1
|
Alli VJ, Singh SK, Darna M, Suresh V, Sule SA, Jangam A, Kattula B, Pusarapu SL, Thomas J, Sardana Y, Gundla KP, Burra AG, Chandra Sekhar K, Patnaik SS, Reddi B, Muralidharan K, Bokara KK, Addlagatta A, Jadav SS. Development of acylhydrazone linked thiazoles as non-covalent dual inhibitors of SARS-CoV-2 proteases. Eur J Med Chem 2025; 290:117509. [PMID: 40132498 DOI: 10.1016/j.ejmech.2025.117509] [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: 11/20/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
SARS-CoV-2's papain-like protease (PLPro) and main protease (MPro) are essential for viral maturation and replication. Currently, Paxlovid is recommended to treat viral infections, but the emergence of Nirmatrelvir resistance new variants poses serious global risks. Dual targeting agents restrict viral replication, act on other crucial viral pathways, or exert simultaneous protease inhibition, increasing the complexity for the virus to develop resistance, and the design of dual inhibitors is an attractive strategy. Herein, we present research on new thiazole-aryl and thiazole-ester compounds that function as cysteine specific non-covalent competitive dual inhibitors of SARS-CoV-2's papain-like protease (PLPro) and main protease (MPro). Twelve of the 36 compounds demonstrated dual inhibition in the range of nanomolar to low micromolar concentrations while five others exhibit selective PLPro inhibition. Minimal cytotoxicity against two mammalian cell lines and no oral toxicity in rats (LD50 > 2000 mg/kg) were observed. SARS-CoV-2 viral load was successfully reduced by several compounds tested. N-acyl hydrazone (NAH)-thiazole core while forms π-π interactions with the catalytic histidine side chain (H41) in MPro active site, it exhibits a series of hydrophobic and hydrophilic interaction in the interface of BL2 loop and the active site of PLPro. Non-covalent dual inhibition demonstrated by novel NAH-thiazole derivatives in this study provides a path for the development of efficient antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Vidya Jyothi Alli
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Shubham Kumar Singh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Mounika Darna
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Vavilapalli Suresh
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Swapnil Anil Sule
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Aruna Jangam
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bhavita Kattula
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Sarva Lakshmi Pusarapu
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Jessie Thomas
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Yogesh Sardana
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Krishna Prasad Gundla
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Amarender Goud Burra
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kunta Chandra Sekhar
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Samata Sai Patnaik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bharati Reddi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kathirvel Muralidharan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kiran Kumar Bokara
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Anthony Addlagatta
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Surender Singh Jadav
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| |
Collapse
|
2
|
Rakesh S, Behera K, Krishnan A. Unveiling the structural and functional implications of uncharacterized NSPs and variations in the molecular toolkit across arteriviruses. NAR Genom Bioinform 2025; 7:lqaf035. [PMID: 40213365 PMCID: PMC11983283 DOI: 10.1093/nargab/lqaf035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/16/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Despite considerable scrutiny of mammalian arterivirus genomes, their genomic architecture remains incomplete, with several unannotated non-structural proteins (NSPs) and the enigmatic absence of methyltransferase (MTase) domains. Additionally, the host range of arteriviruses has expanded to include seven newly sequenced genomes from non-mammalian hosts, which remain largely unannotated and await detailed comparisons alongside mammalian isolates. Utilizing comparative genomics approaches and comprehensive sequence-structure analysis, we provide enhanced genomic architecture and annotations for arterivirus genomes. We identified the previously unannotated C-terminal domain of NSP3 as a winged helix-turn-helix domain and classified NSP7 as a new small β-barrel domain, both likely involved in interactions with viral RNA. NSP12 is identified as a derived variant of the N7-MTase-like Rossmann fold domain that retains core structural alignment with N7-MTases in Nidovirales but likely lacks enzymatic functionality due to the erosion of catalytic residues, indicating a unique role specific to mammalian arteriviruses. In contrast, non-mammalian arteriviruses sporadically retain a 2'-O-MTase and an exonuclease (ExoN) domain, which are typically absent in mammalian arteriviruses, highlighting contrasting evolutionary trends and variations in their molecular toolkit. Similar lineage-specific patterns are observed in the diversification of papain-like proteases and structural proteins. Overall, the study extends our knowledge of arterivirus genomic diversity and evolution.
Collapse
Affiliation(s)
- Siuli Rakesh
- Department of Biological Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Berhampur 760010, India
| | - Kshitij Behera
- Department of Biological Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Berhampur 760010, India
| | - Arunkumar Krishnan
- Department of Biological Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Berhampur 760010, India
| |
Collapse
|
3
|
Lugano D, Mwangi K, Mware B, Kibet G, Osiany S, Kiritu E, Dobi P, Muli C, Njeru R, de Oliveira T, Njenga MK, Routh A, Oyola SO. Characterization of SARS-CoV-2 intrahost genetic evolution in vaccinated and non-vaccinated patients from the Kenyan population. J Virol 2025:e0048225. [PMID: 40326760 DOI: 10.1128/jvi.00482-25] [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: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 05/07/2025] Open
Abstract
Vaccination is a key control measure of coronavirus disease 2019 by preventing severe effects of disease outcomes, reducing hospitalization rates and death, and increasing immunity. However, vaccination can affect the evolution and adaptation of SARS-CoV-2 largely through vaccine-induced immune pressure. Here, we investigated intrahost recombination and single nucleotide variations (iSNVs) on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome in non-vaccinated and vaccinated sequences from the Kenyan population to profile intrahost viral genetic evolution and adaptations driven by vaccine-induced immune pressure. We identified recombination hotspots in the S, N, and ORF1a/b genes and showed the genetic evolution landscape of SARS-CoV-2 by comparing within- and inter-wave recombination events from the beginning of the pandemic (June 2020 to December 2022) in Kenya. We further reveal differential expression of recombinant RNA species between vaccinated and non-vaccinated individuals and perform an in-depth analysis of iSNVs to identify and characterize the functional properties of non-synonymous mutations found in ORF-1 a/b, S, and N genes. Lastly, we detected a minority variant in non-vaccinated patients in Kenya, with an immune escape mutation S255F of the spike gene, and showed differential recombinant RNA species. Overall, this work identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2, which could have significant implications for virus evolution, virulence, and immune escape.IMPORTANCEThe impact of vaccination on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genetic diversity in Kenya and much of Africa remains unknown. This can be attributed to lower sequencing rates; however, this information is relevant to improvement in vaccine and antiviral research. In this study, we investigated how vaccination and SARS-CoV-2 transmission waves affect intrahost non-homologous recombination and single nucleotide variations (iSNVs). We identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2, which could have significant implications for virus evolution, virulence, and immune escape. We also demonstrate a methodology for studying genetic changes in a pathogen by a simultaneous analysis of both intrahost single nucleotide variations and recombination events. The study reveals the diversity of SARS-CoV-2 in Kenya and highlights the need for sustained genomic surveillance in Kenya and Africa to better understand how the virus evolves. Such surveillance ensures detection of drifts in evolution, allowing information for updates in vaccines, policy making, and containment of future variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Doreen Lugano
- International Livestock Research Institute, Nairobi, Kenya
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kennedy Mwangi
- International Livestock Research Institute, Nairobi, Kenya
| | - Bernard Mware
- International Livestock Research Institute, Nairobi, Kenya
| | - Gilbert Kibet
- International Livestock Research Institute, Nairobi, Kenya
| | - Shebbar Osiany
- International Livestock Research Institute, Nairobi, Kenya
| | - Edward Kiritu
- International Livestock Research Institute, Nairobi, Kenya
| | - Paul Dobi
- International Livestock Research Institute, Nairobi, Kenya
| | - Collins Muli
- International Livestock Research Institute, Nairobi, Kenya
| | - Regina Njeru
- International Livestock Research Institute, Nairobi, Kenya
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - M Kariuki Njenga
- Washington State Global Health Program-Kenya, Washington State University, Pullman, Washington, USA
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Samuel O Oyola
- International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
4
|
Gul I, Isham IM, Najimudeen SM, Hassan A, Haq E, Shah RA, Ganai NA, Ahmad SM, Chikan NA, Abdul-Careem MF, Shabir N. An integrated in silico and ex vivo study identifies quinazolinedione L134716 as a potential inhibitor of infectious bronchitis virus. Vet Res Commun 2025; 49:175. [PMID: 40261587 DOI: 10.1007/s11259-025-10742-8] [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: 11/27/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Infectious Bronchitis Virus (IBV) poses a persistent threat to poultry health and productivity, resulting in substantial economic losses. Despite the deployment of live attenuated and inactivated vaccines, effective control of IBV remains challenging, emphasizing the need for alternative strategies to manage infections. This study identifies dual inhibitors targeting the main protease (Mpro) and papain-like protease (PLpro) of infectious bronchitis virus (IBV) using a combinatorial in silico and ex vivo approach. Screening of the MyriaScreen Diversity Library II, comprising 10,000 diverse small molecules, resulted in the selection of two promising compounds, ST092577 and L134716, based on their strong and stable interactions with both proteases. Molecular dynamics (MD) simulations further confirmed the stability of these complexes, with their binding interactions validated through MM-PBSA binding free energy calculations. Ex vivo validation utilizing tracheal organ cultures and quantitative PCR demonstrated that 50 µM of L134716 (4-(4-(benzyloxy)ph)-7,7-dimethyl-4,6,7,8-tetrahydro-2,5(1 H,3 H)-quinazolinedione) significantly reduced the IBV genome load in infected tracheal rings. This reduction in viral load was further corroborated by immunohistochemical analysis. These findings underscore the promising potential of targeting key viral proteases Mpro and PLpro as part of alternative therapeutic strategies against IBV infections in poultry. While the results are encouraging, additional in ovo and in vivo studies are necessary to validate these findings and further explore the efficacy of L134716 in practical applications.
Collapse
Affiliation(s)
- Irfan Gul
- Laboratory of Vaccine Biotechnology, Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India
- Department of Biotechnology, University of Kashmir, Srinagar, 190006, India
| | - Ishara M Isham
- Health Research Innovation Centre, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2 N 4 N1, Canada
| | - Shahnas M Najimudeen
- Health Research Innovation Centre, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2 N 4 N1, Canada
| | - Amreena Hassan
- Laboratory of Vaccine Biotechnology, Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India
- Department of Biotechnology, University of Kashmir, Srinagar, 190006, India
| | - Ehtishamul Haq
- Department of Biotechnology, University of Kashmir, Srinagar, 190006, India
| | - Riaz Ahmad Shah
- Laboratory of Vaccine Biotechnology, Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India
| | - Nazir Ahmad Ganai
- Laboratory of Vaccine Biotechnology, Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India
| | - Syed Mudasir Ahmad
- Laboratory of Vaccine Biotechnology, Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India
| | - Naveed Anjum Chikan
- Division of Computational Biology, Daskdan Innovations, PVT Ltd., Kashmir, 190006, India
| | - Mohamed Faizal Abdul-Careem
- Health Research Innovation Centre, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2 N 4 N1, Canada.
| | - Nadeem Shabir
- Laboratory of Vaccine Biotechnology, Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology, Shuhama, Kashmir, 190006, India.
| |
Collapse
|
5
|
Govender S, Morgan E, Ramahala R, Lobb K, Bishop NT, Tastan Bishop Ö. Transfer learning towards predicting viral missense mutations: A case study on SARS-CoV-2. Comput Struct Biotechnol J 2025; 27:1686-1692. [PMID: 40352476 PMCID: PMC12063013 DOI: 10.1016/j.csbj.2025.04.029] [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: 02/22/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Understanding viral evolution and predicting future mutations are crucial for overcoming drug resistance and developing long-lasting treatments. Previously, we established machine learning (ML) models using dynamic residue network (DRN) metric data and leveraging a vast amount of existing mutation data from the SARS-CoV-2 main protease (Mpro). Here, we sought to assess the generalizability and robustness of the current models across other SARS-CoV-2 proteins. To achieve this, for the first time, we employed a transfer learning (TL) approach, allowing us to determine the extent to which Mpro trained models could be applied to other SARS-CoV-2 proteins. The TL results were highly promising, with artificial neural network (ANN) and random forest (RF) correlation coefficients for Mpro closely matching those of NSP10, NSP16, and PLpro. The ANN |R| value for Mpro was 0.564, while NSP10, NSP16, and PLpro had values of 0.533, 0.527, and 0.464, respectively. Similarly, the RF |R| value for Mpro was 0.673, compared to 0.457, 0.460, and 0.437 for NSP10, NSP16, and PLpro, respectively. Interestingly, we did not observe a strong correlation for the spike (S) protein monomer and its domains. The low p-values that are associated with the correlation |R| values show that the linear correlations between predicted and actual mutation frequencies are statistically significant. This indicates that TL may generalize well across structurally related viral proteins using DRN-derived ML model from Mpro. Overall, we aim to develop a universal ML model for predicting missense mutation frequencies in viral proteins, and this study lays the foundation for that goal.
