapse
Affiliation(s)
- Israful Hoque
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Nittu Singh
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Uddipta Ghosh Dastidar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alna Kuriyickal Martin
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
| | - Akshay Joshi
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Yogesh Sardana
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
| | - Ravneet Singh Chawla
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Nirmal Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Binita Patra
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Renuga Devi
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
| | - Satyajeet Das
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Dipankar Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Sahil Kumar
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Rajesh P Ringe
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Kiran Kumar Bokara
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Krishan Gopal Thakur
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
3
|
Padey B, Droillard C, Dulière V, Fouret J, Lamballerie CND, Milesi C, Laurent E, Brun P, Traversier A, Julien T, Terrier O, Rosa-Calatrava M, Pizzorno A. Host-targeted repurposed diltiazem enhances the antiviral activity of direct acting antivirals against Influenza A virus and SARS-CoV-2. Antiviral Res 2025; 237:106138. [PMID: 40049293 DOI: 10.1016/j.antiviral.2025.106138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/18/2025] [Accepted: 03/02/2025] [Indexed: 03/30/2025]
Abstract
Viral respiratory infections remain a major and recurrent public health threat. Among them, influenza viruses are responsible for ⁓500,000 deaths worldwide and a high economic burden. The recurrent threat of emerging zoonotic or pandemic viruses worsens this scenario, being SARS-CoV-2 and the millions of COVID-19 deaths the most recent example. The rapid evolution of circulating influenza and SARS-CoV-2 viruses allows the emergence and dissemination of variant strains carrying mutations resulting in suboptimal vaccine protection and/or reduced efficacy of current limited therapeutic arsenal. In this context, host-targeted approaches constitute a promising antiviral strategy aiming to achieve broad-spectrum activity and mitigate the emergence of viral resistance against classic direct acting antivirals. Here, we demonstrated that diltiazem, a calcium channel blocker currently used to treat angor, induces an ISG expression profile characteristic of an antiviral cellular state mainly driven by IFN-λ. We then evaluated the potential of the diltiazem-baloxavir combination against Influenza A wild-type and the PA I38T resistant strain in cell culture and human airway epithelia (HAE). We analogously evaluated the diltiazem-molnupiravir combination against SARS-CoV-2, including variants of concern. Our results demonstrate the broad-spectrum antiviral activity of diltiazem against Influenza A viruses, including resistant strains, as well as the capacity to potentiate the antiviral effect of baloxavir. The diltiazem-molnupiravir combination further reduced viral production and protected the integrity of HAE infected with SARS-CoV-2. This study highlights the major interest of combining direct acting and host-targeted agents as a promising strategy against circulating and emerging viruses.
Collapse
Affiliation(s)
- Blandine Padey
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; Signia Therapeutics SAS, Lyon, France
| | - Clément Droillard
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Victoria Dulière
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Julien Fouret
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; Signia Therapeutics SAS, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France; Nexomis, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, 69008, France
| | - Claire Nicolas de Lamballerie
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; Signia Therapeutics SAS, Lyon, France
| | - Cédrine Milesi
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Emilie Laurent
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Pauline Brun
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Aurélien Traversier
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Thomas Julien
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France; Nexomis, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, 69008, France; Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada.
| | - Andrés Pizzorno
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France; International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France.
| |
Collapse
|
4
|
Sanna G, Riabova O, Kazakova E, Lepioshkin A, Monakhova N, Marongiu A, Franci G, Manzin A, Makarov V. Efficacy of dispirotripiperazine PDSTP in a golden Syrian hamster model of SARS-CoV-2 infection. Front Microbiol 2025; 16:1546946. [PMID: 40130242 PMCID: PMC11931052 DOI: 10.3389/fmicb.2025.1546946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/10/2025] [Indexed: 03/26/2025] Open
Abstract
The increasing incidence of viral pandemics calls for new small-molecule therapeutics beyond traditional approaches and targets. Dispirotripiperazine, composed of two positively charged nitrogen atoms, represents an unusual scaffold in drug discovery campaigns, and molecules based on it are known to prevent virus infection by disrupting early host-pathogen interactions. In this study, the adhesion-blocking dispirotripiperazine core compound PDSTP was evaluated against SARS-CoV-2 in vitro and in vivo. We demonstrated that the molecule was acceptably active against two clinical isolates affecting the early stages of the SARS-CoV-2 cycle. In a hamster model of SARS-CoV-2 pneumonia, PDSTP treatment resulted in reduced viral loads in the lungs and turbinates and milder lung tissue lesions. Overall, these data support PDSTP as a preclinical candidate for the treatment of COVID-19.
Collapse
Affiliation(s)
- Giuseppina Sanna
- Department of Biomedical Sciences, Microbiology and Virology Unit, University of Cagliari, Cittadella Universitaria, Cagliari, Italy
| | - Olga Riabova
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Elena Kazakova
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Alexander Lepioshkin
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Natalia Monakhova
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Alessandra Marongiu
- Department of Biomedical Sciences, Microbiology and Virology Unit, University of Cagliari, Cittadella Universitaria, Cagliari, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Aldo Manzin
- Department of Biomedical Sciences, Microbiology and Virology Unit, University of Cagliari, Cittadella Universitaria, Cagliari, Italy
| | - Vadim Makarov
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| |
Collapse
|
5
|
Ahmad S, Alafnan A, Alobaida A, Shahab U, Rehman S, Khan S, Khan MY, Puri P, Pandey RP, Ahmad I, Rafi Z. Decoding the SARS-CoV-2 infection process: Insights into origin, spread, and therapeutic approaches. Microb Pathog 2025; 200:107328. [PMID: 39863091 DOI: 10.1016/j.micpath.2025.107328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/29/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Globally, over 768 million confirmed cases and 6.9 million deaths had been documented as of July 17, 2023. Coronaviruses have a relatively large RNA genome. As with other viruses, SARS-CoV-2 does have an envelope film produced from host cells that are assisted by virally encoded glycoproteins that are required for infectivity, immunological assault, and viral particle production. Although the intermediate source of origin and transmission to humans is unexplained, rapid transmission from human to human has been established. This review focuses on the mechanistic framework for understanding the SARS-CoV-2 viral infection. Additionally, it discusses the origins and implications of COVID-19 using direct quotations from the published scientific literature to avoid misinterpretation of this catastrophic event that resulted in a massive loss of human life and impact on the global economy. The current available information unfolds large number of topics related with COVID-19 and/or the coronavirus (SARS-CoV-2) responsible of the disease. This review article also delves into the multifaceted aspects of COVID-19 and SARS-CoV-2, with a specific focus on a controversial yet essential issue: the possible association between SARS-CoV-2's origin and aldose reductase, an enzyme known for its role in diabetic retinopathy. Exploring this connection holds utmost significance, offering valuable insights into COVID-19's pathogenesis and unlocking new avenues for therapeutic interventions. It is important to trace back the evolution of coronaviruses and reveal the possible origin of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes the coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Saudi Arabia.
| | - Ahmed Alafnan
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail City, 2440, Saudi Arabia.
| | - Ahmed Alobaida
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail City, 2440, Saudi Arabia.
| | - Uzma Shahab
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail City, 2440, Saudi Arabia.
| | - Shahnawaz Rehman
- IIRC-1, Laboratory of Glycation Biology and Metabolic Disorders, Integral University, U.P., India.
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Hail, 2440, Hail, 2440, Saudi Arabia.
| | - Mohd Yasir Khan
- Department of Biotechnology, School of Applied & Life Science, Uttaranchal University Dehradun, India.
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India.
| | - Ramendra Pati Pandey
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana, 131029, India.
| | - Irfan Ahmad
- Central Labs, King Khalid University, AlQura'a, P.O. Box 960, Abha, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Zeeshan Rafi
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India.
| |
Collapse
|
6
|
Li MY, Deng K, Cheng XH, Siu LYL, Gao ZR, Naik TS, Stancheva VG, Cheung PPH, Teo QW, van Leur SW, Wong HH, Lan Y, Lam TTY, Sun MX, Zhang NN, Zhang Y, Cao TS, Yang F, Deng YQ, Sanyal S, Qin CF. ARF4-mediated intracellular transport as a broad-spectrum antiviral target. Nat Microbiol 2025; 10:710-723. [PMID: 39972062 DOI: 10.1038/s41564-025-01940-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025]
Abstract
Host factors that are involved in modulating cellular vesicular trafficking of virus progeny could be potential antiviral drug targets. ADP-ribosylation factors (ARFs) are GTPases that regulate intracellular vesicular transport upon GTP binding. Here we demonstrate that genetic depletion of ARF4 suppresses viral infection by multiple pathogenic RNA viruses including Zika virus (ZIKV), influenza A virus (IAV) and SARS-CoV-2. Viral infection leads to ARF4 activation and virus production is rescued upon complementation with active ARF4, but not with inactive mutants. Mechanistically, ARF4 deletion disrupts translocation of virus progeny into the Golgi complex and redirects them for lysosomal degradation, thereby blocking virus release. More importantly, peptides targeting ARF4 show therapeutic efficacy against ZIKV and IAV challenge in mice by inhibiting ARF4 activation. Our findings highlight the role of ARF4 during viral infection and its potential as a broad-spectrum antiviral target for further development.
Collapse
Affiliation(s)
- Ming-Yuan Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kao Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-He Cheng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Lewis Yu-Lam Siu
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhuo-Ran Gao
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Trupti Shivaprasad Naik
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Peter Pak-Hang Cheung
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi-Wen Teo
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie W van Leur
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Ho-Him Wong
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yun Lan
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Tommy Tsan-Yuk Lam
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Meng-Xu Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Na-Na Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yue Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tian-Shu Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Fan Yang
- Institute of Pathogenic Biology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China.
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Khalili Fakhrabadi A, Shahbazzadeh MJ, Jalali N, Eslami M. A hybrid inception-dilated-ResNet architecture for deep learning-based prediction of COVID-19 severity. Sci Rep 2025; 15:6490. [PMID: 39987169 PMCID: PMC11846838 DOI: 10.1038/s41598-025-91322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 02/19/2025] [Indexed: 02/24/2025] Open
Abstract
Chest computed tomography (CT) scans are essential for accurately assessing the severity of the novel Coronavirus (COVID-19), facilitating appropriate therapeutic interventions and monitoring disease progression. However, determining COVID-19 severity requires a radiologist with significant expertise. This study introduces a pioneering utilization of deep learning (DL) for evaluate COVID-19 severity using lung CT images, presenting a novel and effective method for assessing the severity of pulmonary manifestations in COVID-19 patients. Inception-Residual networks (Inception-ResNet), advanced hybrid models known for their compactness and effectiveness, were used to extract relevant features from CT scans. Inception-ResNet incorporates the dilated mechanism into its ResNet component, enhancing its ability to accurately classify lung involvement stages. This study demonstrates that dilated residual networks (dResNet) outperform their non-dilated counterparts in image classification tasks, as their architectural designs allow the systems to acquire comprehensive global data by expanding their receptive fields. Our study utilized an initial dataset of 1548 human thoracic CT scans, meticulously annotated by two experienced specialists. Lung involvement was determined by calculating a percentage based on observations made at each scan. The hybrid methodology successfully distinguished the ten distinct severity levels associated with COVID-19, achieving a maximum accuracy of 96.40%. This system demonstrates its effectiveness as a diagnostic framework for assessing lung involvement in COVID-19-affected individuals, facilitating disease progression tracking.
Collapse
Affiliation(s)
- Ali Khalili Fakhrabadi
- Department of Electrical Engineering, Kerman Branch, Islamic Azad University, Kerman, Iran
| | | | - Nazanin Jalali
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Neurology Department, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mahdiyeh Eslami
- Department of Electrical Engineering, Kerman Branch, Islamic Azad University, Kerman, Iran
| |
Collapse
|
8
|
Shehzadi K, Kalsoom I, Yu MJ, Liang JH. Design and in-silico evaluation of PNA-based novel pronucleotide analogues targeting RNA-dependent RNA polymerase to combat COVID-19. J Biomol Struct Dyn 2025:1-23. [PMID: 39937582 DOI: 10.1080/07391102.2024.2335287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/19/2024] [Indexed: 02/13/2025]
Abstract
The emergence of highly contagious SARS-CoV-2 variants emphasizes the need for antiviral drugs that can adapt to evolving viral mutations. Despite widespread vaccination efforts, novel variants and recurrence cases raise concerns about COVID-19. Although repurposed drugs like Remdesivir, a nucleoside inhibitor, offer treatment, there is still a critical need for alternative drugs. Inhibiting viral RdRp function remains a key strategy. Structural analysis highlights the importance of pyrrolo-triazine and pyrimidine scaffolds in nucleoside inhibitors. Our study designed Peptide Nucleic Acid (PNA) antisense pronucleotides by combining these scaffolds using structure-guided drug design. Molecular modeling, including molecular docking, pharmacokinetics, molecular dynamics simulations, and MMPBSA binding energy calculations, predicts that modified PNAs can disrupt ribosome assembly at the RdRp translation start site. The neutral backbone of PNAs may enhance sequence-specific RNA binding. MD simulations revealed that complexes of Remdesivir and L14 remained stable throughout, with the phosphate tail of L14 stabilized by a positive amino acid pocket near the RdRp-RNA entry channel, similar to Remdesivir. Additionally, L14's guanine motif interacted with U20, A19, and U18 on the primer RNA strand. The lead PNA analog (L14) showed superior binding free energy to both RdRp (-47.26 kcal/mol) and RdRp-RNA (-85.66 kcal/mol), outperforming Remdesivir. Key amino acid residues critical for binding affinity were identified, providing valuable insights for drug development. This promising PNA-mimetic compound offers dual-target specificity, presenting a compelling avenue for developing potent anti-SARS-CoV-2 agents.
