1
|
Mallimadugula UL, Cruz MA, Vithani N, Zimmerman MI, Bowman GR. Opening and closing of a cryptic pocket in VP35 toggles it between two different RNA-binding modes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609218. [PMID: 39229186 PMCID: PMC11370563 DOI: 10.1101/2024.08.22.609218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Cryptic pockets are of growing interest as potential drug targets, particularly to control protein-nucleic acid interactions that often occur via flat surfaces. However, it remains unclear whether cryptic pockets contribute to protein function or if they are merely happenstantial features that can easily be evolved away to achieve drug resistance. Here, we explore whether a cryptic pocket in the Interferon Inhibitory Domain (IID) of viral protein 35 (VP35) of Zaire ebolavirus aids its ability to bind double-stranded RNA (dsRNA). We use simulations and experiments to study the relationship between cryptic pocket opening and dsRNA binding of the IIDs of two other filoviruses, Reston and Marburg. These homologs have nearly identical structures but block different interferon pathways due to different affinities for blunt ends and backbone of the dsRNA. Simulations and thiol-labeling experiments demonstrate that the homologs have varying probabilities of pocket opening. Subsequent dsRNA-binding assays suggest that closed conformations preferentially bind dsRNA blunt ends while open conformations prefer binding the backbone. Point mutations that modulate pocket opening proteins further confirm this preference. These results demonstrate the open cryptic pocket has a function, suggesting cryptic pockets are under selective pressure and may be difficult to evolve away to achieve drug resistance.
Collapse
|
2
|
Hayat M, Gao T, Cao Y, Rafiq M, Zhuo L, Li YZ. Identification of Prospective Ebola Virus VP35 and VP40 Protein Inhibitors from Myxobacterial Natural Products. Biomolecules 2024; 14:660. [PMID: 38927063 PMCID: PMC11201620 DOI: 10.3390/biom14060660] [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: 05/09/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
The Ebola virus (EBOV) is a lethal pathogen causing hemorrhagic fever syndrome which remains a global health challenge. In the EBOV, two multifunctional proteins, VP35 and VP40, have significant roles in replication, virion assembly, and budding from the cell and have been identified as druggable targets. In this study, we employed in silico methods comprising molecular docking, molecular dynamic simulations, and pharmacological properties to identify prospective drugs for inhibiting VP35 and VP40 proteins from the myxobacterial bioactive natural product repertoire. Cystobactamid 934-2, Cystobactamid 919-1, and Cittilin A bound firmly to VP35. Meanwhile, 2-Hydroxysorangiadenosine, Enhypyrazinone B, and Sorangiadenosine showed strong binding to the matrix protein VP40. Molecular dynamic simulations revealed that, among these compounds, Cystobactamid 919-1 and 2-Hydroxysorangiadenosine had stable interactions with their respective targets. Similarly, molecular mechanics Poisson-Boltzmann surface area (MMPBSA) calculations indicated close-fitting receptor binding with VP35 or VP40. These two compounds also exhibited good pharmacological properties. In conclusion, we identified Cystobactamid 919-1 and 2-Hydroxysorangiadenosine as potential ligands for EBOV that target VP35 and VP40 proteins. These findings signify an essential step in vitro and in vivo to validate their potential for EBOV inhibition.
Collapse
Affiliation(s)
- Muhammad Hayat
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China; (M.H.); (T.G.); (Y.C.); (Y.-Z.L.)
| | - Tian Gao
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China; (M.H.); (T.G.); (Y.C.); (Y.-Z.L.)
| | - Ying Cao
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China; (M.H.); (T.G.); (Y.C.); (Y.-Z.L.)
| | - Muhammad Rafiq
- Department of Microbiology, Faculty of Life Sciences and Informatics, Balochistan University of IT, Engineering and Management Sciences, Quetta 87100, Pakistan
| | - Li Zhuo
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China; (M.H.); (T.G.); (Y.C.); (Y.-Z.L.)