Collapse
Affiliation(s)
- Shaylyn Govender
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6139, South Africa
| | - Emily Morgan
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6139, South Africa
| | - Rabelani Ramahala
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6139, South Africa
| | - Kevin Lobb
- Department of Chemistry, Rhodes University, Makhanda 6139, South Africa
| | - Nigel T. Bishop
- Department of Pure and Applied Mathematics, Rhodes University, Makhanda 6139, South Africa
- National Institute for Theoretical and Computational Studies (NITheCS), South Africa
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6139, South Africa
- National Institute for Theoretical and Computational Studies (NITheCS), South Africa
| |
Collapse
|
6
|
Chen J, Lin Y, Gao C, Wang Z, Xu Y, Zhao Y, Xie C, Liu C, Zhou N, Shan W, Zhuang W, Qin H, Shi C, Liu R, Wang Z, Xing P, Zhu J, Wang B, Li X, Shi D. Design, synthesis and activity evaluation of dual-target inhibitors against papain-like and main proteases of porcine epidemic diarrhea virus. J Adv Res 2025:S2090-1232(25)00265-6. [PMID: 40252827 DOI: 10.1016/j.jare.2025.04.021] [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: 01/18/2025] [Revised: 03/29/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025] Open
Abstract
INTRODUCTION Porcine epidemic diarrhea (PED), caused by porcine epidemic diarrhea virus (PEDV), threatens the global pig industry due to lack of drugs. PEDV replication relies on PLpro and Mpro, which are crucial targets for inhibitors. Additionally, PLpro plays a role in modulating the host's immune response, and the inhibition of PLpro exhibits significant anti-inflammatory properties. OBJECTIVES A series of dual-targeted inhibitors of Mpro and PLpro were designed and synthesized, and their antiviral and anti-inflammatory activities were subsequently evaluated in vitro and in vivo. METHODS Dual-targeted inhibitors of Mpro and PLpro were designed by merging two series of Mpro inhibitors and PLpro inhibitors. sixty-four compounds were synthesized and screened in vitro by FRET for inhibitory activities and by RT-qPCR for antiviral activity on Vero-E6 cells. The anti-PEDV activity of f2 on Vero-E6 cells and IPEC-J2 cells was further confirmed by immunofluorescence. The mechanism by which f2 inhibited PEDV-induced inflammation was investigated by Western blot and RT-qPCR. The anti-colitis activity of f2 was verified in vivo. RESULTS Among the sixty-four synthesized compounds, seventeen potent dual-targeted inhibitors of PLpro and Mpro were identified with IC50 values less than 10 μM. Six compounds demonstrated excellent antiviral activity and safety in cell-based assays. The most potent compound f2 inhibited PEDV replication in Vero-E6 and IPEC-J2 cells with EC50 values of 1.17 ± 0.73 μM and 2.02 ± 0.56 μM, respectively, without cytotoxicity (CC50 > 800 μM). Moreover, f2 was found to inhibit the inflammatory response induced by PEDV infection via suppressing TLR2/PI3K/Akt/NF-κB signaling pathway. Oral f2 attenuated colitis by decreasing p65 phosphorylation, a major PEDV mortality cause. The in vivo acute toxicity test showed that oral administration of f2 did not affect the body weight and internal organs of mice. CONCLUSIONS In summary, a potent dual-targeted inhibitor of PLpro and Mpro, f2, was designed, synthesized, and found to be effective in the inhibition of PEDV replication and inflammatory response in vitro and in vivo.
Collapse
Affiliation(s)
- Jiashu Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Yuxi Lin
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Chenxia Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Zhuoya Wang
- GOSCI Technology Group, Qingdao 266237 Shandong, PR China
| | - Yue Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Yuanyuan Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Chao Xie
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Chao Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Nan Zhou
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Wenlong Shan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Wenli Zhuang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Hongyun Qin
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Cong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Ruihua Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Zemin Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Pan Xing
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Jiqiang Zhu
- Lead High Technology (QingDao) Co., Ltd., Qingdao 266237 Shandong, PR China
| | - Bokan Wang
- Shandong Lead High Biotechnology Co., Ltd., Jinan 250031 Shandong, PR China
| | - Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China.
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237 Shandong, PR China.
| |
Collapse
|
7
|
M Bader S, Calleja DJ, Devine SM, Kuchel NW, Lu BGC, Wu X, Birkinshaw RW, Bhandari R, Loi K, Volpe R, Khakham Y, Au AE, Blackmore TR, Mackiewicz L, Dayton M, Schaefer J, Scherer L, Stock AT, Cooney JP, Schoffer K, Maluenda A, Kleeman EA, Davidson KC, Allison CC, Ebert G, Chen G, Katneni K, Klemm TA, Nachbur U, Georgy SR, Czabotar PE, Hannan AJ, Putoczki TL, Tanzer M, Pellegrini M, Lechtenberg BC, Charman SA, Call MJ, Mitchell JP, Lowes KN, Lessene G, Doerflinger M, Komander D. A novel PLpro inhibitor improves outcomes in a pre-clinical model of long COVID. Nat Commun 2025; 16:2900. [PMID: 40180914 PMCID: PMC11969009 DOI: 10.1038/s41467-025-57905-4] [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: 12/18/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 has highlighted the vulnerability of a globally connected population to zoonotic viruses. The FDA-approved coronavirus antiviral Paxlovid targets the essential SARS-CoV-2 main protease, Mpro. Whilst effective in the acute phase of a COVID infection, Paxlovid cannot be used by all patients, can lead to viral recurrence, and does not protect against post-acute sequelae of COVID-19 (PASC), commonly known as long COVID, an emerging significant health burden that remains poorly understood and untreated. Alternative antivirals that are addressing broader patient needs are urgently required. We here report our drug discovery efforts to target PLpro, a further essential coronaviral protease, for which we report a novel chemical scaffold that targets SARS-CoV-2 PLpro with low nanomolar activity, and which exhibits activity against PLpro of other pathogenic coronaviruses. Our lead compound shows excellent in vivo efficacy in a mouse model of severe acute disease. Importantly, our mouse model recapitulates long-term pathologies matching closely those seen in PASC patients. Our lead compound offers protection against a range of PASC symptoms in this model, prevents lung pathology and reduces brain dysfunction. This provides proof-of-principle that PLpro inhibition may have clinical relevance for PASC prevention and treatment going forward.
Collapse
Affiliation(s)
- Stefanie M Bader
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Dale J Calleja
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Shane M Devine
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - Nathan W Kuchel
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Bernadine G C Lu
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Xinyu Wu
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Richard W Birkinshaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Reet Bhandari
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Katie Loi
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Rohan Volpe
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Yelena Khakham
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Amanda E Au
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Timothy R Blackmore
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Merle Dayton
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jan Schaefer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Lena Scherer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Angus T Stock
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - James P Cooney
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Kael Schoffer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ana Maluenda
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Kathryn C Davidson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Cody C Allison
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gregor Ebert
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Theresa A Klemm
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ueli Nachbur
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Smitha Rose Georgy
- Anatomic Pathology - Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Werribee, VIC, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Tracy L Putoczki
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Maria Tanzer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Marc Pellegrini
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, NSW, Australia
| | - Bernhard C Lechtenberg
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Melissa J Call
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Jeffrey P Mitchell
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Kym N Lowes
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Guillaume Lessene
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia.
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - David Komander
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
8
|
Garcia Lopez V, Plate L. Comparative Interactome Profiling of Nonstructural Protein 3 Across SARS-CoV-2 Variants Emerged During the COVID-19 Pandemic. Viruses 2025; 17:447. [PMID: 40143373 PMCID: PMC11946765 DOI: 10.3390/v17030447] [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: 03/01/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
SARS-CoV-2 virus and its variants remain a global health threat, due to their capacity for rapid evolution. Variants throughout the COVID-19 pandemic exhibited variations in virulence, impacting vaccine protection and disease severity. Investigating nonstructural protein variants is critical to understanding viral evolution and manipulation of host protein interactions. We focus on nonstructural protein 3 (nsp3), with multiple domains with different activities, including viral polyprotein cleavage, host deubiquitylation, de-ISGylation, and double-membrane vesicle formation. Using affinity purification-mass spectrometry (AP-MS), we identify differential protein interactions in nsp3 caused by mutations found in variants identified between 2019 and 2024: Alpha 20I, Beta 20H, Delta 21I, Delta 21J, Gamma 20J, Kappa 21B, Lambda 21G, Omicron 21K, and Omicron 21L. A small set of amino acid substitutions in the N-terminal region of nsp3 (nsp3.1) could be traced to increased interactions with RNA-binding proteins, which are vital in viral replication. Meanwhile, variants of the central region of nsp3 (nsp3.2) were found to share interactions with protein quality control machinery, including ER-associated degradation. In this construct, shared trends in interactor enrichment are observed between Omicron 21K and Delta 21I. These results underscore how minor mutations reshape host interactions, emphasizing the evolutionary arms race between the host and virus. We provide a roadmap to track the interaction changes driven by SARS-CoV-2 variant evolution.
Collapse
Affiliation(s)
- Valeria Garcia Lopez
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37240, USA;
| | - Lars Plate
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37240, USA;
- Department of Chemistry, Vanderbilt University, Nashville, TN 37240, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
9
|
Lacasse É, Dubuc I, Gudimard L, Andrade ACDSP, Gravel A, Greffard K, Chamberland A, Oger C, Galano JM, Durand T, Philipe É, Blanchet MR, Bilodeau JF, Flamand L. Delayed viral clearance and altered inflammatory responses affect severity of SARS-CoV-2 infection in aged mice. Immun Ageing 2025; 22:11. [PMID: 40075368 PMCID: PMC11899864 DOI: 10.1186/s12979-025-00503-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Epidemiological investigations consistently demonstrate an overrepresentation of the elderly in COVID-19 hospitalizations and fatalities, making the advanced age as a major predictor of disease severity. Despite this, a comprehensive understanding of the cellular and molecular mechanisms explaining how old age represents a major risk factor remain elusive. To investigate this, we compared SARS-CoV-2 infection outcomes in young adults (2 months) and geriatric (15-22 months) mice. Both groups of K18-ACE2 mice were intranasally infected with 500 TCID50 of SARS-CoV-2 Delta variant with analyses performed on days 3, 5, and 7 post-infection (DPI). Analyses included pulmonary cytokines, lung RNA-seq, viral loads, lipidomic profiles, and histological assessments, with a concurrent evaluation of the percentage of mice reaching humane endpoints. The findings unveiled notable differences, with aged mice exhibiting impaired viral clearance, reduced survival, and failure to recover weight loss due to infection. RNA-seq data suggested greater lung damage and reduced respiratory function in infected aged mice. Additionally, elderly-infected mice exhibited a deficient antiviral response characterized by reduced Th1-associated mediators (IFNγ, CCL2, CCL3, CXCL9) and diminished number of macrophages, NK cells, and T cells. Furthermore, mass-spectrometry analysis of the lung lipidome indicated altered expression of several lipids with immunomodulatory and pro-resolution effects in aged mice such as Resolvin, HOTrEs, and NeuroP, but also DiHOMEs-related ARDS. These findings indicate that aging affects antiviral immunity, leading to prolonged infection, greater lung damage, and poorer clinical outcomes. This underscores the potential efficacy of immunomodulatory treatments for elderly subjects experiencing symptoms of severe COVID-19.
Collapse
Affiliation(s)
- Émile Lacasse
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
- Département de Microbiologie, Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Isabelle Dubuc
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
| | - Leslie Gudimard
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
| | - Ana Claudia Dos S P Andrade
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
| | - Annie Gravel
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
| | - Karine Greffard
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
| | | | - Camille Oger
- Institut Des Biomolécules Max Mousseron, UMR 5247, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Marie Galano
- Institut Des Biomolécules Max Mousseron, UMR 5247, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Thierry Durand
- Institut Des Biomolécules Max Mousseron, UMR 5247, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Éric Philipe
- Département de Chirurgie, Faculté de Médecine, Université, Québec, QC, Canada
| | - Marie-Renée Blanchet
- Département de Médecine, Faculté de Médecine, Université, Québec, QC, Canada
- Centre de Recherche de L'Institut de Cardiologie de Québec, Université, Québec, QC, Canada
| | - Jean-François Bilodeau
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada
- Département de Médecine, Faculté de Médecine, Université, Québec, QC, Canada
| | - Louis Flamand
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier, Universitaire de Québec- Université Laval, Québec, QC, Canada.
- Département de Microbiologie, Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
10
|
De Falco A, Greene-Cramer R, Surina BA, Zakian S, Acton TB, Ramelot TA, Montelione GT. From Plasmid to Pure Protein: Production and Characterization of SARS-CoV-2 PL pro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642282. [PMID: 40161688 PMCID: PMC11952365 DOI: 10.1101/2025.03.09.642282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Papain-like protease (PLpro) from SARS-CoV-2 is a high-priority target for COVID-19 antiviral drug development. We present protocols for PLpro production in Escherichia coli. PLpro expressed as a fusion with the Saccharomyces cerevisiae Smt3 protein (SUMO), is purified and obtained in its native form upon hydrolysis, with yields as high as 38 mg L-1. The protocol also provides isotope-enriched samples suitable for NMR studies. Protocols are also presented for PLpro characterization by mass spectrometry, 1D 19F-NMR and 2D heteronuclear NMR, and a fluorescence-based enzyme assay.
Collapse
Affiliation(s)
- Anna De Falco
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Technical contacts
| | - Rebecca Greene-Cramer
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Technical contacts
| | - Ben A. Surina
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
| | - Suren Zakian
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
| | - Thomas B. Acton
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
| | - Theresa A. Ramelot
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
| | - Gaetano T. Montelione
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, 12180, USA
- Lead contact
| |
Collapse
|
11
|
Lugano D, Mwangi K, Mware B, Kibet G, Osiany S, Kiritu E, Dobi P, Muli C, Njeru R, de Oliveira T, Njenga MK, Routh A, Oyola SO. Characterization of SARS-CoV-2 intrahost genetic evolution in vaccinated and non-vaccinated patients from the Kenyan population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323296. [PMID: 40093225 PMCID: PMC11908290 DOI: 10.1101/2025.03.03.25323296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Vaccination is a key control measure of COVID-19 by preventing severe effects of disease outcomes, reducing hospitalization rates and death, and increasing immunity. However, vaccination can affect the evolution and adaptation of SARS-CoV-2, largely through vaccine-induced immune pressure. Here we investigated intrahost recombination and single nucleotide variations (iSNVs) on the SARS-CoV-2 genome in non-vaccinated and vaccinated sequences from the Kenyan population to profile intrahost viral genetic evolution and adaptations driven by vaccine-induced immune pressure. We identified recombination hotspots in the S, N, and ORF1a/b genes and showed the genetic evolution landscape of SARS-CoV-2 by comparing within-wave and inter-wave recombination events from the beginning of the pandemic (June 2020) to (December 2022) in Kenya. We further reveal differential expression of recombinant RNA species between vaccinated and non-vaccinated individuals and perform an in-depth analysis of iSNVs to identify and characterize the functional properties of non-synonymous mutations found in ORF-1 a/b, S, and N genes. Lastly, we detected a minority variant in non-vaccinated patients in Kenya, with an immune escape mutation S255F of the spike gene and showed differential recombinant RNA species. Overall, this work identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2 which could have significant implications for virus evolution, virulence, and immune escape.