Collapse
Affiliation(s)
- Kiran Shehzadi
- Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Iqra Kalsoom
- Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Ming-Jia Yu
- Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Jian-Hua Liang
- Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
9
|
Luong QXT, Hoang PT, Ho PT, Ayun RQ, Lee TK, Lee S. Potential Broad-Spectrum Antiviral Agents: A Key Arsenal Against Newly Emerging and Reemerging Respiratory RNA Viruses. Int J Mol Sci 2025; 26:1481. [PMID: 40003946 PMCID: PMC11855616 DOI: 10.3390/ijms26041481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
Respiratory viral infections present significant global health challenges, causing substantial morbidity and mortality, particularly among highly susceptible components of the population. The emergence of pandemics and epidemics, such as those caused by influenza viruses and coronaviruses, emphasizes the urgent need for effective antiviral therapeutics. In this review, we explore the potential of broad-spectrum antiviral agents targeting respiratory RNA viruses, including influenza viruses, coronaviruses, respiratory syncytial virus, human metapneumovirus, human parainfluenza viruses, and rhinoviruses. Various broad-spectrum direct-acting and host-targeting antivirals are discussed, including monoclonal antibodies targeting conserved regions of viral surface proteins, molecules interfering with host cell receptors or viral replication machinery, viral protease inhibitors, siRNA therapies, ribonuclease, and 3D8 scFv. Advancements in host-targeting approaches to reduce resistance and RNA-based therapeutics offer significant potential for combating respiratory viral threats. Despite challenges, broad-spectrum antiviral agents represent a crucial strategy, particularly when specific viral pathogens are unidentified or rapid intervention is essential, such as during pandemics or outbreaks.
Collapse
Affiliation(s)
- Quynh Xuan Thi Luong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Q.X.T.L.); (P.T.H.); (P.T.H.); (R.Q.A.)
| | - Phuong Thi Hoang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Q.X.T.L.); (P.T.H.); (P.T.H.); (R.Q.A.)
| | - Phuong Thi Ho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Q.X.T.L.); (P.T.H.); (P.T.H.); (R.Q.A.)
| | - Ramadhani Qurrota Ayun
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Q.X.T.L.); (P.T.H.); (P.T.H.); (R.Q.A.)
| | - Taek Kyun Lee
- Risk Assessment Research Center, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Q.X.T.L.); (P.T.H.); (P.T.H.); (R.Q.A.)
| |
Collapse
|
10
|
Wilders H, Biggs G, Rowe SM, Cawood EE, Riziotis IG, Rendina AR, Grant EK, Pettinger J, Fallon DJ, Skehel M, House D, Tomkinson NCO, Bush JT. Expedited SARS-CoV-2 Main Protease Inhibitor Discovery through Modular 'Direct-to-Biology' Screening. Angew Chem Int Ed Engl 2025; 64:e202418314. [PMID: 39630105 DOI: 10.1002/anie.202418314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/15/2024] [Indexed: 12/12/2024]
Abstract
Reactive fragment (RF) screening has emerged as an efficient method for ligand discovery across the proteome, irrespective of a target's perceived tractability. To date, however, the efficiency of subsequent optimisation campaigns has largely been low-throughput, constrained by the need for synthesis and purification of target compounds. We report an efficient platform for 'direct-to-biology' (D2B) screening of cysteine-targeting chloroacetamide RFs, wherein synthesis is performed in 384-well plates allowing direct assessment in downstream biological assays without purification. Here, the developed platform was used to optimise inhibitors of SARS-CoV-2 main protease (MPro), an established drug target for the treatment of COVID-19. An initial RF hit was developed into a series of potent inhibitors, and further exploration using D2B screening enabled a 'switch' to a reversible inhibitor series. This example of ligand discovery for MPro illustrates the acceleration that D2B chemistry can offer for optimising RFs towards covalent inhibitor candidates, as well as providing future impetus to explore the evolution of RFs into non-covalent ligands.
Collapse
Affiliation(s)
- Harry Wilders
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| | - George Biggs
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Sam M Rowe
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Emma E Cawood
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Ioannis G Riziotis
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Alan R Rendina
- Screening, Profiling and Mechanistic Biology, GSK, 1250 South Collegeville Road, Collegevill, PA, 19426, US
| | - Emma K Grant
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Jonathan Pettinger
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - David J Fallon
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Mark Skehel
- Proteomics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - David House
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Nicholas C O Tomkinson
- Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| | - Jacob T Bush
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| |
Collapse
|
11
|
Tripathi SM, Akash S, Rahman MA, Sundriyal S. Identification of synthetically tractable MERS-CoV main protease inhibitors using structure-based virtual screening and molecular dynamics potential of mean force (PMF) calculations. J Biomol Struct Dyn 2025; 43:787-797. [PMID: 37978909 DOI: 10.1080/07391102.2023.2283780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/01/2023] [Indexed: 11/19/2023]
Abstract
The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a potentially lethal infection that presents a substantial threat to health, especially in Middle East nations. Given that no FDA-approved specific therapy for MERS infection exists, designing and discovering a potent antiviral therapy for MERS-CoV is crucial. One pivotal strategy for inhibiting MERS replication is to focus on the viral main protease (Mpro). In this study, we identify potential novel Mpro inhibitors employing structure-based virtual screening of our recently reported Ugi reaction-derived library (URDL) consisting of cherry-picked molecules from the literature. The key features of the URDL library include synthetic tractability (1-2 pot synthesis) of the molecules scaffold and unexplored chemical space. The hits were ranked based on the docking score, MM-GBSA free energy of binding, and the interaction pattern with the active site residues. A molecular dynamics (MD) simulation study was performed for the first two top-ranked compounds to analyze the stability and free binding energy based on the molecular mechanics Poisson-Boltzmann surface area. The potential mean force calculated from the steered molecular dynamics (SMD) simulations of the hits indicates improved H-bond potential, enhanced conformational stability, and binding affinity toward the target, compared to the cocrystallized ligand. The discovered hits represent novel synthetically tractable scaffolds as potential MERS-CoV Mpro inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shailesh Mani Tripathi
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Rajasthan, India
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Ashulia, Bangladesh
| | | | - Sandeep Sundriyal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Rajasthan, India
| |
Collapse
|
12
|
Liang L, Meng Y, Chang X, Li E, Huang Y, Yan L, Lou Z, Peng Y, Zhu B, Yu W, Chang J. Discovery of a 2'-α-Fluoro-2'-β- C-(fluoromethyl) Purine Nucleotide Prodrug as a Potential Oral Anti-SARS-CoV-2 Agent. J Med Chem 2025; 68:1994-2007. [PMID: 39804580 DOI: 10.1021/acs.jmedchem.4c02769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
A novel 2'-α-fluoro-2'-β-C-(fluoromethyl) purine nucleoside phosphoramidate prodrug 15 has been designed and synthesized to treat SARS-CoV-2 infection. The SARS-CoV-2 central replication transcription complex (C-RTC, nsp12-nsp7-nsp82) catalyzed in vitro RNA synthesis was effectively inhibited by the corresponding bioactive nucleoside triphosphate (13-TP). The cryo-electron microscopy structure of the C-RTC:13-TP complex was also determined. Compound 15 exhibited potent in vitro antiviral activity against the SARS-CoV-2 20SF107 strain (EC50 = 0.56 ± 0.06 μM) and the Omicron BA.5 variant (EC50 = 0.96 ± 0.23 μM) with low cytotoxicity. Furthermore, it was well tolerated in rats at doses of up to 2000 mg/kg, and a single oral dose of this prodrug at 40 mg/kg led to high levels of 13-TP in the target organ lungs of rats with a long half-life. These findings support the further development of compound 15 as an orally available antiviral agent for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lan Liang
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, NMPA Key Laboratory for Research and Evaluation of Innovative Drug, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Yonggang Meng
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaoyu Chang
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ertong Li
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yucen Huang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences and College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Liming Yan
- MOE Key Laboratory of Protein Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhiyong Lou
- MOE Key Laboratory of Protein Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Youmei Peng
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Bo Zhu
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, NMPA Key Laboratory for Research and Evaluation of Innovative Drug, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Wenquan Yu
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junbiao Chang
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, NMPA Key Laboratory for Research and Evaluation of Innovative Drug, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
13
|
Kumar M, Baig MS, Bhardwaj K. Advancements in the development of antivirals against SARS-Coronavirus. Front Cell Infect Microbiol 2025; 15:1520811. [PMID: 39917633 PMCID: PMC11798951 DOI: 10.3389/fcimb.2025.1520811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) caused an outbreak in 2002-2003, spreading to 29 countries with a mortality rate of about 10%. Strict quarantine and infection control methods quickly stopped the spread of the disease. Later research showed that SARS-CoV came from animals (zoonosis) and stressed the possibility of a similar spread from host to human, which was clearly shown by the COVID-19 outbreak. The COVID-19 pandemic, instigated by SARS-CoV-2, has affected 776 million confirmed cases and more than seven million deaths globally as of Sept 15, 2024. The existence of animal reservoirs of coronaviruses continues to pose a risk of re-emergence with improved fitness and virulence. Given the high death rate (up to 70 percent) and the high rate of severe sickness (up to 68.7 percent in long-COVID patients), it is even more critical to identify new therapies as soon as possible. This study combines research on antivirals that target SARS coronaviruses that have been conducted over the course of more than twenty years. It is a beneficial resource that might be useful in directing future studies.
Collapse
Affiliation(s)
- Mrityunjay Kumar
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, India
| | - Mirza Sarwar Baig
- Centre for Virology, School of Interdisciplinary Science and Technology, Jamia Hamdard, New Delhi, India
| | - Kanchan Bhardwaj
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| |
Collapse
|
14
|
Paróczai D, Bikov A, Blidaru A, Bobu E, Lascu A, Mot CI, Mihaicuta S, Frent S. Comparative efficacy of repurposed drugs lopinavir-ritonavir and darunavir-ritonavir in hospitalised COVID-19 patients: insights from a tertiary centre cohort. Front Cell Infect Microbiol 2025; 14:1496176. [PMID: 39885967 PMCID: PMC11779713 DOI: 10.3389/fcimb.2024.1496176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Background Drug repurposing has become a widely adopted strategy to minimise research time, costs, and associated risks. Combinations of protease inhibitors such as lopinavir and darunavir with ritonavir have been repurposed as treatments for COVID-19. Although lopinavir-ritonavir (LPV/r) and darunavir-ritonavir (DRV/r) have shown in vitro efficacy against COVID-19, the results in human studies have been inconsistent. Therefore, our objective was to compare the efficacy of LPV/r and DRV/r in COVID-19 patients admitted to a tertiary centre in Romania. Research design and methods A clinical dataset from 417 hospitalised patients was analysed. Patients were assigned to the LPV/r, DRV/r, or control (standard-of-care) group based on clinical decisions made by the attending infectious disease specialists, aligned with national treatment protocols. Kaplan-Meier and Cox proportional hazards regression analyses were conducted to compare in-hospital mortality and to identify factors associated with clinical improvement or fatal outcomes. Results By day 10, more patients showed improvement with LPV/r and DRV/r (p=0.03 and 0.01, respectively), but only LPV/r was associated with improved survival compared to the control group (p=0.05). Factors associated with mortality included male gender (HR: 3.63, p=0.02), diabetes (HR: 2.49, p=0.03), oxygen saturation below 90% at admission (HR: 5.23, p<0.01), high blood glucose levels (HR: 3.68, p=0.01), age (HR: 1.04, p=0.02), and more than 25% lesion extension on chest CT scan (HR: 2.28, p=0.03). Conclusions LPV/r, but not DRV/r, showed a survival benefit in patients hospitalised with COVID-19, but these findings deserve further investigation in a randomised clinical trial.