- Shenzhen Research Institute, Shandong University, Shenzhen 518057, China
- Suzhou Research Institute, Shandong University, Suzhou 215123, China
| | - Yue-Zhong Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China; (M.H.); (T.G.); (Y.C.); (Y.-Z.L.)
| |
Collapse
|
3
|
Bajrai LH, Almalki AA, Sahoo AK, Dwivedi VD, Azhar EI. Exploring plant-derived small molecules as inhibitors of Marburg virus RNA binding protein activity. J Biomol Struct Dyn 2024:1-11. [PMID: 38270432 DOI: 10.1080/07391102.2024.2306500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
The search for antiviral medications is greatly influenced by the hunt for potent inhibitors of viral proteins. To find possible inhibitors of the RNA binding activity of the Marburg virus VP35 protein, we used a thorough in silico drug discovery approach in this investigation. A comprehensive virtual screening process, followed by a detailed MMGBSA analysis, led to the discovery of four potential inhibitory compounds viz. Kudzuisoflavone A, Miquelianin, Rutin, and Protopseudohypericin. They were identified from an extensive library of phytomolecules derived from three medicinal plants: Adiantum capillus-veneris, Hypericum perforatum, and Pueraria montana. In molecular dynamics (MD) simulations, all these compounds showed steady binding to the target protein and favourable interactions. Notably, the free binding energies of all the selected compounds were better than the myricetin, a well-known blocker of the VP35 protein of the Ebola virus. Overall, this investigation offers insightful information about the molecular interactions and binding dynamics of the identified inhibitors' binding to the VP35 protein of the Marburg virus. The findings highlight the potential of three particular medicinal plants as sources of key chemicals for the creation of brand-new Marburg virus antiviral drugs. More experimental validation and optimization of the identified inhibitors are necessary in order to transform these findings into effective medicines for treating Marburg virus infections.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Leena Hussein Bajrai
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman Abdullah Almalki
- Clinical Laboratory Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, India
| | - Vivek Dhar Dwivedi
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India
- Bioinformatics Research Division, Quanta Calculus, Greater Noida, India
| | - Esam Ibraheem Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Collados Rodríguez M, Maillard P, Journeaux A, Komarova AV, Najburg V, David RYS, Helynck O, Guo M, Zhong J, Baize S, Tangy F, Jacob Y, Munier-Lehmann H, Meurs EF. Novel Antiviral Molecules against Ebola Virus Infection. Int J Mol Sci 2023; 24:14791. [PMID: 37834238 PMCID: PMC10573436 DOI: 10.3390/ijms241914791] [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: 08/30/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Infection with Ebola virus (EBOV) is responsible for hemorrhagic fever in humans with a high mortality rate. Combined efforts of prevention and therapeutic intervention are required to tackle highly variable RNA viruses, whose infections often lead to outbreaks. Here, we have screened the 2P2I3D chemical library using a nanoluciferase-based protein complementation assay (NPCA) and isolated two compounds that disrupt the interaction of the EBOV protein fragment VP35IID with the N-terminus of the dsRNA-binding proteins PKR and PACT, involved in IFN response and/or intrinsic immunity, respectively. The two compounds inhibited EBOV infection in cell culture as well as infection by measles virus (MV) independently of IFN induction. Consequently, we propose that the compounds are antiviral by restoring intrinsic immunity driven by PACT. Given that PACT is highly conserved across mammals, our data support further testing of the compounds in other species, as well as against other negative-sense RNA viruses.
Collapse
Affiliation(s)
- Mila Collados Rodríguez
- School of Infection & Immunity (SII), College of Medical, Veterinary and Life Sciences (MVLS), Sir Michael Stoker Building, MRC-University of Glasgow Centre for Virus Research (CVR), Glasgow G61 1QH, UK
- Unité Hépacivirus et Immunité Innée, CNRS, UMR 3569, Département de Virologie, Institut Pasteur, 75015 Paris, France; (P.M.); (E.F.M.)
| | - Patrick Maillard
- Unité Hépacivirus et Immunité Innée, CNRS, UMR 3569, Département de Virologie, Institut Pasteur, 75015 Paris, France; (P.M.); (E.F.M.)
| | - Alexandra Journeaux
- Unit of Biology of Emerging Viral Infections, Institut Pasteur, 69007 Lyon, France; (A.J.); (S.B.)
| | - Anastassia V. Komarova
- Interactomics, RNA and Immunity Laboratory, Institut Pasteur, 75015 Paris, France;
- Unité de Génomique Virale et Vaccination, Institut Pasteur, 75015 Paris, France; (V.N.); (R.-Y.S.D.); (F.T.)
- Université Paris Cité, 75013 Paris, France;
| | - Valérie Najburg
- Unité de Génomique Virale et Vaccination, Institut Pasteur, 75015 Paris, France; (V.N.); (R.-Y.S.D.); (F.T.)