Collapse
Affiliation(s)
- Doreen Lugano
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, 77550, USA
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230 Kilifi, Kenya
| | - Kennedy Mwangi
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Bernard Mware
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Gilbert Kibet
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Shebbar Osiany
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Edward Kiritu
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Paul Dobi
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Collins Muli
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Regina Njeru
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - M. Kariuki Njenga
- Washington State Global Health Program-Kenya, Washington State University, Nairobi 00200, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA 99164, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, 77550, USA
- Dept Immunology and Microbiology, Scripps Research, La Jolla, CA, 92037
| | - Samuel O. Oyola
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| |
Collapse
|
12
|
Trivedi A, Kushwaha T, Ishani, Vrati S, Gupta D, Kayampeta SR, Parvez MK, Inampudi KK, Appaiahgari MB, Sehgal D. Psoralidin acts as a dual protease inhibitor against PL pro and M pro of SARS-CoV-2. FEBS J 2025; 292:1106-1123. [PMID: 39745898 DOI: 10.1111/febs.17380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/08/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
The emergence of new coronavirus variants and concerns about vaccine effectiveness against these novel variants emphasize the need for broad-spectrum therapeutics targeting conserved coronaviral non-structural proteins. Accordingly, a virtual library of 178 putative inhibitors targeting SARS-CoV-2 Papain-like protease (PLpro) was compiled through a systematic review of published literature and subsequently screened using molecular docking. Selected hits were analyzed for protease inhibitory activities, binding strength, and antiviral activities against HCoV229E-based surrogate system and subsequently against SARS-CoV-2 for validation. Differences in potential modes of action were investigated using an HCoV229E-based system, combined with in silico and biophysical methods against SARS-CoV-2 system. Of the 178 hits, 13 molecules showed superior docking scores against PLpro and met the inclusion criteria for further investigations. Of these, seven showed notable inhibitory activities against PLpro. Particularly, both Psoralidin and Corylifol-A exhibited superior and, importantly, dual activities against SARS-CoV-2 Mpro. Both molecules were found to be biologically active against HCoV229E and SARS-CoV-2; however, Psoralidin exhibited more consistent effects and was relatively well-tolerated. Detailed in silico analyses of their interactions with the two proteases identified differences in their modes of action, primarily due to differences in their binding of PLpro. Based on these findings, we propose Psoralidin as a potential candidate for further development as a broad-spectrum antiviral and Corylifol-A as an ideal candidate for lead optimization.
Collapse
Affiliation(s)
- Aditya Trivedi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Tushar Kushwaha
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Ishani
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Sudhanshu Vrati
- Laboratory of Virology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| | - Dharmender Gupta
- Laboratory of Virology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| | | | - Mohammad Khalid Parvez
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Mohan Babu Appaiahgari
- Yenepoya (deemed to be) University, Mangalore, India
- R&D Wing, Srikara Biologicals Pvt. Ltd., Tirupati, India
| | - Deepak Sehgal
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| |
Collapse
|
13
|
Aziz S, Khan S, Karkashan A, Asim N, Bukhari K, Mohammad Almousa R, Ali Shah Z, Allemailem KS, Al-Megrin WAI. Exploring natural compounds and synthetic derivatives as potential inhibitors of SARS-CoV-2 PLpro: a computational approach with enzyme inhibition and cytotoxicity assessment. J Biomol Struct Dyn 2025:1-21. [PMID: 39972932 DOI: 10.1080/07391102.2024.2333456] [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: 12/10/2023] [Accepted: 03/15/2024] [Indexed: 02/21/2025]
Abstract
To address coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), there is a pressing need for direct-acting antiviral medications. Papain-like protease (PLpro) domain of Nsp3 from SARS-CoV-2 is central to viral replication and detrimentally affects the host immune response by cleaving ubiquitin and interferon-stimulated gene 15 proteins. Consequently, targeting PLpro with small-molecule therapeutics offers a promising strategy to effectively inhibit the virus and mitigate its impact on the host's immune system. Employing a multifaceted computational approach, we identified three phytochemicals demonstrating substantial molecular interactions and binding affinity [α-lapachone (C1) -37.82 ± 0.08 kcal/mol, Lapachol (C2) -48.56 ± 0.04 kcal/mol and Peshawarquinone (C3) -46.64 ± 0.03 kcal/mol)] with PLpro. Molecular dynamics simulations indicated that these compounds formed stable complexes with the naphthalene-inhibitor binding site, inducing a closed PLpro conformation and altering its normal function. Besides, they are predicted to possess favorable physicochemical, pharmacokinetics and drug-like properties. Among these compounds, C3 exhibited the most potent inhibitory potential (IC50 value: 1.76 ± 0.17 µM), followed by C2 (IC50 value; 2.50 ± 0.12 µM) and C1 (IC50 value: 3.42 ± 0.17 µM). Furthermore, evaluating the cytotoxicity of C1-C3 on human BJ cells revealed minimal harmful effects at a 50 µM dosage. These findings suggest that evaluated compounds hold therapeutic promise and could become potential drug candidates for treating SARS-CoV-2.
Collapse
Affiliation(s)
- Shahkaar Aziz
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Sana Khan
- Department of Human Nutrition and Dietetics, Women University Mardan, Mardan, Pakistan
| | - Alaa Karkashan
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Noreen Asim
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Khulud Bukhari
- Department of Microbiology and Parasitology, College of Veterinary Medicine, King Faisal University, Hofuf, Al-Ahsa, Saudi Arabia
| | | | - Zafar Ali Shah
- Department of Agricultural Chemistry and Biochemistry, The University of Agriculture, Peshawar, Pakistan
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Wafa Abdullah I Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Pandey K, Lewis DSM, Heo K, Acharya A, Fields T, Gowda K, Dean G, Rayalam S, Byrareddy SN, Mody V, Taval S. 9-aminominocycline potentiates the efficacy of EIDD-1931 and PF-332 by targeting the papain like protease enzyme of SARS-CoV-2. Sci Rep 2025; 15:5671. [PMID: 39955340 PMCID: PMC11830066 DOI: 10.1038/s41598-025-89717-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
The 3-chymotrypsin-like protease (3CLpro), papain-like protease (PLpro), and RNA-dependent RNA polymerase (RdRp) are key enzymes in SARS-CoV-2 replication and serve as critical targets for an antiviral drug. Currently, Paxlovid® and Lagevrio™ specifically target 3CLpro and RdRp, respectively, for COVID-19 treatment. However, no antivirals target for the SARS-CoV-2 PLpro enzyme, essential for viral replication and suppression of the host antiviral immune response. This study identified 9-aminominocycline (9-AMN) as a potent inhibitor of SARS-CoV-2 PLpro. Unlike the parent compound minocycline, 9-AMN inhibits PLpro's proteolytic and deubiquitinase activities by approximately 90%, with IC50 values of 4.15 µM and 4.55 µM, respectively, while showing no effect on the enzymatic activity of 3CLpro or RdRp. Enzyme kinetics reveal that 9-AMN functions as a mixed PLpro inhibitor and binds to its active site, disrupting its function as predicted by computer modeling. Furthermore, 9-AMN demonstrates, efficacy against the Delta and Omicron variants, with EC50 values of 1.04 µM and 2.35 µM, respectively. When combined with EIDD-1931 (an active form of molnupiravir) or nirmatrelvir (PF-332), 9-AMN exhibits synergistic effects, significantly reducing the doses required to inhibit the Omicron variant. In conclusion, 9-AMN inhibits SARS-CoV-2 replication, and PLpro activity, highlighting its potential as a promising candidate for COVID-19 treatment strategies.
Collapse
Affiliation(s)
- Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE, 68198, USA
| | - Devin Shane M Lewis
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA
| | - Kyeongin Heo
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE, 68198, USA
| | - Travis Fields
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA
| | - Kritika Gowda
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA
- The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - George Dean
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA
| | - Srujana Rayalam
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE, 68198, USA.
| | - Vicky Mody
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA.
| | - Shashidharamurthy Taval
- Department of Pharmaceutical Sciences, School of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA, 30024, USA.
| |
Collapse
|
15
|
Rouch CC, Chatterjee AK, McCarty C, Song L, Chu A, Johnson K, Heacock M, Riva L, McNamara CW, Wolff KC, Greene-Cramer R, Falco AD, Montelione GT, Grabovyi GA. Synthesis and Structure-Activity Relationship of Covalent Inhibitors of SARS-CoV-2 Papain-Like Protease with Antiviral Potency. Bioorg Med Chem Lett 2025; 116:130034. [PMID: 39580003 PMCID: PMC11936407 DOI: 10.1016/j.bmcl.2024.130034] [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: 09/29/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
The papain-like protease (PLpro) is a highly conserved domain encoded by the coronavirus (CoV) genome and it plays an essential role in the replication and maturation of the virus in addition to weakening host immune response. Due to the virus's reliance on PLpro for survival and propagation, small-molecule inhibitors of PLpro serve as an attractive model for direct-acting antiviral therapeutic agents against SARS-CoV-2. Building upon existing work aimed at designing covalent inhibitors against PLpro, we report the synthesis and structure-activity relationship of analogs based on the known covalent inhibitor 1 (Sanders, et al.2023). To evaluate the efficacy of synthesized derivatives, we conducted enzymatic inhibition assays, SARS-CoV-2/HeLa-ACE2 cellular potency and toxicity assays, and profiled the most promising analogs via in vitro ADME and in vivo pharmacokinetic studies. Additionally, we describe computational docking of profiled compounds bound to PLpro to elucidate the structure-activity relationship of compounds based on 1 and offer suggestions for optimizing the potency and selectivity of the electrophilic warhead and improving ADME and PK properties for this chemotype. Relative to the parent compound, new designs demonstrate comparable potency and target selectivity for PLpro. The accomplished SAR campaign provides novel insight for future development of antivirals against SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine C Rouch
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Arnab K Chatterjee
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Connor McCarty
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Lirui Song
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Alan Chu
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Kristen Johnson
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Mina Heacock
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Laura Riva
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Case W McNamara
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Karen C Wolff
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA
| | - Rebecca Greene-Cramer
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Anna De Falco
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Gaetano T Montelione
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Gennadii A Grabovyi
- Calibr-Skaggs Institute for Innovative Medicines, A Division of Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
16
|
Varghese A, Liu J, Liu B, Guo W, Dong F, Patterson TA, Hong H. Analysis of Structures of SARS-CoV-2 Papain-like Protease Bound with Ligands Unveils Structural Features for Inhibiting the Enzyme. Molecules 2025; 30:491. [PMID: 39942596 PMCID: PMC11820935 DOI: 10.3390/molecules30030491] [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: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
The COVID-19 pandemic, driven by the novel coronavirus SARS-CoV-2, has drastically reshaped global health and socioeconomic landscapes. The papain-like protease (PLpro) plays a critical role in viral polyprotein cleavage and immune evasion, making it a prime target for therapeutic intervention. Numerous compounds have been identified as inhibitors of SARS-CoV-2 PLpro, with many characterized through crystallographic studies. To date, over 70 three-dimensional (3D) structures of PLpro complexed ligands have been deposited in the Protein Data Bank, offering valuable insight into ligand-binding features that could aid the discovery and development of effective COVID-19 treatments targeting PLpro. In this study, we reviewed and analyzed these 3D structures, focusing on the key residues involved in ligand interactions. Our analysis revealed that most inhibitors bind to PLpro's substrate recognition sites S3/S4 and SUb2. While these sites are highly attractive and have been extensively explored, other potential binding regions, such as SUb1 and the Zn(II) domain, are less explored and may hold untapped potential for future COVID-19 drug discovery and development. Our structural analysis provides insights into the molecular features of PLpro that could accelerate the development of novel therapeutics targeting this essential viral enzyme.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (A.V.); (J.L.); (B.L.); (W.G.); (F.D.); (T.A.P.)
| |
Collapse
|
17
|
Kuzikov M, Morasso S, Reinshagen J, Wolf M, Monaco V, Cozzolino F, Golič Grdadolnik S, Šket P, Plavec J, Iaconis D, Summa V, Corona A, Paulis A, Esposito F, Tramontano E, Monti M, Beccari AR, Manelfi C, Windshügel B, Gribbon P, Storici P, Zaliani A. Thiol-Reactive or Redox-Active: Revising a Repurposing Screen Led to a New Invalidation Pipeline and Identified a True Noncovalent Inhibitor Against Papain-like Protease from SARS-CoV-2. ACS Pharmacol Transl Sci 2025; 8:66-77. [PMID: 39816795 PMCID: PMC11729419 DOI: 10.1021/acsptsci.4c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/02/2024] [Accepted: 08/27/2024] [Indexed: 01/18/2025]
Abstract
The SARS-CoV-2 papain-like protease PLpro has multiple roles in the viral replication cycle, related to both its polypeptide cleavage function and its ability to antagonize the host immune response. Targeting the PLpro function is recognized as a promising mechanism to modulate viral replication, while supporting host immune responses. However, the development of PLpro-specific inhibitors remains challenging. Comprehensive investigations utilizing enzymatic, binding studies, and cellular assays revealed the previously reported inhibitors to act in an unspecific manner. At present, GRL-0617 and its derivatives remain the best-validated compounds with demonstrated antiviral activity in cells and in mouse models. In this study, we refer to the pitfalls of the redox sensitivity of PLpro. Using a screening-based approach to identify inhibitors of PLpro's proteolytic activity, we made extensive efforts to validate active compounds over a range of conditions and readouts, emphasizing the need for comprehensive orthogonal data when profiling putative PLpro inhibitors. The remaining active compound, CPI-169, was shown to be a noncovalent inhibitor capable of competing with GRL-0617 in NMR-based experiments, suggesting that it occupied a similar binding site and inhibited viral replication in Vero-E6 cells, opening new design opportunities for further development as antiviral agents.