Collapse
Affiliation(s)
- Dóra Paróczai
- Department of Medical Microbiology, University of Szeged, Szeged, Hungary
- Albert Szent-Györgyi Health Center, Pulmonology Clinic, University of Szeged, Deszk, Hungary
| | - András Bikov
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Andreea Blidaru
- Department of Infectious Diseases, Infectious Diseases and Pulmonology Clinical Hospital, Timisoara, Romania
| | - Emanuel Bobu
- Department of Pulmonology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Ana Lascu
- Department of Functional Sciences, Discipline of Pathophysiology, Centre for Translational Research and Systems Medicine, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Institute for Cardiovascular Diseases of Timisoara, Clinic for Cardiovascular Surgery, Timisoara, Romania
| | - Cristian Ion Mot
- ENT Department, Municipal Emergency Hospital Timisoara, Timisoara, Romania
- Department of Surgery, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Stefan Mihaicuta
- Department of Infectious Diseases, Infectious Diseases and Pulmonology Clinical Hospital, Timisoara, Romania
- Centre for Research and Innovation in Precision Medicine of Respiratory Diseases, Department of Pulmonology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Stefan Frent
- Department of Infectious Diseases, Infectious Diseases and Pulmonology Clinical Hospital, Timisoara, Romania
- Centre for Research and Innovation in Precision Medicine of Respiratory Diseases, Department of Pulmonology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| |
Collapse
|
15
|
Bai Y, Liu T, Zhang S, Shi Y, Yang Y, Ding M, Yang X, Guo S, Xu X, Liu Q. Traditional Chinese Medicine for Viral Pneumonia Therapy: Pharmacological Basis and Mechanistic Insights. Int J Biol Sci 2025; 21:989-1013. [PMID: 39897040 PMCID: PMC11781171 DOI: 10.7150/ijbs.105086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025] Open
Abstract
Different respiratory viruses might cause similar symptoms, ranging from mild upper respiratory tract involvement to severe respiratory distress, which can rapidly progress to septic shock, coagulation disorders, and multiorgan failure, ultimately leading to death. The COVID-19 pandemic has shown that predicting clinical outcomes can be challenging because of the complex interactions between the virus and the host. Traditional Chinese medicine (TCM) has distinct benefits in the treatment of respiratory viral illnesses due to its adherence to the principles of "different treatments for the same disease" and "same treatment for different diseases". This paper examines the effectiveness and underlying mechanisms of key TCM treatments for viral pneumonia in recent years. The aim of this study was to discover and confirm the active substances of TCM with potential therapeutic effects on viral pneumonia and their integrative effects and synergistic mechanisms and to provide a scientific basis for elucidating the effectiveness of TCM treatment and drug discovery. Furthermore, a thorough analysis of previous research is necessary to evaluate the effectiveness of TCM in treating viral pneumonia.
Collapse
Affiliation(s)
- Yinglu Bai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Chinese Medicine, Beijing 100010, China
| | - Tengwen Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shuwen Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yifan Shi
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yumei Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Maoyu Ding
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaowei Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shanshan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Chinese Medicine, Beijing 100010, China
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
- Beijing Institute of Chinese Medicine, Beijing 100010, China
| |
Collapse
|
16
|
Koze H, Sudoh M, Onitsuka S, Okamura H, Ishikawa T, Tani F, Miyata-Yabuki Y, Shirouzu M, Baba M, Okamoto M, Hamada T. Sulfoquinovosyl diacylglycerol, a component of Holy Basil Ocimum tenuiflorum, inhibits the activity of the SARS-CoV-2 main protease and viral replication in vitro. J Nat Med 2025; 79:122-133. [PMID: 39585602 PMCID: PMC11735596 DOI: 10.1007/s11418-024-01855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 11/26/2024]
Abstract
The persistence of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the emergence of new mutant strains continue to present a substantial threat with potential for future pandemics. Safe, effective, and readily available COVID-19 therapeutics are urgently needed to prepare for future coronavirus pandemics. To help identify new antiviral agents, the present study focused on natural products in the extracts of Holy Basil, Ocimum tenuiflorum L., which show potential inhibitory effects against the SARS-CoV-2 main protease (Mpro). Bioassay-guided isolation of the MeOH extracts of O. tenuiflorum led to the identification of a sulfur-containing glyceroglycolipid, sulfoquinovosyl diacylglycerol (SQDG: 1), as a potent Mpro inhibitor that effectively inhibited Mpro activity (IC50: 0.42 µM). SQDG (1) also markedly suppressed SARS-CoV-2 replication (EC50, 51.2 µM) in vitro while displaying no cytotoxicity (CC50 > 100 µM). Further inhibition kinetic studies and docking simulations clearly demonstrated that SQDG strongly inhibited SARS-CoV-2 Mpro in a competitive and mixed-inhibition manner. These findings highlight SQDG as a promising lead compound for COVID-19 therapy and emphasize the need to explore new drugs from natural sources.
Collapse
Affiliation(s)
- Hinako Koze
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan
- Faculty of Science, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan
| | - Masayuki Sudoh
- Department of Translational Research, Joint Research Center for Human Retrovirus Infection, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Satoaki Onitsuka
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan
- Faculty of Science, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan
| | - Hiroaki Okamura
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan
- Faculty of Science, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan
| | - Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima, 890-0065, Japan
| | - Fumito Tani
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yukako Miyata-Yabuki
- Drug Discovery Structural Biology Platform Unit, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Mikako Shirouzu
- Drug Discovery Structural Biology Platform Unit, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Masanori Baba
- Division of Infection Control Research, Center for Advanced Science Research and Promotion, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-8580, Japan
| | - Mika Okamoto
- Division of Infection Control Research, Center for Advanced Science Research and Promotion, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-8580, Japan
| | - Toshiyuki Hamada
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan.
- Faculty of Science, Kagoshima University, 1-21-35 Korimoto, Kagoshima, 890-0065, Japan.
| |
Collapse
|
17
|
Uthumange SS, Liew AJH, Chee XW, Yeong KY. Ringing medicinal chemistry: The importance of 3-membered rings in drug discovery. Bioorg Med Chem 2024; 116:117980. [PMID: 39536361 DOI: 10.1016/j.bmc.2024.117980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Scaffold-based drug design has become increasingly prominent in the pharmaceutical field due to the systematic and effective approach through which it facilitates the development of novel drugs. The identification of key scaffolds provides medicinal chemists with a fundamental framework for subsequent research. With mounting evidence suggesting that increased aromaticity could impede the chances of developmental success for oral drug candidates, there is an imperative need for a more thorough exploration of alternative ring systems to mitigate attrition risks. The unique characteristics exhibited by three-membered rings have led to their application in medicinal chemistry. This review explores the use of cyclopropane-, aziridine-, thiirane-, and epoxide-containing compounds in drug discovery, focusing on their roles in approved medicines and drug candidates. Specifically, the importance of the three-membered ring systems in rending biological activity for each drug molecule was highlighted. The undeniable therapeutic value and intriguing features presented by these compounds suggest significant pharmacological potential, providing justification for their incorporation into the design of novel drug candidates.
Collapse
Affiliation(s)
- Sahani Sandalima Uthumange
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor, Malaysia
| | - Angie Jun Hui Liew
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor, Malaysia
| | - Xavier Wezen Chee
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Keng Yoon Yeong
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor, Malaysia.
| |
Collapse
|
18
|
Zakaria MY, Elmaaty AA, El-Shesheny R, Alnajjar R, Kutkat O, Ben Moussa S, Abdullah Alzahrani AY, El-Zahaby SA, Al-Karmalawy AA. Biological and computational assessments of thiazole derivative-reinforced bile salt enriched nano carriers: a new gate in targeting SARS-CoV-2 spike protein. RSC Adv 2024; 14:38778-38795. [PMID: 39654925 PMCID: PMC11627215 DOI: 10.1039/d4ra07316a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
There is merit in investigating novel therapeutic molecules that hit vital targets during the viral infection cycle i.e. disrupting the interaction between SARS-CoV-2's spike glycoprotein and the host's angiotensin converting enzyme 2 (ACE2) receptor, potentially offering new avenues for treatment. Accordingly, lipid-based vesicular systems like liposomes or niosomes are frequently utilized to overcome these hurdles. Thus, chemically synthesized compounds were encapsulated within PEGylated bilosomes (PBs) to improve their solubility and intestinal permeability, thereby enhancing their anti-SARS-CoV-2 effectiveness. The formulae were prepared according to 23 full factorial design which was also used to explore the impact of the change in predetermined formulation variables on the properties of the prepared vesicles (entrapment efficiency EE%, particle size PS, and zeta potential ZP). Additionally, the optimized formula (F4) which is composed of 3% bile salt (BS), 40 mg 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPE) and sodium deoxycholate (SDC) as a bile salt, was selected as an optimum formula with desirability value 0.674 using Design Expert® software. Both the in vitro release and ex vivo experiments results confirmed the significant superiority of the F4 over the drug dispersion. Both cytotoxicity and anti-SARS-CoV-2 activity of all examined compound-loaded PBs (PB3a-PB3g) were assessed in Vero E6 cells via MTT assay. Both compounds PB3c and PB3g displayed the highest IC50 values (0.71 and 1.25 μg mL-1, respectively) ensuring their superior antiviral potential. Moreover, it was revealed that PB3c demonstrated more than 80% virucidal activity and over 80% inhibition of viral adsorption with little effect on the viral replication ∼(5-10%). Moreover, molecular docking and dynamic studies were conducted to pursue the binding affinities of the investigated compounds towards the ACE2 target of the SARS-CoV-2 spike protein, assuring their feasible inhibitory potential. Collectively, the investigated compound-loaded PBs can be treated as promising lead drug delivery panels for COVID-19 management.
Collapse
Affiliation(s)
- Mohamed Y Zakaria
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Salman International University Ras Sudr 46612 South Sinai Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Port Said University Port Said 42526 Egypt
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University Port Said 42526 Egypt
- Medicinal Chemistry Department, Clinical Pharmacy Program, East Port Said National University Port Said 42526 Egypt
| | - Rabeh El-Shesheny
- Center of Scientific Excellence for Influenza Viruses, Water Pollution Research Department, Environmental Research Institute, National Research Centre Dokki-Giza 12622 Egypt
| | - Radwan Alnajjar
- CADD Unit, Faculty of Pharmacy, Libyan International Medical University Benghazi 16063 Libya
- Department of Chemistry, Faculty of Science, University of Benghazi Benghazi 16063 Libya
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, Water Pollution Research Department, Environmental Research Institute, National Research Centre Dokki-Giza 12622 Egypt
| | - Sana Ben Moussa
- Department of Chemistry, Faculty of Science and Arts, King Khalid University Mohail Assir Saudi Arabia
| | | | - Sally A El-Zahaby
- Department of Pharmaceutics and Industrial Pharmacy, PharmD Program, Egypt-Japan University of Science and Technology (E-Just) Alexandria Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq Baghdad 10023 Iraq
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt New Damietta 34518 Egypt
| |
Collapse
|
19
|
Li M, Wu Y, Li B, Lu C, Jian G, Shang X, Chen H, Huang J, He B. ACVPICPred: Inhibitory activity prediction of anti-coronavirus peptides based on artificial neural network. Comput Struct Biotechnol J 2024; 23:3625-3633. [PMID: 39469670 PMCID: PMC11513478 DOI: 10.1016/j.csbj.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Peptides, as small molecular compounds, exhibit prominent advantages in the inhibition of coronaviruses due to their safety, efficacy, and specificity, holding great promise as drugs against coronaviruses. The rapid and efficient determination of the activity of anti-coronavirus peptides (ACovPs) can greatly accelerate the development of drugs for treating coronavirus-related diseases. Hence, we present ACVPICPred, a computational model designed to predict the inhibitory activity of ACovPs based on their sequences and structural information. By leveraging bioinformatics tools AlphaFold3 for structural predictions and several feature extraction methods, the model integrates both sequence and structural features to enhance prediction accuracy. To address the limitations of existing datasets, we employed data augmentation techniques, including the introduction of noise and the SMOGN, to improve the model robustness. The model's performance was evaluated through five-fold cross-validation, achieving a Pearson correlation coefficient of 0.7668 (p < 0.05) and an R² of 0.5880 on the training dataset. Overall, in our study, compared to models that only use sequence features, models that combine structural features have achieved more robust results in various evaluation metrics. ACVPICPred is freely accessible at the following URL: http://i.uestc.edu.cn/acvpICPred/main/Main.php.