- Université Paris Cité, 75013 Paris, France;
| | - Raul-Yusef Sanchez David
- Unité de Génomique Virale et Vaccination, Institut Pasteur, 75015 Paris, France; (V.N.); (R.-Y.S.D.); (F.T.)
- Blizard Institute—Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Olivier Helynck
- Unité de Chimie et Biocatalyse, CNRS, UMR 3523, Institut Pasteur, Université de Paris, 75015 Paris, France; (O.H.); (H.M.-L.)
| | - Mingzhe Guo
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of Viral Hepatitis, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai 200023, China; (M.G.); (J.Z.)
| | - Jin Zhong
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of Viral Hepatitis, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai 200023, China; (M.G.); (J.Z.)
| | - Sylvain Baize
- Unit of Biology of Emerging Viral Infections, Institut Pasteur, 69007 Lyon, France; (A.J.); (S.B.)
| | - Frédéric Tangy
- Unité de Génomique Virale et Vaccination, Institut Pasteur, 75015 Paris, France; (V.N.); (R.-Y.S.D.); (F.T.)
- Université Paris Cité, 75013 Paris, France;
| | - Yves Jacob
- Université Paris Cité, 75013 Paris, France;
- Unité Génétique Moléculaire des Virus à ARN, CNRS, UMR 3569, Département de Virologie, Institut Pasteur, 75015 Paris, France
| | - Hélène Munier-Lehmann
- Unité de Chimie et Biocatalyse, CNRS, UMR 3523, Institut Pasteur, Université de Paris, 75015 Paris, France; (O.H.); (H.M.-L.)
| | - Eliane F. Meurs
- Unité Hépacivirus et Immunité Innée, CNRS, UMR 3569, Département de Virologie, Institut Pasteur, 75015 Paris, France; (P.M.); (E.F.M.)
| |
Collapse
|
5
|
Wang Z, Yang L. The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection. Nutrients 2023; 15:3443. [PMID: 37571380 PMCID: PMC10421531 DOI: 10.3390/nu15153443] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
The exploration of non-toxic and cost-effective dietary components, such as epigallocatechin 3-gallate and myricetin, for health improvement and disease treatment has recently attracted substantial research attention. The recent COVID-19 pandemic has provided a unique opportunity for the investigation and identification of dietary components capable of treating viral infections, as well as gathering the evidence needed to address the major challenges presented by public health emergencies. Dietary components hold great potential as a starting point for further drug development for the treatment and prevention of SARS-CoV-2 infection owing to their good safety, broad-spectrum antiviral activities, and multi-organ protective capacity. Here, we review current knowledge of the characteristics-chemical composition, bioactive properties, and putative mechanisms of action-of natural bioactive dietary flavonoids with the potential for targeting SARS-CoV-2 and its variants. Notably, we present promising strategies (combination therapy, lead optimization, and drug delivery) to overcome the inherent deficiencies of natural dietary flavonoids, such as limited bioavailability and poor stability.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus, Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, China
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
6
|
Giordano D, Facchiano A, Carbone V. Food Plant Secondary Metabolites Antiviral Activity and Their Possible Roles in SARS-CoV-2 Treatment: An Overview. Molecules 2023; 28:molecules28062470. [PMID: 36985442 PMCID: PMC10058909 DOI: 10.3390/molecules28062470] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Natural products and plant extracts exhibit many biological activities, including that related to the defense mechanisms against parasites. Many studies have investigated the biological functions of secondary metabolites and reported evidence of antiviral activities. The pandemic emergencies have further increased the interest in finding antiviral agents, and efforts are oriented to investigate possible activities of secondary plant metabolites against human viruses and their potential application in treating or preventing SARS-CoV-2 infection. In this review, we performed a comprehensive analysis of studies through in silico and in vitro investigations, also including in vivo applications and clinical trials, to evaluate the state of knowledge on the antiviral activities of secondary metabolites against human viruses and their potential application in treating or preventing SARS-CoV-2 infection, with a particular focus on natural compounds present in food plants. Although some of the food plant secondary metabolites seem to be useful in the prevention and as a possible therapeutic management against SARS-CoV-2, up to now, no molecules can be used as a potential treatment for COVID-19; however, more research is needed.