Collapse
Affiliation(s)
- Maria Kuzikov
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
- School
of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| | - Stefano Morasso
- Protein
Targets for Drug Discovery Lab, Elettra-Sincrotrone
Trieste S.C.p.A., SS 14 - km 163,5 in AREA Science Park, 34149 Basovizza, Trieste, Italy
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgeri 1, 34127 Trieste, Italy
| | - Jeanette Reinshagen
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Markus Wolf
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Vittoria Monaco
- Department
of Chemical Sciences, University of Naples
“Federico II’, Comunale Cinthia 26, 80126 Naples, Italy
- CEINGE
Advanced-Biotechnologies “Franco Salvatore”, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Flora Cozzolino
- Department
of Chemical Sciences, University of Naples
“Federico II’, Comunale Cinthia 26, 80126 Naples, Italy
- CEINGE
Advanced-Biotechnologies “Franco Salvatore”, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Simona Golič Grdadolnik
- Laboratory
for Molecular Structural Dynamics, National
Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Primož Šket
- Slovenian
NMR Center, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Janez Plavec
- Slovenian
NMR Center, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Daniela Iaconis
- EXSCALATE
- Dompé Farmaceutici SpA, via Tommaso De Amicis 95, 80131 Naples, Italy
| | - Vincenzo Summa
- Department
of Pharmacy, University of Naples “Federico
II”, Via D. Montesano, 49 80131 Naples, Italy
| | - Angela Corona
- Dipartimento
di Scienze della vita e dell’ambiente, Cittadella Universitaria di Monserrato, SS-554, Monserrato, 09042 Cagliari, Italy
| | - Annalaura Paulis
- Dipartimento
di Scienze della vita e dell’ambiente, Cittadella Universitaria di Monserrato, SS-554, Monserrato, 09042 Cagliari, Italy
| | - Francesca Esposito
- Dipartimento
di Scienze della vita e dell’ambiente, Cittadella Universitaria di Monserrato, SS-554, Monserrato, 09042 Cagliari, Italy
| | - Enzo Tramontano
- Dipartimento
di Scienze della vita e dell’ambiente, Cittadella Universitaria di Monserrato, SS-554, Monserrato, 09042 Cagliari, Italy
| | - Maria Monti
- Department
of Chemical Sciences, University of Naples
“Federico II’, Comunale Cinthia 26, 80126 Naples, Italy
- CEINGE
Advanced-Biotechnologies “Franco Salvatore”, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Andrea R. Beccari
- EXSCALATE
- Dompé Farmaceutici SpA, via Tommaso De Amicis 95, 80131 Naples, Italy
| | - Candida Manelfi
- EXSCALATE
- Dompé Farmaceutici SpA, via Tommaso De Amicis 95, 80131 Naples, Italy
| | - Björn Windshügel
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
- School
of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| | - Philip Gribbon
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Paola Storici
- Protein
Targets for Drug Discovery Lab, Elettra-Sincrotrone
Trieste S.C.p.A., SS 14 - km 163,5 in AREA Science Park, 34149 Basovizza, Trieste, Italy
| | - Andrea Zaliani
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, Discovery
Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| |
Collapse
|
18
|
Thuy La VN, Kang L, Minh DDL. Enzyme kinetics model for the coronavirus main protease including dimerization and ligand binding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.01.631001. [PMID: 39803422 PMCID: PMC11722214 DOI: 10.1101/2025.01.01.631001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The coronavirus main protease (MPro) plays a pivotal role in viral replication and is the target of several antivirals against SARS-CoV-2. In some species, CRCs of MPro enzymatic activity can exhibit biphasic behavior in which low ligand concentrations activate the enzyme whereas higher ones inhibit it. While this behavior has been attributed to ligand-induced dimerization, quantitative enzyme kinetics models have not been fit to it. Here, we develop a kinetic model integrating dimerization and ligand binding. We perform a Bayesian regression to globally fit the model to multiple types of biochemical and biophysical data. The reversible covalent inhibitor GC376 strongly induces dimerization and binds to the dimer with no cooperativity. In contrast, the fluorescent peptide substrate has a minor effect on dimerization but binds to the dimer with positive cooperativity. The biphasic concentration response curve occurs because compared to substrate, the inhibitor accelerates turnover in the opposite catalytic site.
Collapse
Affiliation(s)
- Van Ngoc Thuy La
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Lulu Kang
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - David D L Minh
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
19
|
Fischer T, Frasson D, Sievers M, Riedl R. Phthalimide derivatives as a new class of papain-like protease inhibitors in SARS-CoV-2. Arch Pharm (Weinheim) 2025; 358:e2400714. [PMID: 39648568 DOI: 10.1002/ardp.202400714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) papain-like cysteine protease (PLpro) represents one of only two essential cysteine proteases involved in the regulation of viral replication. It, therefore, qualifies as a promising therapeutic target for the development of antiviral agents. We identified a previously synthesized protease inhibitor, resulting from an earlier project, as a PLpro inhibitor and crafted a structure-activity relationship around the hit, leading to the more potent inhibitors ZHAWOC6941 (17h) and ZHAWOC25153 (17o) displaying IC50 values of 8 and 7 µM, respectively. The two compounds represent a new class of PLpro inhibitors and, with single-digit micromolar IC50 values, are comparable to inhibitors found in the literature.
Collapse
Affiliation(s)
- Thomas Fischer
- Competence Center for Drug Discovery, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Wädenswil, Switzerland
| | - David Frasson
- Competence Center for Drug Discovery, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Wädenswil, Switzerland
| | - Martin Sievers
- Competence Center for Drug Discovery, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Wädenswil, Switzerland
| | - Rainer Riedl
- Competence Center for Drug Discovery, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Wädenswil, Switzerland
| |
Collapse
|
20
|
Al-Humaidi JY, Gomha SM, Nayl AA, Aly AA, Ibrahim MAA, Zaki MEA, Bräse S, Haggam RA. Synthesis, Characterization, and Molecular Modeling Studies of Novel Indenopyridazine-thiazole Molecular Hybrids. Curr Org Synth 2025; 22:79-89. [DOI: 10.2174/0115701794266795231129074028] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/11/2023] [Accepted: 10/23/2023] [Indexed: 05/14/2025]
Abstract
Background:
Previous studies have reported various biological activities of indeno-pyridazine and thiazole derivatives, including antiviral activity and CoV-19 inhibition. In this paper, the authors aimed to design, synthesize, and characterize a novel series of indenopyridazinethiazoles, starting with 2-(4-cyano-3-oxo-2,3-dihydro-9H-indeno[2,1-c]pyridazin-9-ylidene)-hydrazine-1-car-bothioamide and available laboratory reagents.
Methods:
The strategy involved the synthesis of indeno[2,1-c]pyridazincarbothioamide, followed by its reaction with various hydrazonoyl chlorides and α-halocompounds (phenacyl bromides and α-chloroketones) to obtain the desired indenopyridazinethiazole derivatives. The synthesized structures were confirmed using IR, NMR, mass spectra, elemental analysis, and alternative synthesis when possible. Docking scores and poses of thirteen synthesized compounds were examined using Auto-Dock4.2.6 software against multiple targets of SARS-CoV-2, including 3C-like protease (3CLpro), helicase, receptor binding domain (RBD), papain-like protease (PLpro), neuropilin-1 (NRP-1), RNA-dependent RNA polymerase (RdRp), and human angiotensin‐converting enzyme 2 (ACE2).
Results:
Docking predictions revealed that compound 13d exhibited high potency against 3CLpro and helicase, with docking scores of -10.9 and -10.5 kcal/mol, respectively. Compound 10c showed su-perior docking scores against RBD and ACE2, with values of -8.7 and -11.8 kcal/mol, respectively. Compounds 10a, 13c, and 7b demonstrated excellent docking scores against RdRp, PLpro, and NRP-1, with values of -10.3, -10.4, and -8.6 kcal/mol, respectively.
Conclusion:
The authors recommend further experimental assessments of compounds 13d, 10c, 10a, 13c, and 7b against SARS-CoV-2 multi-targets, considering their promising docking scores.
Collapse
Affiliation(s)
- Jehan Y. Al-Humaidi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Sobhi M. Gomha
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah, 42351, Saudi Arabia
| | - AbdElAziz A. Nayl
- Department of Chemistry, College of Science, Jouf University, Sakaka, 72341, Al Jouf, Saudi Arabia
| | - Ashraf A. Aly
- Department of Chemistry, Faculty of Science, Organic Division, Minia University, El-Minia, 61519, Menia, Egypt
| | - Mahmoud A. A. Ibrahim
- Computational Chemistry Laboratory, Department of Chemistry, Faculty of Science, Minia University, Minia, 61519, Egypt
- School of Health Sciences, University of Kwa-Zulu-Natal, Westville, Durban, 4000, South Africa
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Stefan Bräse
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76133, Karlsruhe, Germany
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Director Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Reda A. Haggam
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah, 42351, Saudi Arabia
- De-partment of Chemistry, Faculty of Science, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
21
|
Wang M, Sun X, Peng S, Wang F, Zhao K, Wang D. Deciphering the cleavage sites of 3C-like protease in Gammacoronaviruses and Deltacoronaviruses. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141057. [PMID: 39454742 DOI: 10.1016/j.bbapap.2024.141057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/26/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Coronaviruses replicate by using the 3C-like protease (3CLpro) to cleave polyprotein precursors and host proteins. However, current tools for identifying 3CLpro cleavage sites are limited, particularly in Gammacoronaviruses (GammaCoV) and Deltacoronaviruses (DeltaCoV). This study aims to fill this gap by identifying 3CLpro cleavage sites in these viruses to provide deeper insights into their pathogenic mechanisms. By integrating sequence alignments and structural model comparisons, we developed a position-specific scoring matrix (PSSM) based on self-cleavage motifs, revealing specific preferences for each residue. Utilizing AlphaFold2's predicted alignment error (PAE) and predicted local distance difference test (pLDDT), we found that most cleavage sequences are located in regions with high PAE and low pLDDT values. KEGG pathway analysis showed that potential host protein cleavage targets are mainly concentrated in pathways related to nucleo-cytoplasmic transport and endocytosis. Through in vitro cleavage experiments and mutational analysis, we identified and validated three high-scoring proteins-nucleoporin 58 (NUP58), cell division cycle 73 (CDC73), and signal transducing adaptor molecule 2 (STAM2). These findings suggest that 3CLpro not only plays a vital role in viral replication but may also influence host cell functions by cleaving host proteins. This study provides an effective tool for identifying 3CLpro cleavage sites, revealing the pathogenic mechanisms of coronaviruses, and offering new insights for developing potential therapeutic targets.
Collapse
Affiliation(s)
- Mengxue Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Xinyi Sun
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Shijiang Peng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Feifan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Kangli Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Dang Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| |
Collapse
|
22
|
Yaghi R, Wylie DC, Andrews CL, Dickert OH, Ram A, Iverson BL. An Investigation of Nirmatrelvir (Paxlovid) Resistance in SARS-CoV-2 M pro. ACS BIO & MED CHEM AU 2024; 4:280-290. [PMID: 39712205 PMCID: PMC11659887 DOI: 10.1021/acsbiomedchemau.4c00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 12/24/2024]
Abstract
The high throughput YESS 2.0 platform was used to screen a large library of SARS-CoV-2 Mpro variants in the presence of nirmatrelvir. Of the 100 individual most prevalent mutations identified in the screen and reported here, the most common were E166V, L27V, N142S, A173V, and Y154N, along with their various combinations. In vitro analysis revealed that resistance to nirmatrelvir for these individual mutations, as well as all of the combinations we analyzed, was accompanied by decreased catalytic activity with the native substrate. Importantly, the mutations we identified have not appeared as significantly enriched in SARS-CoV-2 Mpro sequences isolated from COVID-19 patients following the introduction of nirmatrelvir. We also analyzed three of the most common SARS-CoV-2 Mpro mutations that have been seen in patients recently, and only a measured increase in nirmatrelvir resistance was seen when the more recently appearing A285V is added to both P132H and K90R. Taken together, our results predict that resistance to nirmatrelvir will be slower to develop than expected based on experience with other viral protease inhibitors, perhaps due in part to the close structural correspondence between nirmatrelvir and SARS-CoV-2 Mpro's preferred substrates.
Collapse
Affiliation(s)
- Rasha
M. Yaghi
- Department
of Chemistry, The University of Texas at
Austin, Austin, Texas 78712, The United States of America
| | - Dennis C. Wylie
- Center
of Biomedical Research Support, The University
of Texas at Austin, Austin, Texas 78712, The United States of America
| | - Collin L. Andrews
- Department
of Chemistry, The University of Texas at
Austin, Austin, Texas 78712, The United States of America
| | - Olivia H. Dickert
- Department
of Chemistry, The University of Texas at
Austin, Austin, Texas 78712, The United States of America
| | - Anjana Ram
- Department
of Chemistry, The University of Texas at
Austin, Austin, Texas 78712, The United States of America
| | - Brent L. Iverson
- Department
of Chemistry, The University of Texas at
Austin, Austin, Texas 78712, The United States of America
| |
Collapse
|
23
|
Sousa BP, Mottin M, Seanego D, Jurisch CD, Rodrigues BSA, da Silva VLS, Andrade MA, Morais GS, Boerin DF, Froes TQ, Motta FN, Nonato MC, Bastos IDM, Chibale K, Gessner RK, Andrade CH. Discovery of Non-Covalent Inhibitors for SARS-CoV-2 PLpro: Integrating Virtual Screening, Synthesis, and Experimental Validation. ACS Med Chem Lett 2024; 15:2140-2149. [PMID: 39691531 PMCID: PMC11647681 DOI: 10.1021/acsmedchemlett.4c00420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
The SARS-CoV-2 pandemic has significantly challenged global public health, highlighting the need for effective therapeutic options. This study focuses on the papain-like protease (PLpro) of SARS-CoV-2, which is a critical enzyme for viral polyprotein processing, maturation, and immune evasion. We employed a combined approach that began with computational models in a virtual screening campaign, prioritizing compounds from our in-house chemical library against PLpro. Out of 81 virtual hits evaluated through enzymatic and biophysical assays, we identified a modest inhibitor featuring a naphthyridine core with an IC50 of 73.61 μM and a K i of 22 μM. Expanding our exploration, we synthesized and assessed 30 naphthyridine analogues, three of which emerged as promising noncovalent, nonpeptidomimetic inhibitors with IC50 values between 15.06 and 51.81 μM. Furthermore, in vitro ADMET assays revealed these compounds to possess moderate aqueous solubility, low cytotoxicity, and high microsomal stability, making them excellent candidates for further development targeting SARS-CoV-2 PLpro.