Collapse
Affiliation(s)
- Min Li
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Yifei Wu
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Bowen Li
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Chunying Lu
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Guifen Jian
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Xing Shang
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Heng Chen
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, No.2006, Xiyuan Ave, West Hi‑Tech Zone, Chengdu 6173001, Sichuan, China
| | - Bifang He
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
- State Key Laboratory of Public Big Data, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| |
Collapse
|
20
|
Guo L, Zeng P, Zhou X, Li W, Zhang J, Li J. Structural basis of main proteases of MERS-CoV bound to antineoplastic drug carmofur. Biochem Biophys Res Commun 2024; 735:150469. [PMID: 39106601 DOI: 10.1016/j.bbrc.2024.150469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 07/21/2024] [Accepted: 07/28/2024] [Indexed: 08/09/2024]
Abstract
Recurrent epidemics of coronaviruses have posed significant threats to human life and health. The mortality rate of patients infected with the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is 35 %. The main protease (Mpro) plays a crucial role in the MERS-CoV life cycle, and Mpro exhibited a high degree of conservation among different coronaviruses. Therefore inhibition of Mpro has become an effective strategy for the development of broad-spectrum anti-coronaviral drugs. The inhibition of SARS-CoV-2 Mpro by the anti-tumor drug carmofur has been revealed, but structural studies of carmofur in complex with Mpro from other types of coronavirus have not been reported. Hence, we revealed the structure of the MERS-CoV Mpro-carmofur complex, analysed the structural basis for the binding of carmofur to MERS-CoV Mpro in detail, and compared the binding patterns of carmofur to Mpros of two different coronaviruses, MERS-CoV and SARS-CoV-2. Considering the importance of Mpros for coronavirus therapy, structural understanding of Mpro inhibition by carmofur could contribute to the design and development of novel antiviral drugs with safe and broad-spectrum efficacy.
Collapse
Affiliation(s)
- Li Guo
- Shenzhen Crystalo Biopharmaceutical Co., Ltd., Shenzhen, China
| | - Pei Zeng
- Jiangxi Jmerry Biopharmaceutical Co., Ltd., Ganzhou, China
| | - Xuelan Zhou
- Jiangxi Jmerry Biopharmaceutical Co., Ltd., Ganzhou, China
| | - Wenwen Li
- Jiangxi Jmerry Biopharmaceutical Co., Ltd., Ganzhou, China
| | - Jin Zhang
- Shenzhen Crystalo Biopharmaceutical Co., Ltd., Shenzhen, China; Jiangxi Jmerry Biopharmaceutical Co., Ltd., Ganzhou, China.
| | - Jian Li
- Shenzhen Crystalo Biopharmaceutical Co., Ltd., Shenzhen, China; Jiangxi Jmerry Biopharmaceutical Co., Ltd., Ganzhou, China.
| |
Collapse
|
21
|
Wang W, Tan Y, Mao J, Xiong W. Swertiamarin and sweroside are potential inhibitors of COVID-19 based on the silico analysis. Medicine (Baltimore) 2024; 103:e40425. [PMID: 39533611 PMCID: PMC11556981 DOI: 10.1097/md.0000000000040425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
The severity of the respiratory disease caused by the severe acute respiratory syndrome coronavirus 2 has escalated rapidly in recent years, posing a significant threat to global health. Sweroside and swertiamarin are bioactive iridoid glycosides extracted mainly from Swertia davidii Franch. It remains unclear how Swertia davidii Franch. Specifically affects COVID-19 and its underlying mechanisms. We first employed network pharmacology and molecular docking techniques to investigate how sweroside and swertiamarin affect COVID-19 in order to explore its potential mechanism. We found that 35 potential target genes can be used for the treatment of COVID-19, with androgen receptor (AR), HSP90AA1, RAC-alpha serine/threonine-protein kinase, cyclin-dependent kinase 1, epidermal growth factor receptor, and glycogen synthase kinase-3 beta emerging as particularly promising candidates. Additionally, sweroside and swertiamarin demonstrated unambiguous interactions with the 3CL protease AR through molecular docking research. At the active site, sweroside and swertiamarin can bind to AR (1T65), the main protease (5R82), and 3CL protease (6M2N), showing therapeutic potential.
Collapse
Affiliation(s)
- Wenxiang Wang
- Chongqing Three Gorges Medical College, Chongqing, China
| | - Ying Tan
- Chongqing Three Gorges Medical College, Chongqing, China
| | - Jingxin Mao
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Wei Xiong
- Chongqing Three Gorges Medical College, Chongqing, China
| |
Collapse
|
22
|
Li R, Zhang W, Huang B, Sun G, Xie Y, Song J, Wang S, Du G. Dayuan Yin alleviates symptoms of HCoV-229E-induced pneumonia and modulates the Ras/Raf1/MEK/ERK pathway. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:58. [PMID: 39495380 PMCID: PMC11534925 DOI: 10.1007/s13659-024-00474-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/15/2024] [Indexed: 11/05/2024]
Abstract
Viral pneumonia is characterized by inflammation in the lungs triggered by respiratory viruses. Dayuan Yin (DYY), a traditional Chinese medicine formula known for treating infectious diseases, is hypothesized to offer therapeutic benefits in treating viral pneumonia, although its specific molecular impacts remain understudied. This study aimed to evaluate the therapeutic effects of DYY in mitigating HCoV-229E virus-induced pneumonia in mice. This study employed an HCoV-229E virus-infected mouse model to investigate the therapeutic potential and underlying molecular mechanisms of DYY on virus-induced pneumonia. The respiratory function and organ indices post-treatment were assessed. Lung tissue and tracheal lesions were evaluated via immunohistochemistry. Spleen immune cell composition was analyzed using flow cytometry. Inflammatory cytokines and viral loads were quantified using hypersensitive multiplex electrochemiluminescence method and PCR analysis, respectively. The expression levels of MAS1, Ras, Raf1, MEK1/2, and ERK1/2 in lung tissues were determined through western blot analysis. DYY significantly improved respiratory function, and reduced organ pathology in infected mice. It effectively decreased viral loads and inflammatory cytokines such as IL-6, IL-1β, and TNF-α in lung tissues. Enhancements in immune response were evidenced by increased CD4/CD8 ratios in the spleen. DYY also notably upregulated MAS1 protein levels and suppressed the activation of the Ras/Raf1/MEK/ERK signaling pathway. DYY enhanced respiratory function and exerted significant antiviral and immunomodulatory effects in mice infected with the HCoV-229E virus, primarily by modulating MAS1 expression and inhibiting the Ras/Raf1/MEK/ERK pathway.
Collapse
Affiliation(s)
- Rui Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | | | - Bei Huang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Guotong Sun
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yifei Xie
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Junke Song
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shumei Wang
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
23
|
Dehghan M, Askari H, Tohidfar M, Siadat S, Fatemi F. Improvement of RBD-FC Immunogenicity by Using Alum-Sodium Alginate Adjuvant Against SARS-COV-2. Influenza Other Respir Viruses 2024; 18:e70018. [PMID: 39478310 PMCID: PMC11525037 DOI: 10.1111/irv.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Adjuvants use several mechanisms to boost immunogenicity and to modulate immune response. The strength of adsorption of antigen by adjuvants can be a determinant factor for significant improvement of immunopotentiation. METHODS We expressed recombinant RBD-FC in PichiaPink Strain 4 and examined the vaccination of mice by vaccine formulation with different adjuvants (sodium alginate and aluminum hydroxide, alone and together). RESULTS Sodium alginate significantly increased the immunogenicity and stability of RBD-FC antigen, so RBD-FC formulated with combined alginate and alum (AlSa) and sodium alginate alone showed higher antibody titer and stability. Immunogenicity of RBD-FC:AlSa was determined by serological assays including direct enzyme-linked immunosorbent assay (ELISA) and surrogate virus neutralization test (sVNT). High levels of IgGs and neutralizing antibodies were measured in serum of mice immunized with the RBD-FC:AlSa formulation. On the other hand, cytokines IL-10 and INF-γ were severely accumulated in response to RBD-FC:AlSa, and after 10 days, their accumulation was significantly declined, whereas IL-4 showed the highest and the lowest accumulation in response to alum and alginate, respectively. CONCLUSIONS Our data may suggest that combination of alum and sodium alginate has a better compatibility with RBD-FC in vaccine formulation.
Collapse
MESH Headings
- Alginates/chemistry
- Animals
- Mice
- Alum Compounds/administration & dosage
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- SARS-CoV-2/immunology
- Mice, Inbred BALB C
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Female
- Adjuvants, Vaccine
- COVID-19/prevention & control
- COVID-19/immunology
- Immunogenicity, Vaccine
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/immunology
- Aluminum Hydroxide/chemistry
- Humans
- Immunoglobulin G/blood
- Cytokines
- Immunoglobulin Fc Fragments/immunology
Collapse
Affiliation(s)
- Mahboobeh Dehghan
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| | - Hossein Askari
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| | - Masoud Tohidfar
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| | | | - Fataneh Fatemi
- Protein Research CenterShahid Beheshti UniversityTehranIran
| |
Collapse
|
24
|
Bashir A, Li S, Ye Y, Zheng Q, Knanghat R, Bashir F, Shah NN, Yang D, Xue M, Wang H, Zheng C. SARS-CoV-2 S protein harbors furin cleavage site located in a short loop between antiparallel β-strand. Int J Biol Macromol 2024; 281:136020. [PMID: 39368587 DOI: 10.1016/j.ijbiomac.2024.136020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/22/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Furin cleavage site (FCS) of the SARS-CoV-2 S protein, which connects the S1/S2 junction, is essential for facilitating fusion with the host cells. Wild-type (Wt) SARS-CoV-2 S protein, PDB ID: 6yvb, lacks a sequence of amino acid residues, including the FCS that links the S1/S2 junction. For the first time, we demonstrated that a stretch of 14 amino acid residues (677QTNSPRRARSVASQ689) forms an antiparallel β-sheet comprising of PRRAR sequence in the FCS within a short loop. Upon comparing the loop content of the S1/S2 junction with that of Wt SARS-CoV-2 containing PRRAR in the FCS, we observed a decrease in antiparallel β-sheet content and an increase in loop content in the B.1.1.7 variant with HRRAR in the FCS. This short loop within antiparallel β-sheet can serve as a docking site for various proteases, including TMPRSS2 and α1AT. We performed a 300-ns simulation of the SARS-CoV-2 receptor binding domain (RBD) using several antibacterial and antiviral ligands commonly used to treat various infections. Our findings indicate that the receptor binding domain (RBD) comprising the receptor binding motif (RBM) utilizes β6 and a significant portion of the loop to bind with ligands, suggesting its potential for treating SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Arif Bashir
- Department of Clinical Biochemistry & Biotechnology, Government College for Women, Nawa-Kadal, Srinagar 190002, India
| | - Shun Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yu Ye
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350004, China
| | - Rajani Knanghat
- Department of Biotechnology, Indian Institute of Technology, Chennai 600036, India
| | - Fahim Bashir
- Department of Environmental Science, University of Kashmir, 190006, India
| | - Naveed Nazir Shah
- Department of Chest Medicine, Government Medical College, Srinagar, Jammu and Kashmir 190001, India
| | - Debin Yang
- Department of Pediatrics, Children's Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
25
|
Islam MA, Pathak K, Saikia R, Pramanik P, Das A, Talukdar P, Shakya A, Ghosh SK, Singh UP, Bhat HR. An in-depth analysis of COVID-19 treatment: Present situation and prospects. Arch Pharm (Weinheim) 2024; 357:e2400307. [PMID: 39106224 DOI: 10.1002/ardp.202400307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 08/09/2024]
Abstract
Coronavirus disease 2019 (COVID-19) the most contagious infection caused by the unique type of coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), produced a global pandemic that wreaked havoc on the health-care system, resulting in high morbidity and mortality. Several methods were implemented to tackle the virus, including the repurposing of existing medications and the development of vaccinations. The purpose of this article is to provide a complete summary of the current state and future possibilities for COVID-19 therapies. We describe the many treatment classes, such as antivirals, immunomodulators, and monoclonal antibodies, that have been repurposed or developed to treat COVID-19. We also looked at the clinical evidence for these treatments, including findings from observational studies and randomized-controlled clinical trials, and highlighted the problems and limitations of the available evidence. Furthermore, we reviewed existing clinical trials and prospective COVID-19 therapeutic options, such as novel medication candidates and combination therapies. Finally, we discussed the long-term consequences of COVID-19 and the importance of ongoing research into the development of viable treatments. This review will help physicians, researchers, and policymakers to understand the prevention and mitigation of COVID-19.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Pallab Pramanik
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Aparoop Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Prasenjit Talukdar
- Department of Petroleum Engineering, DUIET, Dibrugarh, University, Assam, India
| | - Anshul Shakya
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Surajit Kumar Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Udaya Pratap Singh
- Drug Design & Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh, India
| | - Hans Raj Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| |
Collapse
|
26
|
Hue BTB, Nguyet Huong Giang H, Nguyen CQ, Chou FP, La Duc Thanh D, Tran QD, Hieu VT, Hoang Phuong Mai L, Lin HC, Wu TK. Discovery of a novel benzimidazole conjugated quinazolinone derivative as a promising SARS-CoV-2 3CL protease inhibitor. RSC Adv 2024; 14:33820-33829. [PMID: 39450066 PMCID: PMC11500731 DOI: 10.1039/d4ra03267e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/02/2024] [Indexed: 10/26/2024] Open
Abstract
This report presents the design and synthesis of quinazolinone-based derivatives as promising SARS-CoV-2 3CL protease inhibitors. Two novel series, namely, febrifugine analogues 4a-i and quinazolinone conjugated benzimidazoles 9a-c, were successfully synthesized starting from isatoic anhydride. The synthesized quinazolinone derivatives were evaluated for their cytotoxicity against cancer cell lines and SARS-CoV-2 3CL inhibitory activity. The results showed that the synthesized compounds did not have significant toxicity for the non-cancer HEK293 cell line and MCF-7, MDA-MB-231, HEPG2 and HEPG2.2.15 cancer cell lines. Notably, compound 9b exhibited anti-3CL enzymatic activity in a dose-dependent manner, with the calculated IC50 value of 10.73 ± 1.17 μM. Docking results highlighted the interaction between 9b and 3CL protease through hydrogen bonding with key amino acids, including His41, Met49, Cys145, Met165, Arg188, His164, and Glu166, at the active site of the protease. Pharmacokinetic studies and ADME analyses provide valuable insights into the potential of compound 9b as a drug candidate. These findings support the new scaffold as a candidate for 3CLpro inhibition and advanced anti-coronavirus drug research.