Collapse
Affiliation(s)
- Bruna
K. P. Sousa
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Melina Mottin
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Donald Seanego
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Christopher D. Jurisch
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Beatriz S. A. Rodrigues
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Verônica L. S. da Silva
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Milene Aparecida Andrade
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Gilberto S. Morais
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Diogo F. Boerin
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratório
de Cristalografia de Proteínas, Faculdade de Ciências
Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 05508-070, Brazil
| | - Thamires Q. Froes
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratório
de Cristalografia de Proteínas, Faculdade de Ciências
Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 05508-070, Brazil
| | - Flávia Nader Motta
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
- Faculdade
de Ceilândia, Universidade de Brasília, Brasília, Distrito
Federal 73345-010, Brazil
| | - M. Cristina Nonato
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratório
de Cristalografia de Proteínas, Faculdade de Ciências
Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 05508-070, Brazil
| | - Izabela D. M. Bastos
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Kelly Chibale
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
- South
African Medical Research Council Drug Discovery and Development Research
Unit, University of Cape Town, Cape Town 7701, South Africa
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa
| | - Richard K. Gessner
- Holistic
Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Carolina Horta Andrade
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), Faculdade de Ciências Farmaceuticas de Ribeirão
Preto, Universidade de São Paulo, Ribeirão Preto, São
Paulo 05508-070, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Instituto de Informática, Universidade Federal de Goiás, Goiânia, Goiás 74690-900, Brazil
| |
Collapse
|
24
|
Fesatidou M, Petrou A, Geronikaki A. Design, Synthesis, Biological Evaluation and Molecular Docking Studies of New Thiazolidinone Derivatives as NNRTIs and SARS-CoV-2 Main Protease Inhibitors. Chem Biodivers 2024; 21:e202401697. [PMID: 39442074 PMCID: PMC11644116 DOI: 10.1002/cbdv.202401697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024]
Abstract
HIV-1 remains a major health problem worldwide since the virus has developed drug-resistant strains, so, the need for novel agents is urgent. The protein reverse transcriptase plays fundamental role in the viruses' replication cycle. FDA approved Delavirdine bearing a sulfonamide moiety, while thiazolidinone has demonstrated significant anti-HIV activity as a core heterocycle or derivative of substituted heterocycles. In this study, thirty new thiazolidinone derivatives (series A, B and C) bearing sulfonamide group were designed, synthesized and evaluated for their HIV-1 RT inhibition activity predicted by computer program PASS taking into account the best features of available NNRTIs as well as against SARS-COV-2 main protease. Seven compounds showed good anti-HIV inhibitory activity, with two of them, C1 and C2 being better (IC50 0.18 μΜ & 0.12 μΜ respectively) than the reference drug nevirapine (IC50 0.31 μΜ). The evaluation of molecules to inhibit the main protease revealed that 6 of the synthesized compounds exhibited excellent to moderate activity with two of them (B4 and B10) having better IC50 values (0.15 & 0.19 μΜ respectively) than the reference inhibitor GC376 (IC50 0.439 μΜ). The docking studies is coincides with experimental results, showing good binding mode to both enzymes.
Collapse
Affiliation(s)
- Maria Fesatidou
- Department of Pharmaceutical ChemistrySchool of PharmacyAristotle University of ThessalonikiThessaloniki54124Greece
| | - Anthi Petrou
- Department of Pharmaceutical ChemistrySchool of PharmacyAristotle University of ThessalonikiThessaloniki54124Greece
| | - Athina Geronikaki
- Department of Pharmaceutical ChemistrySchool of PharmacyAristotle University of ThessalonikiThessaloniki54124Greece
| |
Collapse
|
25
|
Woo HS, Lee KH, Park KH, Kim DW. Flavonoids Derived from the Roots of Lespedeza bicolor Inhibit the Activity of SARS-CoV Papain-like Protease. PLANTS (BASEL, SWITZERLAND) 2024; 13:3319. [PMID: 39683112 DOI: 10.3390/plants13233319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
Despite the now infamous coronavirus disease outbreaks caused by severe acute respiratory syndrome coronavirus (SARS-CoV), this virus continues to be a threat to the global population. Although a huge research effort has targeted SARS-CoV, no report exists regarding natural small molecules targeting one of its key enzymes, papain-like protease (PLpro). In this study, nine flavonoids displaying SARS-CoV PLpro inhibitory activity were isolated from the root bark of Lespedeza bicolor. The compounds were identified as erythrabyssin II (1), lespebuergine G4 (2), 1-methoxyerythrabyssin II (3), bicolosin A (4), bicolosin B (5), bicolosin (6), xanthoangelol (7), (±)-lespeol (8), and parvisoflavanone (9). Most compounds (1-4 and 6-8) inhibited SARS-CoV PLpro activity in a dose-dependent manner, with their Kis ranging from 5.56 to 75.37 μM. The structure-activity analysis of pterocarpans (1-6) showed that activity was enhanced by C1-OCH3, but it was reduced by C8-CH3. A mechanistic analysis revealed that all inhibitors were noncompetitive. Some of the key compounds isolated in this study are pterocarpans, which are abundantly present in the Leguminosae family. Overall, a rich source of SARS-CoV papain-like protease inhibitors was identified in this study.
Collapse
Affiliation(s)
- Hyun Sim Woo
- Forest Bioresources Department, Baekdudaegan National Arboretum, Bonghwa-gun 36209, Republic of Korea
| | - Kon Ho Lee
- Department of Microbiology, School of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Ki Hun Park
- Division of Applied Life Science (BK21 Four), IALS, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Dae Wook Kim
- Forest Bioresources Department, Baekdudaegan National Arboretum, Bonghwa-gun 36209, Republic of Korea
| |
Collapse
|
26
|
Baroni S, Carletti T, Donalisio M, Arduino I, Cazzaniga I, Giorgino T, Esposito F, Porta A, Diomede L, De Luigi A, Gobbi M, Lembo D, Marcello A, Tramontano E, Milani M, Mastrangelo E. The antipsychotic drug lurasidone inhibits coronaviruses by affecting multiple targets. Front Cell Infect Microbiol 2024; 14:1487604. [PMID: 39654978 PMCID: PMC11625747 DOI: 10.3389/fcimb.2024.1487604] [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: 08/28/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Coronaviruses (CoVs) share key genomic elements critical for viral replication, suggesting the feasibility of developing therapeutics with efficacy across different viruses. In a previous work, we demonstrated the antiviral activity of the antipsychotic drug lurasidone against both SARS-CoV-2 and HCoV-OC43. In this study, our investigations on the mechanism of action of lurasidone suggested that the drug exhibits antiviral activity by targeting the papain-like protease (PL-Pro) of both viruses, and the Spike protein of SARS-CoV-2, thereby hampering both the entry and the viral replication. In vitro assays demonstrate that lurasidone significantly reduces viral load in infected cells, showing that the drug is a promising candidate for further development as a dual-action antiviral, offering a potential new strategy in the fight against COVID-19 and other coronavirus-related diseases.
Collapse
Affiliation(s)
- Sara Baroni
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - Tea Carletti
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Manuela Donalisio
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, Regione Gonzole, Turin, Italy
| | - Irene Arduino
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, Regione Gonzole, Turin, Italy
| | - Irene Cazzaniga
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Milano, Italy
| | - Toni Giorgino
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Milano, Italy
| | - Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Alessia Porta
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - Marco Gobbi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milano, Italy
| | - David Lembo
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, Regione Gonzole, Turin, Italy
| | - Alessandro Marcello
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Mario Milani
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Milano, Italy
| | - Eloise Mastrangelo
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Milano, Italy
| |
Collapse
|
27
|
Lu Y, Yang Q, Ran T, Zhang G, Li W, Zhou P, Tang J, Dai M, Zhong J, Chen H, He P, Zhou A, Xue B, Chen J, Zhang J, Yang S, Wu K, Wu X, Tang M, Zhang WK, Guo D, Chen X, Chen H, Shang J. Discovery of orally bioavailable SARS-CoV-2 papain-like protease inhibitor as a potential treatment for COVID-19. Nat Commun 2024; 15:10169. [PMID: 39580525 PMCID: PMC11585628 DOI: 10.1038/s41467-024-54462-0] [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: 05/24/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024] Open
Abstract
The RNA-dependent RNA polymerase (RdRp), 3C-like protease (3CLpro), and papain-like protease (PLpro) are pivotal components in the viral life cycle of SARS-CoV-2, presenting as promising therapeutic targets. Currently, all FDA-approved antiviral drugs against SARS-CoV-2 are RdRp or 3CLpro inhibitors. However, the mutations causing drug resistance have been observed in RdRp and 3CLpro from SARS-CoV-2, which makes it necessary to develop antivirals with novel mechanisms. Through the application of a structure-based drug design (SBDD) approach, we discover a series of novel potent non-covalent PLpro inhibitors with remarkable in vitro potency and in vivo PK properties. The co-crystal structures of PLpro with lead compounds reveal that the residues D164 and Q269 around the S2 site are critical for improving the inhibitor's potency. The lead compound GZNL-P36 not only inhibits SARS-CoV-2 and its variants at the cellular level with EC50 ranging from 58.2 nM to 306.2 nM, but also inhibits HCoV-NL63 and HCoV-229E with EC50 of 81.6 nM and 2.66 μM, respectively. Oral administration of the GZNL-P36 results in significantly improved survival and notable reductions in lung viral loads and lesions in SARS-CoV-2 infection mouse model, consistent with RNA-seq data analysis. Our results indicate that PLpro inhibitors represent a promising SARS-CoV-2 therapy.
Collapse
Affiliation(s)
- Yongzhi Lu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ting Ran
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Guihua Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Wenqi Li
- Guangzhou National Laboratory, Guangzhou, 510005, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Peiqi Zhou
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Minxian Dai
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jinpeng Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Hua Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Pan He
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Anqi Zhou
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jiayi Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jiyun Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Sidi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Kunzhong Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xinyu Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Miru Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Wei K Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Deyin Guo
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Hongming Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jinsai Shang
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
28
|
Wang Y, Wang J, Zhao Y, Liu Y, Zhang M, Deng X, Zhu J, Li G, Liu G. Isolation and Genomic Characteristics of a Novel Pathogenicity Type I Feline Coronavirus in Mainland China. Transbound Emerg Dis 2024; 2024:4162458. [PMID: 40303063 PMCID: PMC12016700 DOI: 10.1155/2024/4162458] [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: 10/10/2023] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 05/02/2025]
Abstract
Feline coronavirus (FCoV) is an enveloped, positive-sense RNA virus, which is widespread among feline populations, and can cause a fatal serious disease called feline infectious peritonitis (FIP). According to the differences of antigen and genetic composition, FCoV consists of two genotypes, FCoV I and FCoV II. In this study, we have isolated and identified a FCoV I strain named HL2019. Based on the complete genome of HL2019, phylogenetic analysis showed that HL2019 strain formed in the cluster FCoV I which is more closed to human coronavirus 229E (HCoV 229E) and HCoV NL63, while the FCoV I stains is distantly related to FCoV II strains. Analyzing with RDP4 and Simplot software showed that the virus HL2019 is recombinant by the FCoV I China/ZJU1709 and FCoV I Netherlands/UU16 strains. Furthermore, the pathogenicity of HL2019 was evaluated in 9-12-month-old cats. Two of three challenged cats developed serious clinical signs and died at 28-day postchallenge (dpc). Real-time polymerase chain reaction (PCR) analysis showed that HL2019 has broad tissue tropism, especially in the duodenum with viral load up to 104 copies/mg. In summary, our data show that we have successfully isolated a strain of FCoV I named HL2019 that is highly pathogenic to cats.
Collapse
Affiliation(s)
- Yuanhong Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Junna Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - You Zhao
- Research Center of General Administration of Customs, Beijing 100011, China
| | - Yun Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Miao Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiaoying Deng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jie Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Guoxin Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Guangqing Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| |
Collapse
|
29
|
Chen J, Zhang Y, Zhang B, Wang Z. In Vitro Characterization of Inhibition Function of Calcifediol to the Protease Activity of SARS-COV-2 PLpro. J Med Virol 2024; 96:e70085. [PMID: 39588768 DOI: 10.1002/jmv.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/14/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024]
Abstract
Vitamin D3 and its metabolites calcifediol have been recommended as effective drugs for novel coronavirus disease 2019 (COVID-19) therapy in many studies, since the outbreak of this global dramatic pandemic. In this study, we made a striking discovery that Calcifediol demonstrates robust inhibitive effect on the of the papain-like cysteine protease (PLpro), a critical proteolytic enzyme for the severe acute respiratory syndrome coronavirus-2(SARS-COV-2), through a small-scale FRET-based screening experiment. The practical bindings of Calcifediol to PLpro were also demonstrated by several in vitro interaction studies. All the evidence had revealed the inhibition might be caused by the targeted binding event. Consequently, our discovery represents a significant finding that the beneficial therapeutic impact of Calcifediol on COVID-19 may be attributed not only to its immunoregulatory properties but also to its inhibition of PLpro. This finding strongly bolsters the case for the clinical use of Vitamin D3 and its derivative Calcifediol in the treatment of COVID-19.
Collapse
Affiliation(s)
- Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yaya Zhang
- Department of Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Bingchang Zhang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Zhanxiang Wang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
30
|
Kerti L, Frecer V. Design of inhibitors of SARS-CoV-2 papain-like protease deriving from GRL0617: Structure-activity relationships. Bioorg Med Chem 2024; 113:117909. [PMID: 39288705 DOI: 10.1016/j.bmc.2024.117909] [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: 06/19/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024]
Abstract
The unique and complex structure of papain-like protease (PLpro) of the SARS-CoV-2 virus represents a difficult challenge for antiviral development, yet it offers a compelling validated target for effective therapy of COVID-19. The surge in scientific interest in inhibiting this cysteine protease emerged after its demonstrated connection to the cytokine storm in patients with COVID-19 disease. Furthermore, the development of new inhibitors against PLpro may also be beneficial for the treatment of respiratory infections caused by emerging coronavirus variants of concern. This review article provides a comprehensive overview of PLpro inhibitors, focusing on the structural framework of the known inhibitor GRL0617 and its analogs. We categorize PLpro inhibitors on the basis of their structures and binding site: Glu167 containing site, BL2 groove, Val70Ub site, and Cys111 containing catalytic site. We summarize and evaluate the majority of GRL0617-like inhibitors synthesized so far, highlighting their published biochemical parameters, which reflect their efficacy. Published research has shown that strategic modifications to GRL0617, such as decorating the naphthalene ring, extending the aromatic amino group or the orthomethyl group, can substantially decrease the IC50 from micromolar up to nanomolar concentration range. Some advantageous modifications significantly enhance inhibitory activity, paving the way for the development of new potent compounds. Our review places special emphasis on structures that involve direct modifications to the GRL0617 scaffold, including piperidine carboxamides and modified benzylmethylnaphthylethanamines (Jun9 scaffold). All these compounds are believed to inhibit the proteolytic, deubiquitination, and deISGylation activity of PLpro, biochemical processes linked to the severe progression of COVID-19. Finally, we summarize the development efforts for SARS-CoV-2 PLpro inhibitors, in detailed structure-activity relationships diagrams. This aims to inform and inspire future research in the search for potent antiviral agents against PLpro of current and emerging coronavirus threats.
Collapse
Affiliation(s)
- Lukas Kerti
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, SK-83232 Bratislava, Slovakia
| | - Vladimir Frecer
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, SK-83232 Bratislava, Slovakia.
| |
Collapse
|
31
|
Lazou M, Bekar-Cesaretli AA, Vajda S, Joseph-McCarthy D. Identification and Ranking of Binding Sites from Structural Ensembles: Application to SARS-CoV-2. Viruses 2024; 16:1647. [PMID: 39599762 PMCID: PMC11599001 DOI: 10.3390/v16111647] [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: 08/25/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Target identification and evaluation is a critical step in the drug discovery process. Although time-intensive and complex, the challenge becomes even more acute in the realm of infectious disease, where the rapid emergence of new viruses, the swift mutation of existing targets, and partial effectiveness of approved antivirals can lead to outbreaks of significant public health concern. The COVID-19 pandemic, caused by the SARS-CoV-2 virus, serves as a prime example of this, where despite the allocation of substantial resources, Paxlovid is currently the only effective treatment. In that case, significant effort pre-pandemic had been expended to evaluate the biological target for the closely related SARS-CoV. In this work, we utilize the computational hot spot mapping method, FTMove, to rapidly identify and rank binding sites for a set of nine SARS-CoV-2 drug/potential drug targets. FTMove takes into account protein flexibility by mapping binding site hot spots across an ensemble of structures for a given target. To assess the applicability of the FTMove approach to a wide range of drug targets for viral pathogens, we also carry out a comprehensive review of the known SARS-CoV-2 ligandable sites. The approach is able to identify the vast majority of all known sites and a few additional sites, which may in fact be yet to be discovered as ligandable. Furthermore, a UMAP analysis of the FTMove features for each identified binding site is largely able to separate predicted sites with experimentally known binders from those without known binders. These results demonstrate the utility of FTMove to rapidly identify actionable sites across a range of targets for a given indication. As such, the approach is expected to be particularly useful for assessing target binding sites for any emerging pathogen, as well as for indications in other disease areas, and providing actionable starting points for structure-based drug design efforts.
Collapse
Affiliation(s)
- Maria Lazou
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; (M.L.); (S.V.)
| | | | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; (M.L.); (S.V.)
- Department of Chemistry, Boston University, Boston, MA 02215, USA;
| | - Diane Joseph-McCarthy
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; (M.L.); (S.V.)
- Department of Chemistry, Boston University, Boston, MA 02215, USA;
| |
Collapse
|
32
|
James VK, Godula RN, Perez JM, Beckham JT, Butalewicz JP, Sipe SN, Huibregtse JM, Brodbelt JS. Native Mass Spectrometry Reveals Binding Interactions of SARS-CoV-2 PLpro with Inhibitors and Cellular Targets. ACS Infect Dis 2024; 10:3597-3606. [PMID: 39303064 PMCID: PMC11533220 DOI: 10.1021/acsinfecdis.4c00444] [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] [Indexed: 09/22/2024]
Abstract
Here we used native mass spectrometry (native MS) to probe a SARS-CoV protease, PLpro, which plays critical roles in coronavirus disease by affecting viral protein production and antagonizing host antiviral responses. Ultraviolet photodissociation (UVPD) and variable temperature electrospray ionization (vT ESI) were used to localize binding sites of PLpro inhibitors and revealed the stabilizing effects of inhibitors on protein tertiary structure. We compared PLpro from SARS-CoV-1 and SARS-CoV-2 in terms of inhibitor and ISG15 interactions to discern possible differences in protease function. A PLpro mutant lacking a single cysteine was used to localize inhibitor binding, and thermodynamic measurements revealed that inhibitor PR-619 stabilized the folded PLpro structure. These results will inform further development of PLpro as a therapeutic target against SARS-CoV-2 and other emerging coronaviruses.
Collapse
Affiliation(s)
- Virginia K. James
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Rianna N. Godula
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jessica M. Perez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Josh T. Beckham
- Freshman Research Initiative, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jamie P. Butalewicz
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Sarah N. Sipe
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jon M. Huibregtse
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer S. Brodbelt
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
33
|
Patel DK, Kumar H, Sobhia ME. Exploring the binding dynamics of covalent inhibitors within active site of PL pro in SARS-CoV-2. Comput Biol Chem 2024; 112:108132. [PMID: 38959551 DOI: 10.1016/j.compbiolchem.2024.108132] [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: 02/27/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
In the global fight against the COVID-19 pandemic caused by the highly transmissible SARS-CoV-2 virus, the search for potent medications is paramount. With a focused investigation on the SARS-CoV-2 papain-like protease (PLpro) as a promising therapeutic target due to its pivotal role in viral replication and immune modulation, the catalytic triad of PLpro comprising Cys111, His272, and Asp286, highlights Cys111 as an intriguing nucleophilic center for potential covalent bonds with ligands. The detailed analysis of the binding site unveils crucial interactions with both hydrophobic and polar residues, demonstrating the structural insights of the cavity and deepening our understanding of its molecular landscape. The sequence of PLpro among variants of concern (Alpha, Beta, Gamma, Delta and Omicron) and the recent variant of interest, JN.1, remains conserved with no mutations at the active site. Moreover, a thorough exploration of apo, non-covalently bound, and covalently bound PLpro conformations exposes significant conformational changes in loop regions, offering invaluable insights into the intricate dynamics of ligand-protein complex formation. Employing strategic in silico medication repurposing, this study swiftly identifies potential molecules for target inhibition. Within the domain of covalent docking studies and molecular dynamics, using reported inhibitors and clinically tested molecules elucidate the formation of stable covalent bonds with the cysteine residue, laying a robust foundation for potential therapeutic applications. These details not only deepen our comprehension of PLpro inhibition but also play a pivotal role in shaping the dynamic landscape of COVID-19 treatment strategies.
Collapse
Affiliation(s)
- Deepesh Kumar Patel
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Harish Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India.
| |
Collapse
|
34
|
Ferreira JC, Villanueva AJ, Al Adem K, Fadl S, Alzyoud L, Ghattas MA, Rabeh WM. Identification of novel allosteric sites of SARS-CoV-2 papain-like protease (PLpro) for the development of COVID-19 antivirals. J Biol Chem 2024; 300:107821. [PMID: 39342997 PMCID: PMC11538808 DOI: 10.1016/j.jbc.2024.107821] [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: 06/06/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024] Open
Abstract
Coronaviruses such as SARS-CoV-2 encode a conserved papain-like protease (PLpro) that is crucial for viral replication and immune evasion, making it a prime target for antiviral drug development. In this study, three surface pockets on SARS-CoV-2 PLpro that may function as sites for allosteric inhibition were computationally identified. To evaluate the effects of these pockets on proteolytic activity, 52 residues were separately mutated to alanine. In Pocket 1, located between the Ubl and thumb domains, the introduction of alanine at T10, D12, T54, Y72, or Y83 reduced PLpro activity to <12% of that of WT. In Pocket 2, situated at the interface of the thumb, fingers, and palm domains, Q237A, S239A, H275A, and S278A inactivated PLpro. Finally, introducing alanine at five residues in Pocket 3, between the fingers and palm domains, inactivated PLpro: S212, Y213, Y251, K254, and Y305. Pocket 1 has a higher druggability score than Pockets 2 and 3. MD simulations showed that interactions within and between domains play critical roles in PLpro activity and thermal stability. The essential residues in Pockets 1 and 2 participate in a combination of intra- and inter-domain interactions. By contrast, the essential residues in Pocket three predominantly participate in inter-domain interactions. The most promising targets for therapeutic development are Pockets one and 3, which have the highest druggability score and the largest number of essential residues, respectively. Non-competitive inhibitors targeting these pockets may be antiviral agents against COVID-19 and related coronaviruses.
Collapse
Affiliation(s)
- Juliana C Ferreira
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Adrian J Villanueva
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Kenana Al Adem
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Samar Fadl
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Lara Alzyoud
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Mohammad A Ghattas
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Wael M Rabeh
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
35
|
Diogo MA, Cabral AGT, de Oliveira RB. Advances in the Search for SARS-CoV-2 M pro and PL pro Inhibitors. Pathogens 2024; 13:825. [PMID: 39452697 PMCID: PMC11510351 DOI: 10.3390/pathogens13100825] [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: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 10/26/2024] Open
Abstract
SARS-CoV-2 is a spherical, positive-sense, single-stranded RNA virus with a large genome, responsible for encoding both structural proteins, vital for the viral particle's architecture, and non-structural proteins, critical for the virus's replication cycle. Among the non-structural proteins, two cysteine proteases emerge as promising molecular targets for the design of new antiviral compounds. The main protease (Mpro) is a homodimeric enzyme that plays a pivotal role in the formation of the viral replication-transcription complex, associated with the papain-like protease (PLpro), a cysteine protease that modulates host immune signaling by reversing post-translational modifications of ubiquitin and interferon-stimulated gene 15 (ISG15) in host cells. Due to the importance of these molecular targets for the design and development of novel anti-SARS-CoV-2 drugs, the purpose of this review is to address aspects related to the structure, mechanism of action and strategies for the design of inhibitors capable of targeting the Mpro and PLpro. Examples of covalent and non-covalent inhibitors that are currently being evaluated in preclinical and clinical studies or already approved for therapy will be also discussed to show the advances in medicinal chemistry in the search for new molecules to treat COVID-19.
Collapse
Affiliation(s)
| | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.D.); (A.G.T.C.)
| |
Collapse
|
36
|
Cui W, Duan Y, Gao Y, Wang W, Yang H. Structural review of SARS-CoV-2 antiviral targets. Structure 2024; 32:1301-1321. [PMID: 39241763 DOI: 10.1016/j.str.2024.08.005] [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: 05/13/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 09/09/2024]
Abstract
The coronavirus disease 2019 (COVID-19), the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents the most disastrous infectious disease pandemic of the past century. As a member of the Betacoronavirus genus, the SARS-CoV-2 genome encodes a total of 29 proteins. The spike protein, RNA-dependent RNA polymerase, and proteases play crucial roles in the virus replication process and are promising targets for drug development. In recent years, structural studies of these viral proteins and of their complexes with antibodies and inhibitors have provided valuable insights into their functions and laid a solid foundation for drug development. In this review, we summarize the structural features of these proteins and discuss recent progress in research regarding therapeutic development, highlighting mechanistically representative molecules and those that have already been approved or are under clinical investigation.
Collapse
Affiliation(s)
- Wen Cui
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Yinkai Duan
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yan Gao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201203, China
| | - Wei Wang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201203, China.
| |
Collapse
|
37
|
Liang JJ, Pitsillou E, Lau HLY, Mccubbery CP, Gan H, Hung A, Karagiannis TC. Utilization of the EpiMed Coronabank Chemical Collection to identify potential SARS-CoV-2 antivirals: in silico studies targeting the nsp14 ExoN domain and PL pro naphthalene binding site. J Mol Graph Model 2024; 131:108803. [PMID: 38815531 DOI: 10.1016/j.jmgm.2024.108803] [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: 12/08/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome encodes 29 proteins including four structural, 16 nonstructural (nsps), and nine accessory proteins (https://epimedlab.org/sars-cov-2-proteome/). Many of these proteins contain potential targetable sites for the development of antivirals. Despite the widespread use of vaccinations, the emergence of variants necessitates the investigation of new therapeutics and antivirals. Here, the EpiMed Coronabank Chemical Collection (https://epimedlab.org/crl/) was utilized to investigate potential antivirals against the nsp14 exoribonuclease (ExoN) domain. Molecular docking was performed to evaluate the binding characteristics of our chemical library against the nsp14 ExoN site. Based on the initial screen, trisjuglone, ararobinol, corilagin, and naphthofluorescein were identified as potential lead compounds. Molecular dynamics (MD) simulations were subsequently performed, with the results highlighting the stability of the lead compounds in the nsp14 ExoN site. Protein-RNA docking revealed the potential for the lead compounds to disrupt the interaction with RNA when bound to the ExoN site. Moreover, hypericin, cyanidin-3-O-glucoside, and rutin were previously identified as lead compounds targeting the papain-like protease (PLpro) naphthalene binding site. Through performing MD simulations, the stability and interactions of lead compounds with PLpro were further examined. Overall, given the critical role of the exonuclease activity of nsp14 in ensuring viral fidelity and the multifunctional role of PLpro in viral pathobiology and replication, these nsps represent important targets for antiviral drug development. Our databases can be utilized for in silico studies, such as the ones performed here, and this approach can be applied to other potentially druggable SARS-CoV-2 protein targets.