Collapse
Affiliation(s)
- Bui Thi Buu Hue
- College of Natural Sciences, Can Tho University Can Tho 94000 Vietnam
| | - Huynh Nguyet Huong Giang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University Hsinchu 30010 Taiwan
| | - Cuong Quoc Nguyen
- College of Natural Sciences, Can Tho University Can Tho 94000 Vietnam
| | - Feng-Pai Chou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University Hsinchu 30010 Taiwan
- Center for Emergent Functional Matter Science, National Yang Ming Chiao Tung University 1001 Ta-Hsueh Rd. Hsinchu 30010 Taiwan
| | - Danh La Duc Thanh
- College of Natural Sciences, Can Tho University Can Tho 94000 Vietnam
- Department of Material Science, National Yang Ming Chiao Tung University 1001 Ta-Hsueh Rd. Hsinchu 30010 Taiwan
| | - Quang De Tran
- College of Natural Sciences, Can Tho University Can Tho 94000 Vietnam
| | - Vo Trung Hieu
- College of Natural Sciences, Can Tho University Can Tho 94000 Vietnam
| | | | - Hong-Cheu Lin
- Department of Material Science, National Yang Ming Chiao Tung University 1001 Ta-Hsueh Rd. Hsinchu 30010 Taiwan
- Center for Emergent Functional Matter Science, National Yang Ming Chiao Tung University 1001 Ta-Hsueh Rd. Hsinchu 30010 Taiwan
| | - Tung-Kung Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University Hsinchu 30010 Taiwan
- Center for Emergent Functional Matter Science, National Yang Ming Chiao Tung University 1001 Ta-Hsueh Rd. Hsinchu 30010 Taiwan
| |
Collapse
|
27
|
Gharui S, Sengupta D, Das A. Characterization of the Conformational Hotspots of the RNA-Dependent RNA Polymerase Complex Identifies a Unique Structural Malleability of nsp8. J Phys Chem B 2024; 128:9959-9975. [PMID: 39356135 DOI: 10.1021/acs.jpcb.4c03851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Several antiviral therapeutic approaches have been targeted toward the RNA-dependent RNA polymerase (RdRp) complex that is involved in viral genome replication. In SARS-CoV-2, although the RdRp is a multiprotein complex, the focus has been on the ligand binding catalytic core (nonstructural protein nsp12), and not the multiprotein functional dynamics. In this study, we focus on the conformational ensembles of the RdRp complex and their modulation by the presence of RNA, performing comprehensive microsecond-scale atomistic simulations of the apo- and RNA-bound complex. We delineate the differential impact of RNA on the constituent proteins, such as conformational polymorphisms, dominant segment-specific fluctuations, and the switch in dynamical crosstalk within the complex. We distinguish dynamical signatures of nsp7, nsp8, and nsp12 in the apo-state that are reduced in the presence of the RNA and appear to "prime" the complex for activity. Importantly, we identify a unique structural malleability of the nsp8 protein with high conformational heterogeneity in the apo state, especially at three sites (Y71 for nsp8A, and D52 and A66 for nsp8B). Our work highlights the functional implications of the polymorphism of nsp8 structures and reveals possibilities for the development of allosteric inhibitors.
Collapse
Affiliation(s)
- Sowmomita Gharui
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
| | - Durba Sengupta
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Atanu Das
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
28
|
Xue J, Li H, Wang R, Wang M, Chen X, Deng Y, Lu J, Li Y, Song Y, Xu J, Zhu T, Chen L, Liu S. Novel Alkynylamide-Based Nonpeptidic Allosteric Inhibitors for SARS-CoV-2 3-Chymotrypsin-like Protease. ACS Pharmacol Transl Sci 2024; 7:3170-3191. [PMID: 39421662 PMCID: PMC11481521 DOI: 10.1021/acsptsci.4c00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
Although the coronavirus disease 2019 (COVID-19) crisis has passed, there remains a necessity for continuous efforts toward developing more targeted drugs and preparing for potential future virus attacks. Currently, most of the drugs received authorization for the treatment of COVID-19 have exhibited several limitations, such as poor metabolic stability, formidable preparation, and uncertain effectiveness. It is still significant to develop novel, structurally diverse small-molecule antiviral drugs targeting SARS-CoV-2 3-chymotrypsin-like protease (3CLpro). Herein, we report a class of alkynylamide-based nonpeptidic 3CLpro inhibitors that can be prepared conveniently by our previously developed one-pot synthetic method. The structure-activity relationships of alkynylamides as SARS-CoV-2 3CLpro inhibitors have been carefully investigated and discussed in this study. The two stereoisomers of the resulting molecules exhibit stereoselective interaction with 3CLpro, and the optimized compound (S,R)-4y inhibits 3CLpro with high potency (IC50 = 0.43 μM), low cytotoxicity, and acceptable cell permeability. Compound (S,R)-4y presents as a noncovalent inhibitor of 3CLpro against SARS-CoV-2 by the time-dependent inhibition assay (TDI) and mass spectrometry analysis. The Lineweaver-Burk plots, binding energy, surface plasmon resonance, and molecular docking studies suggest that (S,R)-4y specifically binds to an allosteric pocket of the SARS-CoV-2 3CLpro. These findings provide a novel class of nonpeptidic alkynylamide-based allosteric inhibitors with high selectivity against SARS-CoV-2 3CLpro featured by a simplified one-pot synthesis at room temperature in air.
Collapse
Affiliation(s)
- Jian Xue
- Shanghai Engineering Research Center of Molecular
Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering,
East China Normal University, Shanghai 200062,
China
| | - Hongtao Li
- Shanghai Frontiers Science Center of TCM Chemical
Biology, Institute of Interdisciplinary Integrative Medicine Research,
Shanghai University of Traditional Chinese Medicine, Shanghai
200062, China
| | - Ruyu Wang
- Shanghai Frontiers Science Center of TCM Chemical
Biology, Institute of Interdisciplinary Integrative Medicine Research,
Shanghai University of Traditional Chinese Medicine, Shanghai
200062, China
| | - Meiting Wang
- Shanghai Engineering Research Center of Molecular
Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering,
East China Normal University, Shanghai 200062,
China
| | - Xixiang Chen
- Academy of Integrative Medicine, Shanghai Frontiers
Science Center of TCM Chemical Biology, Shanghai University of Traditional
Chinese Medicine, Shanghai 200062, China
| | - Yaqi Deng
- Shanghai Engineering Research Center of Molecular
Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering,
East China Normal University, Shanghai 200062,
China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical
Biology, Institute of Interdisciplinary Integrative Medicine Research,
Shanghai University of Traditional Chinese Medicine, Shanghai
200062, China
| | - Yexi Li
- Shanghai Frontiers Science Center of TCM Chemical
Biology, Institute of Interdisciplinary Integrative Medicine Research,
Shanghai University of Traditional Chinese Medicine, Shanghai
200062, China
| | - Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical
Biology, Institute of Interdisciplinary Integrative Medicine Research,
Shanghai University of Traditional Chinese Medicine, Shanghai
200062, China
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai Frontiers
Science Center of TCM Chemical Biology, Shanghai University of Traditional
Chinese Medicine, Shanghai 200062, China
| | - Tong Zhu
- Shanghai Engineering Research Center of Molecular
Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering,
East China Normal University, Shanghai 200062,
China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical
Biology, Institute of Interdisciplinary Integrative Medicine Research,
Shanghai University of Traditional Chinese Medicine, Shanghai
200062, China
| | - Shunying Liu
- Shanghai Engineering Research Center of Molecular
Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering,
East China Normal University, Shanghai 200062,
China
| |
Collapse
|
29
|
Yang C, Yu Y, Peng Q, Song J, Sun B, Shi Y, Ding Q. Drupacine as a potent SARS-CoV-2 replication inhibitor in vitro. BIOSAFETY AND HEALTH 2024; 6:270-278. [PMID: 40078736 PMCID: PMC11895013 DOI: 10.1016/j.bsheal.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 03/14/2025] Open
Abstract
Despite the availability of vaccines and antiviral treatments, the continued emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and breakthrough infections underscores the need for new, potent antiviral therapies. In a previous study, we established a transcription and replication-competent SARS-CoV-2 virus-like particle (trVLP) system that recapitulates the complete viral life cycle. In this study, we combined high-content screening (HCS) with the SARS-CoV-2 trVLP cell culture system, providing a powerful phenotype-oriented approach to assess the antiviral potential of compounds on a large scale. We screened a library of 3,200 natural compounds and identified drupacine as a potential candidate against SARS-CoV-2 infection. Furthermore, we utilized a SARS-CoV-2 replicon system to demonstrate that drupacine could inhibit viral genome transcription and replication. However, in vitro, enzymatic assays revealed that the inhibition could not be attributed to conventional antiviral targets, such as the viral non-structural proteins nsp5 (MPro) or nsp12 (RdRp). In conclusion, our findings position drupacine as a promising antiviral candidate against SARS-CoV-2, providing a novel scaffold for developing anti-coronavirus disease 2019 therapeutics. Further investigation is required to pinpoint its precise target and mechanism of action.
Collapse
Affiliation(s)
- Chen Yang
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yanying Yu
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Qi Peng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingwei Song
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Bo Sun
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Ding
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
30
|
Yu TWB. A phenotypic drug discovery approach by latent interaction in deep learning. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240720. [PMID: 40191531 PMCID: PMC11972434 DOI: 10.1098/rsos.240720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 04/09/2025]
Abstract
Contemporary drug discovery paradigms rely heavily on binding assays about the bio-physicochemical processes. However, this dominant approach suffers from overlooked higher-order interactions arising from the intricacies of molecular mechanisms, such as those involving cis-regulatory elements. It introduces potential impairments and restrains the potential development of computational methods. To address this limitation, I developed a deep learning model that leverages an end-to-end approach, relying exclusively on therapeutic information about drugs. By transforming textual representations of drug and virus genetic information into high-dimensional latent representations, this method evades the challenges arising from insufficient information about binding specificities. Its strengths lie in its ability to implicitly consider complexities such as epistasis and chemical-genetic interactions, and to handle the pervasive challenge of data scarcity. Through various modeling skills and data augmentation techniques, the proposed model demonstrates outstanding performance in out-of-sample validations, even in scenarios with unknown complex interactions. Furthermore, the study highlights the importance of chemical diversity for model training. While the method showcases the feasibility of deep learning in data-scarce scenarios, it reveals a promising alternative for drug discovery in situations where knowledge of underlying mechanisms is limited.