Collapse
Affiliation(s)
- Julia J Liang
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia; Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia
| | - Hannah L Y Lau
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cian P Mccubbery
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Hockxuen Gan
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia
| | - Tom C Karagiannis
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC, 3053, Australia; Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
38
|
Rahebi P, Aryapour H. Reconstruction of the unbinding pathways of new inhibitors of the SARS-CoV-2 papain-like protease using molecular dynamics simulation. J Biomol Struct Dyn 2024; 42:7501-7514. [PMID: 37505097 DOI: 10.1080/07391102.2023.2240424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Developing novel antiviral drugs against the SARS-CoV-2 virus and COVID-19 disease is imperative as the vaccines may not offer absolute protection. PLpro plays a crucial role in the viral life cycle, making it an attractive target for drug development. Several PLpro inhibitors have been developed, and their 3D structures in complex with PLpro are available. In this work, we employed Supervised Molecular Dynamics (SuMD), a specific Unbiased Molecular Dynamics (UMD) method, to investigate unbinding pathways of the novel inhibitors of PLpro (PDB IDs: 7LBR, 7RZC, 7SDR and 7E35) and GRL0617 (PDB ID: 7JRN) as a reference. We conducted three simulations for each ligand and achieved unbinding events in the nanosecond timescale in all simulations. We found that unbinding events are commonly affected by altering the conformation of the BL2 loop, which is caused by the natural fluctuations of the loop that are required to trap the substrate and throw out the product. BL2 loop is crucial for keeping the ligand and unbinding and acts as a double-edged sword. Any inhibitor designed to be effective must prevent the loop's natural fluctuations. We perceived that increasing ligands interactions with the binding pocket interior and the BL2 loop will help prevent natural fluctuation of the BL2 loop, Although the interactions with the binding pocket's inner side are more critical than the BL2 loop. These findings may be helpful in developing more potent inhibitors against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pouya Rahebi
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| | - Hassan Aryapour
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| |
Collapse
|
39
|
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
|
40
|
Ciardullo G, Orlando C, Russo N, Marchese E, Galano A, Marino T, Prejanò M. On the dual role of (+)-catechin as primary antioxidant and inhibitor of viral proteases. Comput Biol Med 2024; 180:108953. [PMID: 39089115 DOI: 10.1016/j.compbiomed.2024.108953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/03/2024]
Abstract
Natural antioxidants have become the subject of many investigations due to the role that they play in the reduction of oxidative stress. Their main scavenging mechanisms concern the direct inactivation of free radicals and the coordination of metal ions involved in Fenton-like reactions. Recently, increasing attention has been paid to non-covalent inhibition of enzymes involved in different diseases by the antioxidants. Here, a computational investigation on the primary antioxidant power of (+)-catechin against the •OOH radical has been performed in both lipid-like and aqueous environments, taking into account the relevant species present in the simulated acid-base equilibria at the physiological pH. Hydrogen Atom Transfer (HAT), Single Electron Transfer (SET), and Radical Adduct Formation (RAF) mechanisms were studied, and relative rate constants were estimated. The potential inhibitory activity of the (+)-catechin towards the most important proteases from SARS-CoV-2, 3C-like (Mpro) and papain-like (PLpro) proteases was also investigated by MD simulations to provide deeper atomistic insights on the binding sites. Based on the antioxidant and antiviral properties also unravelled by comparison with other molecules having similar chemical scaffold, our results propose that (+)-CTc satisfies can explicate a dual action as antioxidant and antiviral in particular versus Mpro from SARS-CoV-2.
Collapse
Affiliation(s)
- Giada Ciardullo
- Dipartimento di Chimica e Tecnologie Chimiche, Università Della Calabria, Rende, (CS), Italy
| | - Carla Orlando
- Dipartimento di Chimica e Tecnologie Chimiche, Università Della Calabria, Rende, (CS), Italy
| | - Nino Russo
- Dipartimento di Chimica e Tecnologie Chimiche, Università Della Calabria, Rende, (CS), Italy
| | - Emanuela Marchese
- Dipartimento di Chimica e Tecnologie Chimiche, Università Della Calabria, Rende, (CS), Italy
| | - Annia Galano
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, 09310, Mexico
| | - Tiziana Marino
- Dipartimento di Chimica e Tecnologie Chimiche, Università Della Calabria, Rende, (CS), Italy.
| | - Mario Prejanò
- Dipartimento di Chimica e Tecnologie Chimiche, Università Della Calabria, Rende, (CS), Italy
| |
Collapse
|
41
|
Zeppilli D, Madabeni A, Nogara PA, Rocha JBT, Orian L. Reactivity of Zinc Fingers in Oxidizing Environments: Insight from Molecular Models Through Activation Strain Analysis. Chempluschem 2024; 89:e202400252. [PMID: 38842473 DOI: 10.1002/cplu.202400252] [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: 04/05/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/07/2024]
Abstract
The reactivity of Zn2+ tetrahedral complexes with H2O2 was investigated in silico, as a first step in their disruption process. The substrates were chosen to represent the cores of three different zinc finger protein motifs, i. e., a Zn2+ ion coordinated to four cysteines (CCCC), to three cysteines and one histidine (CCCH), and to two cysteines and two histidines (CCHH). The cysteine and histidine ligands were further simplified to methyl thiolate and imidazole, respectively. H2O2 was chosen as an oxidizing agent due to its biological role as a metabolic product and species involved in signaling processes. The mechanism of oxidation of a coordinated cysteinate to sulfenate-κS and the trends for the different substrates were rationalized through activation strain analysis and energy decomposition analysis in the framework of scalar relativistic Density Functional Theory (DFT) calculations at ZORA-M06/TZ2P ae // ZORA-BLYP-D3(BJ)/TZ2P. CCCC is oxidized most easily, an outcome explained considering both electrostatic and orbital interactions. The isomerization to sulfenate-κO was attempted to assess whether this step may affect the ligand dissociation; however, it was found to introduce a kinetic barrier without improving the energetics of the dissociation. Lastly, ligand exchange with free thiolates and selenolates was investigated as a trigger for ligand dissociation, possibly leading to metal ejection; molecular docking simulations also support this hypothesis.
Collapse
Affiliation(s)
- Davide Zeppilli
- Dipartimento di Scienze Chimiche Università degli Studi di Padova, Via Marzolo 1, 35131, Padova, Italy
| | - Andrea Madabeni
- Dipartimento di Scienze Chimiche Università degli Studi di Padova, Via Marzolo 1, 35131, Padova, Italy
| | - Pablo A Nogara
- Departamento de Bioquímica e Biologia Molecolar, Universidade Federal de Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia Sul-rio-grandense (IFSul), Av. Leonel de Moura Brizola, 2501, 96418-400, Bagé, RS, Brasil
| | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecolar, Universidade Federal de Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil
| | - Laura Orian
- Dipartimento di Scienze Chimiche Università degli Studi di Padova, Via Marzolo 1, 35131, Padova, Italy
| |
Collapse
|
42
|
Velma G, Shen Z, Holberg C, Fu J, Soleymani F, Cooper L, Ramos OL, Indukuri D, Musku SR, Rychetsky P, Slilaty S, Li Z, Ratia K, Rong L, Schenten D, Xiong R, J Thatcher GR. Non-Covalent Inhibitors of SARS-CoV-2 Papain-Like Protease (PLpro): In Vitro and In Vivo Antiviral Activity. J Med Chem 2024; 67:13681-13702. [PMID: 39102360 PMCID: PMC11345844 DOI: 10.1021/acs.jmedchem.4c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
The SARS-CoV-2 papain-like protease (PLpro), essential for viral processing and immune response disruption, is a promising target for treating acute infection of SARS-CoV-2. To date, there have been no reports of PLpro inhibitors with both submicromolar potency and animal model efficacy. To address the challenge of PLpro's featureless active site, a noncovalent inhibitor library with over 50 new analogs was developed, targeting the PLpro active site by modulating the BL2-loop and engaging the BL2-groove. Notably, compounds 42 and 10 exhibited strong antiviral effects and were further analyzed pharmacokinetically. 10, in particular, showed a significant lung accumulation, up to 12.9-fold greater than plasma exposure, and was effective in a mouse model of SARS-CoV-2 infection, as well as against several SARS-CoV-2 variants. These findings highlight the potential of 10 as an in vivo chemical probe for studying PLpro inhibition in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ganga
Reddy Velma
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Zhengnan Shen
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Cameron Holberg
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Jiqiang Fu
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Farinaz Soleymani
- Department
of Chemistry & Biochemistry, Colleges of Science and Medicine, University of Arizona, Tucson 85721, Arizona, United States
| | - Laura Cooper
- Department
of Microbiology, College of Medicine, University
of Illinois at Chicago (UIC), Chicago 60612, Illinois, United States
| | - Omar Lozano Ramos
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Divakar Indukuri
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Soumya Reddy Musku
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Pavel Rychetsky
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Steve Slilaty
- Sunshine
Biopharma Inc, 333 Las Olas Way, CU4 Suite 433, Fort Lauderdale 33301, Florida, United States
| | - Zuomei Li
- Sunshine
Biopharma Inc, 333 Las Olas Way, CU4 Suite 433, Fort Lauderdale 33301, Florida, United States
| | - Kiira Ratia
- Research
Resources Center, University of Illinois
at Chicago (UIC), Chicago 60612, Illinois, United States
| | - Lijun Rong
- Department
of Microbiology, College of Medicine, University
of Illinois at Chicago (UIC), Chicago 60612, Illinois, United States
| | - Dominik Schenten
- Department
of Immunology, College of Medicine, University
of Arizona, Tucson 85721, Arizona, United States
| | - Rui Xiong
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
| | - Gregory R. J Thatcher
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson 85721, Arizona, United States
- Department
of Chemistry & Biochemistry, Colleges of Science and Medicine, University of Arizona, Tucson 85721, Arizona, United States
| |
Collapse
|
43
|
Gaiya DD, Muhammad A, Musa JS, Auta R, Dadah AJ, Bello RO, Hassan M, Eke SS, Odihi RI, Sankey M. In silico analysis of balsaminol as anti-viral agents targeting SARS-CoV-2 main protease, spike receptor binding domain and papain-like protease receptors. In Silico Pharmacol 2024; 12:75. [PMID: 39155972 PMCID: PMC11329488 DOI: 10.1007/s40203-024-00241-0] [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: 10/09/2023] [Accepted: 07/13/2024] [Indexed: 08/20/2024] Open
Abstract
Plant-derived phytochemicals from medicinal plants are becoming increasingly attractive natural sources of antimicrobial and antiviral agents due to their therapeutic value, mechanism of action, level of toxicity and bioavailability. The continued emergence of more immune-evasive strains and the rate of resistance to current antiviral drugs have created a need to identify new antiviral agents against SARS-CoV-2. This study investigated the antiviral potential of balsaminol, a bioactive compound from Momordica balsamina, and its inhibitory activities against SARS-CoV-2 receptor proteins. In this study, three Food and Drug Administration (FDA) COVID-19 approved drugs namely; nirmatrelvir, ritonavir and remdesivir were used as positive control. Molecular docking was performed to determine the predominant binding mode (most negative Gibbs free energy of binding/ΔG) and inhibitory activity of balsaminol against SARS-CoV-2 receptor proteins. The pharmacokinetics, toxicity, physicochemical and drug-like properties of balsaminol were evaluated to determine its potential as an active oral drug candidate as well as its non-toxicity in humans. The results show that balsaminol E has the highest binding affinity to the SARS CoV-2 papain-like protease (7CMD) with a free binding energy of - 8.7 kcal/mol, followed by balsaminol A interacting with the spike receptor binding domain (6VW1) with - 8.5 kcal/mol and balsaminol C had a binding energy of - 8.1 kcal/mol with the main protease (6LU7) comparable to the standard drugs namely ritonavir, nirmatrelvir and remdesivir. However, the ADMET and drug-like profile of balsaminol F favours it as a better potential drug candidate and inhibitor of the docked SARS-CoV-2 receptor proteins. Further preclinical studies are therefore recommended. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00241-0.
Collapse
Affiliation(s)
- Daniel Danladi Gaiya
- Biology Unit, Air Force Institute of Technology, Nigerian Air Force Base, P.M.B 2104, Kaduna, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1045, Samaru Zaria, Nigeria
| | - Joy Sim Musa
- Department of Chemistry, Faculty of Physical Sciences, Ahmadu Bello University, P.M.B. 1045, Samaru Zaria, Nigeria
| | - Richard Auta
- Department of Biochemistry, Faculty of Life Sciences, Kaduna State University, Tafawa Balewa Way, P.M.B. 2339, Kaduna, Nigeria
| | - Anthony John Dadah
- Department of Microbiology, Faculty of Life Sciences, Kaduna State University, Tafawa Balewa Way, P.M.B. 2339, Kaduna, Nigeria
| | | | - Madinat Hassan
- Biology Unit, Air Force Institute of Technology, Nigerian Air Force Base, P.M.B 2104, Kaduna, Nigeria
| | - Samuel Sunday Eke
- Biology Unit, Air Force Institute of Technology, Nigerian Air Force Base, P.M.B 2104, Kaduna, Nigeria
| | - Rebecca Imoo Odihi
- Department of Biological Science, Nigerian Defence Academy, Kaduna, Nigeria
| | - Musa Sankey
- Department of Chemistry, Kaduna State College of Education, Gidan Waya, Kaduna, Nigeria
| |
Collapse
|
44
|
Ulzurrun E, Grande-Pérez A, del Hoyo D, Guevara C, Gil C, Sorzano CO, Campillo NE. Unlocking the puzzle: non-defining mutations in SARS-CoV-2 proteome may affect vaccine effectiveness. Front Public Health 2024; 12:1386596. [PMID: 39228849 PMCID: PMC11369981 DOI: 10.3389/fpubh.2024.1386596] [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: 02/15/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
Introduction SARS-CoV-2 variants are defined by specific genome-wide mutations compared to the Wuhan genome. However, non-clade-defining mutations may also impact protein structure and function, potentially leading to reduced vaccine effectiveness. Our objective is to identify mutations across the entire viral genome rather than focus on individual mutations that may be associated with vaccine failure and to examine the physicochemical properties of the resulting amino acid changes. Materials and methods Whole-genome consensus sequences of SARS-CoV-2 from COVID-19 patients were retrieved from the GISAID database. Analysis focused on Dataset_1 (7,154 genomes from Italy) and Dataset_2 (8,819 sequences from Spain). Bioinformatic tools identified amino acid changes resulting from codon mutations with frequencies of 10% or higher, and sequences were organized into sets based on identical amino acid combinations. Results Non-defining mutations in SARS-CoV-2 genomes belonging to clades 21 L (Omicron), 22B/22E (Omicron), 22F/23A (Omicron) and 21J (Delta) were associated with vaccine failure. Four sets of sequences from Dataset_1 were significantly linked to low vaccine coverage: one from clade 21L with mutations L3201F (ORF1a), A27- (S) and G30- (N); two sets shared by clades 22B and 22E with changes A27- (S), I68- (S), R346T (S) and G30- (N); and one set shared by clades 22F and 23A containing changes A27- (S), F486P (S) and G30- (N). Booster doses showed a slight improvement in protection against Omicron clades. Regarding 21J (Delta) two sets of sequences from Dataset_2 exhibited the combination of non-clade mutations P2046L (ORF1a), P2287S (ORF1a), L829I (ORF1b), T95I (S), Y145H (S), R158- (S) and Q9L (N), that was associated with vaccine failure. Discussion Vaccine coverage associations appear to be influenced by the mutations harbored by marketed vaccines. An analysis of the physicochemical properties of amino acid revealed that primarily hydrophobic and polar amino acid substitutions occurred. Our results suggest that non-defining mutations across the proteome of SARS-CoV-2 variants could affect the extent of protection of the COVID-19 vaccine. In addition, alteration of the physicochemical characteristics of viral amino acids could potentially disrupt protein structure or function or both.
Collapse
Affiliation(s)
- Eugenia Ulzurrun
- Center for Biological Research Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
- National Center for Biotechnology, Spanish National Research Council (CSIC), Madrid, Spain
- Institute of Mathematical Sciences, Spanish National Research Council (CSIC), Madrid, Spain
| | - Ana Grande-Pérez
- Department of Cellular Biology, Genetics, and Physiology, University of Malaga, Málaga, Spain
| | - Daniel del Hoyo
- National Center for Biotechnology, Spanish National Research Council (CSIC), Madrid, Spain
| | - Cesar Guevara
- Mechatronics and Interactive Systems - MIST Research Center, Universidad Tecnológica Indoamérica, Quito, Ecuador
| | - Carmen Gil
- Center for Biological Research Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
| | - Carlos Oscar Sorzano
- National Center for Biotechnology, Spanish National Research Council (CSIC), Madrid, Spain
| | - Nuria E. Campillo
- Center for Biological Research Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
45
|
Liu X, Zheng M, Zhang H, Feng B, Li J, Zhang Y, Zhang J, Zhao N, Li C, Song N, Song B, Yang D, Chen J, Qi A, Zhao L, Luo C, Zang Y, Liu H, Li J, Zhang B, Zhou Y, Zheng J. Characterization and noncovalent inhibition of the K63-deubiquitinase activity of SARS-cov-2 PLpro. Antiviral Res 2024; 228:105944. [PMID: 38914283 DOI: 10.1016/j.antiviral.2024.105944] [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: 01/05/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/26/2024]
Abstract
SARS-CoV-2 papain-like protease (PLpro) could facilitate viral replication and host immune evasion by respectively hydrolyzing viral polyprotein and host ubiquitin conjugates, thereby rendering itself as an important antiviral target. Yet few noncovalent PLpro inhibitors of SARS-CoV-2 have been reported with improved directed towards pathogenic deubiquitinating activities inhibition. Herein, we report that coronavirus PLpro proteases have distinctive substrate bias and are conserved to deubiquitylate K63-linked polyubiquitination, thereby attenuating host type I interferon response. We identify a noncovalent compound specifically optimized towards halting the K63-deubiquitinase activity of SARS-CoV-2 PLpro, but not other coronavirus (CoV) counterparts or host deubiquitinase. Contrasting with GRL-0617, a SARS-CoV-1 PLpro inhibitor, SIMM-036 is 50-fold and 7-fold (half maximal inhibitory concentration (IC50)) more potent to inhibit viral replication during SARS-CoV-2 infection and restore the host interferon-β (IFN-β) response in human angiotensin-converting enzyme 2 (hACE2)-HeLa cells, respectively. Structure-activity relationship (SAR) analysis further reveals the importance of BL2 groove of PLpro, which could determine the selectivity of K63-deubiquitinase activity of the enzyme.
Collapse
Affiliation(s)
- Xin Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Miao Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hongqing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Bo Feng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiaqi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yanan Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Ji Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Na Zhao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chaoqiang Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ning Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bin Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dongyuan Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jin Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ao Qi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Linxiang Zhao
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Cheng Luo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi Zang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hong Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jia Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yu Zhou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
46
|
Samrat SK, Kumar P, Liu Y, Chen K, Lee H, Li Z, Chen Y, Li H. An ISG15-Based High-Throughput Screening Assay for Identification and Characterization of SARS-CoV-2 Inhibitors Targeting Papain-like Protease. Viruses 2024; 16:1239. [PMID: 39205213 PMCID: PMC11359932 DOI: 10.3390/v16081239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Emergence of newer variants of SARS-CoV-2 underscores the need for effective antivirals to complement the vaccination program in managing COVID-19. The multi-functional papain-like protease (PLpro) of SARS-CoV-2 is an essential viral protein that not only regulates the viral replication but also modulates the host immune system, making it a promising therapeutic target. To this end, we developed an in vitro interferon stimulating gene 15 (ISG15)-based Förster resonance energy transfer (FRET) assay and screened the National Cancer Institute (NCI) Diversity Set VI compound library, which comprises 1584 small molecules. Subsequently, we assessed the PLpro enzymatic activity in the presence of screened molecules. We identified three potential PLpro inhibitors, namely, NSC338106, 651084, and 679525, with IC50 values in the range from 3.3 to 6.0 µM. These molecules demonstrated in vitro inhibition of the enzyme activity and exhibited antiviral activity against SARS-CoV-2, with EC50 values ranging from 0.4 to 4.6 µM. The molecular docking of all three small molecules to PLpro suggested their specificity towards the enzyme's active site. Overall, our study contributes promising prospects for further developing potential antivirals to combat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Prashant Kumar
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Yuchen Liu
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Ke Chen
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Hyun Lee
- Department of Pharmaceutical Sciences, College of Pharmacy and Biophysics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Zhong Li
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Yin Chen
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Hongmin Li
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
- Department of Chemistry and Biochemistry, College of Science & College of Medicine, The University of Arizona, Tucson, AZ 85721, USA
- The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
47
|
Ibrahim PEGF, Zuccotto F, Zachariae U, Gilbert I, Bodkin M. Accurate prediction of dynamic protein-ligand binding using P-score ranking. J Comput Chem 2024; 45:1762-1778. [PMID: 38647338 PMCID: PMC11980828 DOI: 10.1002/jcc.27370] [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: 10/20/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
Protein-ligand binding prediction typically relies on docking methodologies and associated scoring functions to propose the binding mode of a ligand in a biological target. Significant challenges are associated with this approach, including the flexibility of the protein-ligand system, solvent-mediated interactions, and associated entropy changes. In addition, scoring functions are only weakly accurate due to the short time required for calculating enthalpic and entropic binding interactions. The workflow described here attempts to address these limitations by combining supervised molecular dynamics with dynamical averaging quantum mechanics fragment molecular orbital. This combination significantly increased the ability to predict the experimental binding structure of protein-ligand complexes independent from the starting position of the ligands or the binding site conformation. We found that the predictive power could be enhanced by combining the residence time and interaction energies as descriptors in a novel scoring function named the P-score. This is illustrated using six different protein-ligand targets as case studies.
Collapse
Affiliation(s)
- Peter E. G. F. Ibrahim
- Drug Discovery Unit, Division of Biological Chemistry and Drug DiscoveryUniversity of DundeeDundeeUK
| | - Fabio Zuccotto
- Drug Discovery Unit, Division of Biological Chemistry and Drug DiscoveryUniversity of DundeeDundeeUK
| | - Ulrich Zachariae
- Drug Discovery Unit, Division of Biological Chemistry and Drug DiscoveryUniversity of DundeeDundeeUK
| | - Ian Gilbert
- Drug Discovery Unit, Division of Biological Chemistry and Drug DiscoveryUniversity of DundeeDundeeUK
| | - Mike Bodkin
- Drug Discovery Unit, Division of Biological Chemistry and Drug DiscoveryUniversity of DundeeDundeeUK
| |
Collapse
|
48
|
Hangyu W, Panpan L, Jie S, Hongyan W, Linmiao W, Kangning H, Yichen S, Shuai W, Cheng W. Advancements in Antiviral Drug Development: Comprehensive Insights into Design Strategies and Mechanisms Targeting Key Viral Proteins. J Microbiol Biotechnol 2024; 34:1376-1384. [PMID: 38934770 PMCID: PMC11294656 DOI: 10.4014/jmb.2403.03008] [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: 03/07/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 06/28/2024]
Abstract
Viral infectious diseases have always been a threat to human survival and quality of life, impeding the stability and progress of human society. As such, researchers have persistently focused on developing highly efficient, low-toxicity antiviral drugs, whether for acute or chronic infectious diseases. This article presents a comprehensive review of the design concepts behind virus-targeted drugs, examined through the lens of antiviral drug mechanisms. The intention is to provide a reference for the development of new, virus-targeted antiviral drugs and guide their clinical usage.
Collapse
Affiliation(s)
- Wang Hangyu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Li Panpan
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Shen Jie
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Wang Hongyan
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Wei Linmiao
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Han Kangning
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Shi Yichen
- School of Stomatology, Shandong Second Medical University, Weifang 261053, P.R. China
| | - Wang Shuai
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia 010050, P.R. China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, Inner Mongolia 010110, P.R. China
| | - Wang Cheng
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia 010050, P.R. China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, Inner Mongolia 010110, P.R. China
| |
Collapse
|
49
|
Zhang W, Xiao L, Li D, Hu Y, Yu W. New Strategies for Responding to SARS-CoV-2: The Present and Future of Dual-Target Drugs. J Med Chem 2024; 67:11522-11542. [PMID: 38967785 DOI: 10.1021/acs.jmedchem.4c00384] [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: 07/06/2024]
Abstract
The 2019 coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in millions of deaths, posing a serious threat to public health and safety. Rapid mutations of SARS-CoV-2 and complex interactions among multiple targets during infection pose a risk of expiry for small molecule inhibitors. This suggests that the traditional concept of "one bug, one drug" could be ineffective in dealing with the coronavirus. The dual-target drug strategy is expected to be the key to ending coronavirus infections. However, the lack of design method and improper combination of dual-targets poses obstacle to the discovery of new dual-target drugs. In this Perspective, we summarized the profiles concerning drug design methods, structure-activity relationships, and pharmacological parameters of dual-target drugs for the treatment of COVID-19. Importantly, we underscored how target combination and rational drug design illuminate the development of dual-target drugs for SARS-CoV-2.
Collapse
Affiliation(s)
- Wenyi Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lecheng Xiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Dianyang Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuxuan Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Wenying Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
50
|
Yaghi R, Andrews CL, Wylie DC, Iverson BL. High-Resolution Substrate Specificity Profiling of SARS-CoV-2 M pro; Comparison to SARS-CoV M pro. ACS Chem Biol 2024; 19:1474-1483. [PMID: 38865301 PMCID: PMC11267570 DOI: 10.1021/acschembio.4c00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/14/2024]
Abstract
The SARS-CoV-2 Mpro protease from COVID-19 cleaves the pp1a and pp2b polyproteins at 11 sites during viral maturation and is the target of Nirmatrelvir, one of the two components of the frontline treatment sold as Paxlovid. We used the YESS 2.0 platform, combining protease and substrate expression in the yeast endoplasmic reticulum with fluorescence-activated cell sorting and next-generation sequencing, to carry out the high-resolution substrate specificity profiling of SARS-CoV-2 Mpro as well as the related SARS-CoV Mpro from SARS 2003. Even at such a high level of resolution, the substrate specificity profiles of both enzymes are essentially identical. The population of cleaved substrates isolated in our sorts is so deep, the relative catalytic efficiencies of the different cleavage sites on the SARS-CoV-2 polyproteins pp1a and pp2b are qualitatively predicted. These results not only demonstrated the precise and reproducible nature of the YESS 2.0/NGS approach to protease substrate specificity profiling but also should be useful in the design of next generation SARS-CoV-2 Mpro inhibitors, and by analogy, SARS-CoV Mpro inhibitors as well.
Collapse
Affiliation(s)
- Rasha
M. Yaghi
- Department
of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
of America
| | - Collin L. Andrews
- Department
of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
of America
| | - Dennis C. Wylie
- Center
of Biomedical Research Support, University
of Texas at Austin, Austin, Texas 78712, United States of America
| | - Brent L. Iverson
- Department
of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
of America
| |
Collapse
|