Collapse
Affiliation(s)
- Tat Wai Billy Yu
- Macao Polytechnic University, Macau SAR, People’s Republic of China
| |
Collapse
|
31
|
Li DY, Donadu MG, Shue T, Dangas G, Athanasiadis A, Lan S, Wen X, Battah B, Zanetti S, Mazzarello V, Sarafianos SG, Ferrari M, Michailidis E. Myrtus communis L. Essential Oil Exhibits Antiviral Activity against Coronaviruses. Pharmaceuticals (Basel) 2024; 17:1189. [PMID: 39338351 PMCID: PMC11435418 DOI: 10.3390/ph17091189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Human coronaviruses are a continuous threat to the human population and have limited antiviral treatments, and the recent COVID-19 pandemic sparked interest in finding new antiviral strategies, such as natural products, to combat emerging coronaviruses. Rapid efforts in the scientific community to identify effective antiviral agents for coronaviruses remain a focus to minimize mortalities and global setbacks. In this study, an essential oil derived from Myrtus communis L. (MEO) is effective against HCoV-229E and HCoV-OC43 virus infections in comparison to two FDA-approved drugs, Remdesivir and Nirmatrelvir. Gas-chromatography and mass spectrometry were used to identify the chemical composition of MEO. Slight antioxidant activity was observed in MEO, indicating a role in oxidative stress. A dose-response curve measuring the EC50 indicates a high potency against HCoV-229E and HCoV-OC43 virus infections on Huh7.5 cells with low cytotoxicity using a PrestoBlue cell viability assay. Our findings demonstrate that MEO exhibits potent antiviral activity against HCoV-229E and HCoV-OC43 on Huh7.5 cells within a low-cytotoxicity range, but not on SARS-CoV-2. Artificial bacterial chromosome plasmids that expressed SARS-CoV-2 used for replicon-to determine viral replication and viral assembly/egress on HEK293T/17 cells-and virus-like particles on Huh7.5-AT cells-to determine viral entry and assembly/egress-showed no antiviral activity with MEO in comparison to Remdesivir. This study reveals the potential effectiveness of MEO as an alternative natural remedy to treat human coronaviruses and a potential antiviral agent for future coronavirus infections.
Collapse
Affiliation(s)
- Dar-Yin Li
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Matthew G Donadu
- Scuola di Specializzazione in Farmacia Ospedaliera, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
- Hospital Pharmacy, Giovanni Paolo II Hospital, ASL Gallura, 07026 Olbia, Italy
| | - Taylor Shue
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Georgios Dangas
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Antonis Athanasiadis
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Shuiyun Lan
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Xin Wen
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Basem Battah
- Department of Biochemistry and Microbiology, Antioch Syrian Private University, M5, Damascus 22734, Syria
| | - Stefania Zanetti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Vittorio Mazzarello
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| | - Marco Ferrari
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Via dell'Istria, 65, 34137 Trieste, Italy
| | - Eleftherios Michailidis
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, and Children's Healthcare of Atlanta, 1750 Haygood Drive, Atlanta, GA 30322, USA
| |
Collapse
|
32
|
Huang Y, Wang T, Zhong L, Zhang W, Zhang Y, Yu X, Yuan S, Ni T. Molecular architecture of coronavirus double-membrane vesicle pore complex. Nature 2024; 633:224-231. [PMID: 39143215 PMCID: PMC11374677 DOI: 10.1038/s41586-024-07817-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Coronaviruses remodel the intracellular host membranes during replication, forming double-membrane vesicles (DMVs) to accommodate viral RNA synthesis and modifications1,2. SARS-CoV-2 non-structural protein 3 (nsp3) and nsp4 are the minimal viral components required to induce DMV formation and to form a double-membrane-spanning pore, essential for the transport of newly synthesized viral RNAs3-5. The mechanism of DMV pore complex formation remains unknown. Here we describe the molecular architecture of the SARS-CoV-2 nsp3-nsp4 pore complex, as resolved by cryogenic electron tomography and subtomogram averaging in isolated DMVs. The structures uncover an unexpected stoichiometry and topology of the nsp3-nsp4 pore complex comprising 12 copies each of nsp3 and nsp4, organized in 4 concentric stacking hexamer rings, mimicking a miniature nuclear pore complex. The transmembrane domains are interdigitated to create a high local curvature at the double-membrane junction, coupling double-membrane reorganization with pore formation. The ectodomains form extensive contacts in a pseudo-12-fold symmetry, belting the pore complex from the intermembrane space. A central positively charged ring of arginine residues coordinates the putative RNA translocation, essential for virus replication. Our work establishes a framework for understanding DMV pore formation and RNA translocation, providing a structural basis for the development of new antiviral strategies to combat coronavirus infection.
Collapse
Affiliation(s)
- Yixin Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tongyun Wang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lijie Zhong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wenxin Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiulian Yu
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Tao Ni
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, China.
| |
Collapse
|
33
|
Nisar H, Wajid B, Anwar F, Ahmad A, Javaid A, Attique SA, Nisar W, Saeed A, Shahid S, Sadaf S. Bioinformatics and systems biology analysis revealed PMID26394986-Compound-10 as potential repurposable drug against covid-19. J Biomol Struct Dyn 2024; 42:7972-7985. [PMID: 37534820 DOI: 10.1080/07391102.2023.2242500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
The global health pandemic known as COVID-19, which stems from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a significant concern worldwide. Several treatment methods exist for COVID-19; however, there is an urgent demand for previously established drugs and vaccines to effectively combat the disease. Since, discovering new drugs poses a significant challenge, making the repurposing of existing drugs can potentially reduce time and costs compared to developing entirely new drugs from scratch. The objective of this study is to identify hub genes and associated repurposed drugs targeting them. We analyzed differentially expressed genes (DEGs) by analyzing RNA-seq transcriptomic datasets and integrated with genes associated with COVID-19 present in different databases. We detected 173 Covid-19 associated genes for the construction of a protein-protein interaction (PPI) network which resulted in the identification of the top 10 hub genes/proteins (STAT1, IRF7, MX1, IRF9, ISG15, OAS3, OAS2, OAS1, IRF3, and IRF1). Hub genes were subjected to GO functional and KEGG pathway enrichment analyses, which indicated some key roles and signaling pathways that were strongly related to SARS-CoV-2 infections. We conducted drug repurposing analysis using CMap, TTD, and DrugBank databases with these 10 hub genes, leading to the identification of Piceatannol, CKD-712, and PMID26394986-Compound-10 as top-ranked candidate drugs. Finally, drug-gene interactions analysis through molecular docking and validated via molecular dynamic simulation for 80 ns suggests PMID26394986-Compound-10 as the only potential drug. Our research demonstrates how in silico analysis might produce repurposing candidates to help respond faster to new disease outbreaks.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Haseeb Nisar
- Department of Life-Sciences, University of Management and Technology, Lahore, Pakistan
| | - Bilal Wajid
- Ibn Sina Research & Development Division, Sabz-Qalam, Lahore, Pakistan
- Department of Electrical Engineering, University of Engineering and Technology, Lahore, Pakistan
| | | | - Ashfaq Ahmad
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Anum Javaid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Syed Awais Attique
- Bioinformatics Institute, Agency for Science, Technology and Research (A(*)STAR), Singapore, Singapore
| | - Wardah Nisar
- Department of Public Health, University of Health Sciences, Lahore, Pakistan
| | - Amir Saeed
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Samiah Shahid
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
34
|
Li J, Xiao H, Zhang C, Liu G, Liu X. From virus to immune system: Harnessing membrane-derived vesicles to fight COVID-19 by interacting with biological molecules. Eur J Immunol 2024; 54:e2350916. [PMID: 38778737 DOI: 10.1002/eji.202350916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
Emerging and re-emerging viral pandemics have emerged as a major public health concern. Highly pathogenic coronaviruses, which cause severe respiratory disease, threaten human health and socioeconomic development. Great efforts are being devoted to the development of safe and efficacious therapeutic agents and preventive vaccines to combat them. Nevertheless, the highly mutated virus poses a challenge to drug development and vaccine efficacy, and the use of common immunomodulatory agents lacks specificity. Benefiting from the burgeoning intersection of biological engineering and biotechnology, membrane-derived vesicles have shown superior potential as therapeutics due to their biocompatibility, design flexibility, remarkable bionics, and inherent interaction with phagocytes. The interactions between membrane-derived vesicles, viruses, and the immune system have emerged as a new and promising topic. This review provides insight into considerations for developing innovative antiviral strategies and vaccines against SARS-CoV-2. First, membrane-derived vesicles may provide potential biomimetic decoys with a high affinity for viruses to block virus-receptor interactions for early interruption of infection. Second, membrane-derived vesicles could help achieve a balanced interplay between the virus and the host's innate immunity. Finally, membrane-derived vesicles have revealed numerous possibilities for their employment as vaccines.
Collapse
Affiliation(s)
- Jiayuan Li
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Haiqing Xiao
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Chang Zhang
- Clinical Center for Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Gang Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xuan Liu
- Clinical Center for Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Shen Zhen Research Institute of Xiamen University, Xiamen University, Shenzhen, China
| |
Collapse
|
35
|
Bolinger AA, Li J, Xie X, Li H, Zhou J. Lessons learnt from broad-spectrum coronavirus antiviral drug discovery. Expert Opin Drug Discov 2024; 19:1023-1041. [PMID: 39078037 PMCID: PMC11390334 DOI: 10.1080/17460441.2024.2385598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Highly pathogenic coronaviruses (CoVs), such as severe acute respiratory syndrome CoV (SARS-CoV), Middle East respiratory syndrome CoV (MERS-CoV), and the most recent SARS-CoV-2 responsible for the COVID-19 pandemic, pose significant threats to human populations over the past two decades. These CoVs have caused a broad spectrum of clinical manifestations ranging from asymptomatic to severe distress syndromes (ARDS), resulting in high morbidity and mortality. AREAS COVERED The accelerated advancements in antiviral drug discovery, spurred by the COVID-19 pandemic, have shed new light on the imperative to develop treatments effective against a broad spectrum of CoVs. This perspective discusses strategies and lessons learnt in targeting viral non-structural proteins, structural proteins, drug repurposing, and combinational approaches for the development of antivirals against CoVs. EXPERT OPINION Drawing lessons from the pandemic, it becomes evident that the absence of efficient broad-spectrum antiviral drugs increases the vulnerability of public health systems to the potential onslaught by highly pathogenic CoVs. The rapid and sustained spread of novel CoVs can have devastating consequences without effective and specifically targeted treatments. Prioritizing the effective development of broad-spectrum antivirals is imperative for bolstering the resilience of public health systems and mitigating the potential impact of future highly pathogenic CoVs.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jun Li
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
36
|
Li M, Yang Y, Wang P, Que W, Zhong L, Cai Z, Liu Y, Yang L, Liu Y. Transcriptome dynamics of the BHK21 cell line in response to human coronavirus OC43 infection. Microbiol Res 2024; 285:127750. [PMID: 38761489 DOI: 10.1016/j.micres.2024.127750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The progress of viral infection involves numerous transcriptional regulatory events. The identification of the newly synthesized transcripts helps us to understand the replication mechanisms and pathogenesis of the virus. Here, we utilized a time-resolved technique called metabolic RNA labeling approach called thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM-seq) to differentially elucidate the levels of steady-state and newly synthesized RNAs of BHK21 cell line in response to human coronavirus OC43 (HCoV-OC43) infection. Our results showed that the Wnt/β-catenin signaling pathway was significantly enriched with the newly synthesized transcripts of BHK21 cell line in response to HCoV-OC43 infection. Moreover, inhibition of the Wnt pathway promoted viral replication in the early stage of infection, but inhibited it in the later stage of infection. Furthermore, remdesivir inhibits the upregulation of the Wnt/β-catenin signaling pathway induced by early infection with HCoV-OC43. Collectively, our study showed the diverse roles of Wnt/β-catenin pathway at different stages of HCoV-OC43 infection, suggesting a potential target for the antiviral treatment. In addition, although infection with HCoV-OC43 induces cytopathic effects in BHK21 cells, inhibiting apoptosis does not affect the intracellular replication of the virus. Monitoring newly synthesized RNA based on such time-resolved approach is a highly promising method for studying the mechanism of viral infections.
Collapse
Affiliation(s)
- Mianhuan Li
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, People's Republic of China; Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Yang Yang
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Pusen Wang
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Weitao Que
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Lin Zhong
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Zhao Cai
- Shenzhen Mindray Bio-Medical Electronics Co.,Ltd, Shenzhen 518057, People's Republic of China
| | - Yang Liu
- Southern University of Science and Technology Hospital, Shenzhen 518055, People's Republic of China
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, People's Republic of China; Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China.
| | - Yingxia Liu
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China.
| |
Collapse
|
37
|
Fomina AD, Uvarova VI, Kozlovskaya LI, Palyulin VA, Osolodkin DI, Ishmukhametov AA. Ensemble docking based virtual screening of SARS-CoV-2 main protease inhibitors. Mol Inform 2024; 43:e202300279. [PMID: 38973780 DOI: 10.1002/minf.202300279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/21/2024] [Accepted: 03/03/2024] [Indexed: 07/09/2024]
Abstract
During the first years of COVID-19 pandemic, X-ray structures of the coronavirus drug targets were acquired at an unprecedented rate, giving hundreds of PDB depositions in less than a year. The main protease (Mpro) of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) is the primary validated target of direct-acting antivirals. The selection of the optimal ensemble of structures of Mpro for the docking-driven virtual screening campaign was thus non-trivial and required a systematic and automated approach. Here we report a semi-automated active site RMSD based procedure of ensemble selection from the SARS-CoV-2 Mpro crystallographic data and virtual screening of its inhibitors. The procedure was compared with other approaches to ensemble selection and validated with the help of hand-picked and peer-reviewed activity-annotated libraries. Prospective virtual screening of non-covalent Mpro inhibitors resulted in a new chemotype of thienopyrimidinone derivatives with experimentally confirmed enzyme inhibition.
Collapse
Affiliation(s)
- Anastasia D Fomina
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), 108819, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Victoria I Uvarova
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), 108819, Moscow, Russia
| | - Liubov I Kozlovskaya
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), 108819, Moscow, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Vladimir A Palyulin
- Department of Chemistry, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Dmitry I Osolodkin
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), 108819, Moscow, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Aydar A Ishmukhametov
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), 108819, Moscow, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| |
Collapse
|
38
|
Yoosefian M, Sabaghian H. Silver nanoparticle-based drug delivery systems in the fight against COVID-19: enhancing efficacy, reducing toxicity and improving drug bioavailability. J Drug Target 2024; 32:794-806. [PMID: 38742854 DOI: 10.1080/1061186x.2024.2356147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/26/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Nanoparticles (NPs) have played a pivotal role in various biomedical applications, spanning from sensing to drug delivery, imaging and anti-viral therapy. The therapeutic utilisation of NPs in clinical trials was established in the early 1990s. Silver nanoparticles (AgNPs) possess anti-microbial, anti-cancer and anti-viral properties, which make them a possible anti-viral drug to combat the COVID-19 virus. Free radicals and reactive oxygen species are produced by AgNPs, which causes apoptosis induction and prevents viral contamination. The shape and size of AgNPs can influence their interactions and biological activities. Therefore, it is recommended that silver nanoparticles (AgNPs) be used as a valuable tool in the management of COVID-19 pandemic. These nanoparticles possess strong anti-microbial properties, allowing them to penetrate and destroy microbial cells. Additionally, the toxicity level of nanoparticles depends on the administered dose, and surface modifications are necessary to reduce toxicity, preventing direct interaction between metal surfaces and cells. By utilising silver nanoparticles, drugs can be targeted to specific areas in the body. For example, in the case of COVID-19, anti-viral drugs can be stimulated as nanoparticles in the lungs to accelerate disease recovery. Nanoparticle-based systems have the capability to transport drugs and treat specific body parts. This review offers an examination of silver nanoparticle-based drug delivery systems for combatting COVID-19, with the objective of boosting the bioavailability of existing medications, decreasing their toxicity and raising their efficiency.
Collapse
Affiliation(s)
- Mehdi Yoosefian
- Department of Chemistry, Graduate University of Advanced Technology, Kerman, Iran
| | - Hanieh Sabaghian
- Department of Chemistry, Graduate University of Advanced Technology, Kerman, Iran
| |
Collapse
|
39
|
Dampalla CS, Kim Y, Zabiegala A, Howard DJ, Nguyen HN, Madden TK, Thurman HA, Cooper A, Liu L, Battaile KP, Lovell S, Chang KO, Groutas WC. Structure-Guided Design of Potent Coronavirus Inhibitors with a 2-Pyrrolidone Scaffold: Biochemical, Crystallographic, and Virological Studies. J Med Chem 2024; 67:11937-11956. [PMID: 38953866 DOI: 10.1021/acs.jmedchem.4c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Zoonotic coronaviruses are known to produce severe infections in humans and have been the cause of significant morbidity and mortality worldwide. SARS-CoV-2 was the largest and latest contributor of fatal cases, even though MERS-CoV has the highest case-fatality ratio among zoonotic coronaviruses. These infections pose a high risk to public health worldwide warranting efforts for the expeditious discovery of antivirals. Hence, we hereby describe a novel series of inhibitors of coronavirus 3CLpro embodying an N-substituted 2-pyrrolidone scaffold envisaged to exploit favorable interactions with the S3-S4 subsites and connected to an invariant Leu-Gln P2-P1 recognition element. Several inhibitors showed nanomolar antiviral activity in enzyme and cell-based assays, with no significant cytotoxicity. High-resolution crystal structures of inhibitors bound to the 3CLpro were determined to probe and identify the molecular determinants associated with binding, to inform the structure-guided optimization of the inhibitors, and to confirm the mechanism of action of the inhibitors.
Collapse
Affiliation(s)
- Chamandi S Dampalla
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, Kansas 67260, United States
| | - Yunjeong Kim
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Alexandria Zabiegala
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Dennis J Howard
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, Kansas 67260, United States
| | - Harry Nhat Nguyen
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, Kansas 67260, United States
| | - Trent K Madden
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, Kansas 67260, United States
| | - Hayden A Thurman
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, Kansas 67260, United States
| | - Anne Cooper
- Protein Structure and X-ray Crystallography Laboratory, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Lijun Liu
- Protein Structure and X-ray Crystallography Laboratory, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Kevin P Battaile
- NYX, New York Structural Biology Center, Upton, New York 11973, United States
| | - Scott Lovell
- Protein Structure and X-ray Crystallography Laboratory, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - William C Groutas
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, Kansas 67260, United States
| |
Collapse
|
40
|
Song N, Zheng W, Song B, Zheng J. Allosteric Regulation and Inhibition of Coronavirus 3CLpro Revealed by HDX-MS. Chembiochem 2024; 25:e202400001. [PMID: 38720172 DOI: 10.1002/cbic.202400001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/03/2024] [Indexed: 07/03/2024]
Abstract
Coronavirus (CoV) infections have caused contagious and fatal respiratory diseases in humans worldwide. CoV 3-chymotrypsin-like proteases (3CLpro or Mpro) play an important role in viral maturation, and maintenance of their dimeric conformation is crucial for viral activity. Therefore, allosterically regulated dimerization of 3CLpro can be employed as a drug development target. Here, we investigated the allosteric regulatory mechanism of 3CLpro dimerization by using hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) technology. We found that the FLAG tag directly coupled to the N-finger of 3CLpro significantly increased HDX kinetics at the dimer interface, and 3CLpro transformed from a dimer to a monomer. The 3CLpro mutants of SARS-CoV-2, which are monomeric, also exhibited increased deuterium exchange. Binding of the allosteric inhibitor Gastrodenol to most betacoronavirus 3CLpros led to increased allosteric deuterium exchange, resulting in the monomeric conformation of the CoV 3CLpro upon binding. Molecular dynamics (MD) simulation analysis further indicated the molecular mechanism of action of Gastrodenol on CoV 3CLpro: binding of Gastrodenol to SARS-CoV-2 3CLpro destroyed the hydrogen bond in the dimer interface. These results suggest that Gastrodenol may be a potential broad-spectrum anti-betacoronavirus drug.
Collapse
Affiliation(s)
- Ning Song
- Immunological Disease Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, 201203, Shanghai, PR China
- University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408, Beijing, PR China
| | - Wen Zheng
- CADD computer-aided protein Design Simulator, Hubei Yuanda Biotechnology Co., Ltd., Building B6, Optics Valley Biological City, Hongshan District, 430000, Wuhan, Hubei, PR China
| | - Bin Song
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, 200025, Shanghai, PR China
| | - Jie Zheng
- Immunological Disease Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, 201203, Shanghai, PR China
- University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408, Beijing, PR China
| |
Collapse
|
41
|
Siragam V, Maltseva M, Castonguay N, Galipeau Y, Srinivasan MM, Soto JH, Dankar S, Langlois MA. Seasonal human coronaviruses OC43, 229E, and NL63 induce cell surface modulation of entry receptors and display host cell-specific viral replication kinetics. Microbiol Spectr 2024; 12:e0422023. [PMID: 38864599 PMCID: PMC11218498 DOI: 10.1128/spectrum.04220-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
The emergence of the COVID-19 pandemic prompted an increased interest in seasonal human coronaviruses. OC43, 229E, NL63, and HKU1 are endemic seasonal coronaviruses that cause the common cold and are associated with generally mild respiratory symptoms. In this study, we identified cell lines that exhibited cytopathic effects (CPE) upon infection by three of these coronaviruses and characterized their viral replication kinetics and the effect of infection on host surface receptor expression. We found that NL63 produced CPE in LLC-MK2 cells, while OC43 produced CPE in MRC-5, HCT-8, and WI-38 cell lines, while 229E produced CPE in MRC-5 and WI-38 by day 3 post-infection. We observed a sharp increase in nucleocapsid and spike viral RNA (vRNA) from day 3 to day 5 post-infection for all viruses; however, the abundance and the proportion of vRNA copies measured in the supernatants and cell lysates of infected cells varied considerably depending on the virus-host cell pair. Importantly, we observed modulation of coronavirus entry and attachment receptors upon infection. Infection with 229E and OC43 led to a downregulation of CD13 and GD3, respectively. In contrast, infection with NL63 and OC43 leads to an increase in ACE2 expression. Attempts to block entry of NL63 using either soluble ACE2 or anti-ACE2 monoclonal antibodies demonstrated the potential of these strategies to greatly reduce infection. Overall, our results enable a better understanding of seasonal coronaviruses infection kinetics in permissive cell lines and reveal entry receptor modulation that may have implications in facilitating co-infections with multiple coronaviruses in humans.IMPORTANCESeasonal human coronavirus is an important cause of the common cold associated with generally mild upper respiratory tract infections that can result in respiratory complications for some individuals. There are no vaccines available for these viruses, with only limited antiviral therapeutic options to treat the most severe cases. A better understanding of how these viruses interact with host cells is essential to identify new strategies to prevent infection-related complications. By analyzing viral replication kinetics in different permissive cell lines, we find that cell-dependent host factors influence how viral genes are expressed and virus particles released. We also analyzed entry receptor expression on infected cells and found that these can be up- or down-modulated depending on the infecting coronavirus. Our findings raise concerns over the possibility of infection enhancement upon co-infection by some coronaviruses, which may facilitate genetic recombination and the emergence of new variants and strains.
Collapse
MESH Headings
- Humans
- Virus Replication
- Coronavirus NL63, Human/physiology
- Coronavirus NL63, Human/genetics
- Coronavirus 229E, Human/physiology
- Coronavirus 229E, Human/genetics
- Coronavirus OC43, Human/physiology
- Coronavirus OC43, Human/genetics
- Cell Line
- Virus Internalization
- Seasons
- Kinetics
- Receptors, Virus/metabolism
- Receptors, Virus/genetics
- Common Cold/virology
- Common Cold/metabolism
- SARS-CoV-2/physiology
- SARS-CoV-2/genetics
- SARS-CoV-2/metabolism
- RNA, Viral/metabolism
- RNA, Viral/genetics
- Animals
- COVID-19/virology
- COVID-19/metabolism
- Coronavirus/physiology
- Coronavirus/genetics
Collapse
Affiliation(s)
- Vinayakumar Siragam
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Mariam Maltseva
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Nicolas Castonguay
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Mrudhula Madapuji Srinivasan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Justino Hernandez Soto
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Samar Dankar
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- The Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, Canada
| |
Collapse
|
42
|
Chan JFW, Yuan S, Chu H, Sridhar S, Yuen KY. COVID-19 drug discovery and treatment options. Nat Rev Microbiol 2024; 22:391-407. [PMID: 38622352 DOI: 10.1038/s41579-024-01036-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 04/17/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused substantial morbidity and mortality, and serious social and economic disruptions worldwide. Unvaccinated or incompletely vaccinated older individuals with underlying diseases are especially prone to severe disease. In patients with non-fatal disease, long COVID affecting multiple body systems may persist for months. Unlike SARS-CoV and Middle East respiratory syndrome coronavirus, which have either been mitigated or remained geographically restricted, SARS-CoV-2 has disseminated globally and is likely to continue circulating in humans with possible emergence of new variants that may render vaccines less effective. Thus, safe, effective and readily available COVID-19 therapeutics are urgently needed. In this Review, we summarize the major drug discovery approaches, preclinical antiviral evaluation models, representative virus-targeting and host-targeting therapeutic options, and key therapeutics currently in clinical use for COVID-19. Preparedness against future coronavirus pandemics relies not only on effective vaccines but also on broad-spectrum antivirals targeting conserved viral components or universal host targets, and new therapeutics that can precisely modulate the immune response during infection.
Collapse
Affiliation(s)
- Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, China.
| |
Collapse
|
43
|
Li S, Li H, Lian R, Xie J, Feng R. New perspective of small-molecule antiviral drugs development for RNA viruses. Virology 2024; 594:110042. [PMID: 38492519 DOI: 10.1016/j.virol.2024.110042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
High variability and adaptability of RNA viruses allows them to spread between humans and animals, causing large-scale infectious diseases which seriously threat human and animal health and social development. At present, AIDS, viral hepatitis and other viral diseases with high incidence and low cure rate are still spreading around the world. The outbreaks of Ebola, Zika, dengue and in particular of the global pandemic of COVID-19 have presented serious challenges to the global public health system. The development of highly effective and broad-spectrum antiviral drugs is a substantial and urgent research subject to deal with the current RNA virus infection and the possible new viral infections in the future. In recent years, with the rapid development of modern disciplines such as artificial intelligence technology, bioinformatics, molecular biology, and structural biology, some new strategies and targets for antivirals development have emerged. Here we review the main strategies and new targets for developing small-molecule antiviral drugs against RNA viruses through the analysis of the new drug development progress against several highly pathogenic RNA viruses, to provide clues for development of future antivirals.
Collapse
Affiliation(s)
- Shasha Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Huixia Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Ruiya Lian
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Jingying Xie
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China.
| |
Collapse
|
44
|
Chen JY, Huang TR, Hsu SY, Huang CC, Wang HS, Chang JS. Effect and mechanism of quercetin or quercetin-containing formulas against COVID-19: From bench to bedside. Phytother Res 2024; 38:2597-2618. [PMID: 38479376 DOI: 10.1002/ptr.8175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 06/13/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global coronavirus disease 2019 (COVID-19) pandemic since 2019. Immunopathogenesis and thromboembolic events are central to its pathogenesis. Quercetin exhibits several beneficial activities against COVID-19, including antiviral, anti-inflammatory, immunomodulatory, antioxidative, and antithrombotic effects. Although several reviews have been published, these reviews are incomplete from the viewpoint of translational medicine. The authors comprehensively evaluated the evidence of quercetin against COVID-19, both basically and clinically, to apply quercetin and/or its derivatives in the future. The authors searched the PubMed, Embase, and the Cochrane Library databases without any restrictions. The search terms included COVID-19, SARS-CoV-2, quercetin, antiviral, anti-inflammatory, immunomodulatory, thrombosis, embolism, oxidative, and microbiota. The references of relevant articles were also reviewed. All authors independently screened and reviewed the quality of each included manuscript. The Cochrane Risk of Bias Tool, version 2 (RoB 2) was used to assess the quality of the included randomized controlled trials (RCTs). All selected studies were discussed monthly. The effectiveness of quercetin against COVID-19 is not solid due to methodological flaws in the clinical trials. High-quality studies are also required for quercetin-containing traditional Chinese medicines. The low bioavailability and highly variable pharmacokinetics of quercetin hinder its clinical applications. Its positive impact on immunomodulation through reverting dysbiosis of gut microbiota still lacks robust evidence. Quercetin against COVID-19 does not have tough clinical evidence. Strategies to improve its bioavailability and/or to develop its effective derivatives are needed. Well-designed RCTs are also crucial to confirm their effectiveness in the future.
Collapse
Affiliation(s)
- Jhong Yuan Chen
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung Rung Huang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih Yun Hsu
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching Chun Huang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Huei Syun Wang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jung San Chang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
45
|
Feng J, Huang X, Xu Q, Tang R, Zhou Y, Qin S, Xing S, Gao Y, Mei S, He Z. Pharmacological inhibition of the ACE/Ang-2/AT1 axis alleviates mechanical ventilation-induced pulmonary fibrosis. Int Immunopharmacol 2024; 131:111855. [PMID: 38493697 DOI: 10.1016/j.intimp.2024.111855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/22/2024] [Accepted: 03/10/2024] [Indexed: 03/19/2024]
Abstract
Mechanical ventilation (MV) is an essential therapy for acute respiratory distress syndrome (ARDS) and pulmonary fibrosis. However, it can also induce mechanical ventilation-induced pulmonary fibrosis (MVPF) and the underlying mechanism remains unknown. Based on a mouse model of MVPF, the present study aimed to explore the role of the angiotensin-converting enzyme/angiotensin II/angiotensin type 1 receptor (ACE/Ang-2/AT1R) axis in the process of MVPF. In addition, recombinant angiotensin-converting enzyme 2(rACE2), AT1R inhibitor valsartan, AGTR1-directed shRNA and ACE inhibitor perindopril were applied to verify the effect of inhibiting ACE/Ang-2/AT1R axis in the treatment of MVPF. Our study found MV induced an inflammatory reaction and collagen deposition in mouse lung tissue accompanied by the activation of ACE in lung tissue, increased concentration of Ang-2 in bronchoalveolar lavage fluid (BALF), and upregulation of AT1R in alveolar epithelial cells. The process of pulmonary fibrosis could be alleviated by the application of the ACE inhibitor perindopril, ATIR inhibitor valsartan and AGTR1-directed shRNA. Meanwhile, rACE2 could also alleviate MVPF through the degradation of Ang-2. Our finding indicated the ACE/Ang-2/AT1R axis played an essential role in the pathogenesis of MVPF. Pharmacological inhibition of the ACE/Ang-2/AT1R axis might be a promising strategy for the treatment of MVPF.
Collapse
Affiliation(s)
- Jinhua Feng
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xi Huang
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qiaoyi Xu
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ri Tang
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yang Zhou
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shaojie Qin
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shunpeng Xing
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yuan Gao
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shuya Mei
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Zhengyu He
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
46
|
Branda F, Ciccozzi M. Navigating Novel Viral Challenges: Understanding, Tracking, and Mitigating Emerging Threats. Microorganisms 2024; 12:807. [PMID: 38674751 PMCID: PMC11051937 DOI: 10.3390/microorganisms12040807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of new viral threats continues to pose significant challenges to global health security [...].
Collapse
|
47
|
Yan Y, Liu H, Wu D, Gu Z, Guo W, Yao H, Lin K, Li X. Design, synthesis and biological evaluation of novel 3C-like protease inhibitors as lead compounds against SARS-CoV-2. Future Med Chem 2024; 16:887-903. [PMID: 38618977 PMCID: PMC11249163 DOI: 10.4155/fmc-2024-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Background: The epidemic caused by SARS-CoV-2 swept the world in 2019. The 3C-like protease (3CLpro) of SARS-CoV-2 plays a key role in viral replication, and its inhibition could inhibit viral replication. Materials & methods: The virtual screen based on receptor-ligand pharmacophore models and molecular docking were conducted to obtain the novel scaffolds of the 3CLpro. The molecular dynamics simulation was also carried out. All compounds were synthesized and evaluated in biochemical assays. Results: The compound C2 could inhibit 3CLpro with a 72% inhibitory rate at 10 μM. The covalent docking showed that C2 could form a covalent bond with the Cys145 in 3CLpro. Conclusion: C2 could be a potent lead compound of 3CLpro inhibitors against SARS-CoV-2.
Collapse
Affiliation(s)
- Yong Yan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanwen Liu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Di Wu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhihao Gu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Wenhao Guo
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hequan Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kejiang Lin
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xuanyi Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
48
|
Ramachandran B, Nadeem A, Mohanprasanth A, Saravanan M. Prediction of deleterious non-synonymous SNPs of TMPRSS2 protein combined with Molecular Dynamics Simulations and free energy analysis to identify the potential peptide substrates against SARS-CoV-2. J Biomol Struct Dyn 2024:1-15. [PMID: 38592189 DOI: 10.1080/07391102.2024.2330710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Globally the SARS-CoV-2 viral infection demands for the new drugs, the TMPRSS2 target plays a vital role in facilitating the virus entry. The aim of the present study is to identify the potential peptide substrate from the Anti-viral database against TMPRSS2 of SARS-CoV-2. The compound screening and variation analysis were performed using molecular docking analysis and online tools such as PROVEAN and SNAP2 server, respectively. The re-docked crystal structure peptide substrate exhibits -128.151 kcal/mol whereas the RRKK peptide substrate shows -134.158 kcal/mol. Further, the selected compounds were proceeded with Molecular Dynamics Simulation, it explores the stability of the complex by revealing the hotspot residues (His296 and Ser441) were active for nucleophilic attack against TMPRSS2. The average Binding Free Energy values computed through MM/GBSA for RRKK, Camostat, and Crystal Structure were shown -69.9278 kcal/mol, -64.5983 kcal/mol, and -63.9755 kcal/mol, respectively against TMPRSS2. The 'rate of acylation' emerges as an indicator for RRKK's efficacy, it maintains the distance of 3.2 Å with Ser441 resembles, whilst its -NH backbone stabilizes at 2.5 Å 'Michaelis Complex' which leads to prevent the entry of SARS-CoV-2 to human cells. The sequence variation analysis explores that the V160 and G6 substitutions are essential to emphasize the uncover possibilities for the ongoing drug discovery research. Therefore, the identified peptide substrate found to be potent against SARS-CoV-2 and these results will be valuable for ongoing drug discovery research.
Collapse
Affiliation(s)
- Balajee Ramachandran
- Structural and Computational Biology Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, India
- Department of Pharmacology, Physiology and Biophysics, Boston University School of Medicine, Boston, MA, USA
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aruchamy Mohanprasanth
- AMR & Nanotherapeutics Lab, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical, Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Muthupandian Saravanan
- AMR & Nanotherapeutics Lab, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical, Sciences (SIMATS), Chennai, Tamil Nadu, India
| |
Collapse
|
49
|
Ozeer FZ, Nagandran S, Wu YS, Wong LS, Stephen A, Lee MF, Kijsomporn J, Guad RM, Batumalaie K, Oyewusi HA, Verma A, Yadav E, Afzal S, Sekar M, Subramaniyan V, Fuloria NK, Fuloria S, Sarker MMR. A comprehensive review of phytochemicals of Withania somnifera (L.) Dunal (Solanaceae) as antiviral therapeutics. DISCOVER APPLIED SCIENCES 2024; 6:187. [DOI: 10.1007/s42452-024-05845-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/20/2024] [Indexed: 11/22/2024]
Abstract
AbstractViruses have caused millions and billions of infections and high mortality rates without successful immunization due to a lack of antiviral drugs approved for clinical use. Therefore, the discovery of novel antiviral drugs is impertinent and natural products are excellent alternative sources. Withania somnifera (L.) Dunal (Solanaceae) is recognized as one of the most significant herbs in the Ayurvedic system and it had been utilized in various biological actions for more than 3000 years. This review aimed to discuss the therapeutic effects and associated molecular mechanisms of Withania somnifera (WS) and its phytochemicals, withanolides against various viruses in preclinical and clinical settings towards developing potential inhibitors which could target virus proteins or their respective host cell receptors. WS was reported to attenuate coronavirus disease 2019 (COVID-19), serve as a potential ligand against the herpes simplex virus (HSV) DNA polymerase, suppress Alzheimer’s disease progression by inhibiting the cytotoxicity induced by the human immunodeficiency virus 1 (HIV-1)-activated beta-amyloid (Aβ), and attenuate the neuraminidase activity of H1N1 influenza. WS root extracts have also reduced the mortality rates and stress levels in tilapia infected with tilapia lake virus (TiLV), and stimulated antiviral nitric oxide formation in chicks infected with infectious bursal disease (IBD). With increasing evidence from previous literatures, further in vitro and in vivo investigations of WS against other viral infections may provide promising results.
Graphical Abstract
Collapse
|
50
|
Okasha NI, Abdel-Rahman MA, Nafie MS, Abo Shama NM, Mahmoud SH, El-Ebeedy DA, Abdel Azeiz AZ. Identification of potential antiviral compounds from Egyptian sea stars against MERS-CoV with the in vitro and in silico experiments. Nat Prod Res 2024:1-7. [PMID: 38563220 DOI: 10.1080/14786419.2024.2335361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
Recently, the world faced many epidemics which were caused by viral respiratory pathogens. Marine creatures including Asteroidea class have been one of the recent research topics due to their diverse and complex secondary metabolites. Some of these constituents exhibit antiviral activities. The present study aimed to extract and identify the potential antiviral compounds from Pentaceraster cumingi, Astropecten polyacanthus and Pentaceraster mammillatus. The results showed that promising activity of the methanolic extract of P. cumingi with 50% inhibitory concentration (IC50) of 3.21 mg/ml against MERS-CoV with a selective index (SI) of 13.975. The biochemical components of the extracts were identified by GC/MS analysis. The Molecular docking study highlighted the virtual mechanism of binding the identified compounds towards three PDB codes of MERS-CoV non-structural protein 10/16. Interestingly, 2-mono Linolein showed promising binding energy of -14.75 Kcal/mol with the second PDB code (5YNI) and -15.22 Kcal/mol with the third PDB code (5YNQ).
Collapse
Affiliation(s)
- Nadia I Okasha
- College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October, Egypt
| | | | - Mohamed S Nafie
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Noura M Abo Shama
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Sara H Mahmoud
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Dalia A El-Ebeedy
- College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October, Egypt
| | - Ahmed Z Abdel Azeiz
- College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October, Egypt
| |
Collapse
|