Collapse
Affiliation(s)
- Deborah Giordano
- Institute of Food Sciences, National Research Council, via Roma 64, 83100 Avellino, Italy
| | - Angelo Facchiano
- Institute of Food Sciences, National Research Council, via Roma 64, 83100 Avellino, Italy
| | - Virginia Carbone
- Institute of Food Sciences, National Research Council, via Roma 64, 83100 Avellino, Italy
| |
Collapse
|
7
|
Zinzula L, Mereu AM, Orsini M, Seeleitner C, Bracher A, Nagy I, Baumeister W. Ebola and Marburg virus VP35 coiled-coil validated as antiviral target by tripartite split-GFP complementation. iScience 2022; 25:105354. [PMID: 36325051 PMCID: PMC9619376 DOI: 10.1016/j.isci.2022.105354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
Ebola virus (EBOV) and Marburg virus (MARV) are highly pathogenic viruses in humans, against which approved antivirals are lacking. During EBOV and MARV infection, coiled-coil mediated oligomerization is essential for the virion protein 35 (VP35) polymerase co-factor function and type I interferon antagonism, making VP35 coiled-coil an elective drug target. We established a tripartite split-green fluorescent protein (GFP) fluorescence complementation (FC) system based on recombinant GFP-tagged EBOV and MARV VP35, which probes VP35 coiled-coil assembly by monitoring fluorescence on E. coli colonies, or in vitro in 96/384-multiwell. Oligomerization-defective VP35 mutants showed that correct coiled-coil knobs-into-holes pairing within VP35 oligomer is pre-requisite for GFP tags and GFP detector to reconstitute fluorescing full-length GFP. The method was validated by screening a small compound library, which identified Myricetin and 4,5,6,7-Tetrabromobenzotriazole as inhibitors of EBOV and MARV VP35 oligomerization-dependent FC with low-micromolar IC50 values. These findings substantiate the VP35 coiled-coil value as antiviral target. Ebola and Marburg virus VP35 oligomerize via trimeric and tetrameric coiled-coil VP35 coiled-coil assembly triggers fluorescence of a tripartite split-GFP system Mutations perturbing VP35 coiled-coil hamper split-GFP complementation Myricetin and TBBT inhibit split-GFP complementation mediated by VP35 coiled-coil
Collapse
Affiliation(s)
- Luca Zinzula
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
- Corresponding author
| | - Angela Maria Mereu
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Massimiliano Orsini
- Istituto Zooprofilattico Sperimentale delle Venezie, Department of Risk Analysis and Public Health Surveillance, Viale dell’Università 10, 35020 Legnaro, Italy
| | - Christine Seeleitner
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Andreas Bracher
- The Max-Planck Institute of Biochemistry, Department of Cellular Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - István Nagy
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Wolfgang Baumeister
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
- Corresponding author
| |
Collapse
|
8
|
Kuo C, Nikan M, Yeh ST, Chappell AE, Tanowitz M, Seth PP, Prakash TP, Mullick AE. Targeted Delivery of Antisense Oligonucleotides Through Angiotensin Type 1 Receptor. Nucleic Acid Ther 2022; 32:300-311. [PMID: 35612431 DOI: 10.1089/nat.2021.0105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We evaluated the potential of AGTR1, the principal receptor for angiotensin II (Ang II) and a member of the G protein-coupled receptor family, for targeted delivery of antisense oligonucleotides (ASOs) in cells and tissues with abundant AGTR1 expression. Ang II peptide ASO conjugates maintained robust AGTR1 signaling and receptor internalization when ASO was placed at the N-terminus of the peptide, but not at C-terminus. Conjugation of Ang II peptide improved ASO potency up to 12- to 17-fold in AGTR1-expressing cells. Additionally, evaluation of Ang II conjugates in cells lacking AGTR1 revealed no enhancement of ASO potency. Ang II peptide conjugation improves potency of ASO in mouse heart, adrenal, and adipose tissues. The data presented in this report add to a growing list of approaches for improving ASO potency in extrahepatic tissues.
Collapse
Affiliation(s)
- Carol Kuo
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Mehran Nikan
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Steve T Yeh
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | | | | | - Punit P Seth
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | | | | |
Collapse
|
9
|
Cruz MA, Frederick TE, Mallimadugula UL, Singh S, Vithani N, Zimmerman MI, Porter JR, Moeder KE, Amarasinghe GK, Bowman GR. A cryptic pocket in Ebola VP35 allosterically controls RNA binding. Nat Commun 2022; 13:2269. [PMID: 35477718 PMCID: PMC9046395 DOI: 10.1038/s41467-022-29927-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/07/2022] [Indexed: 11/08/2022] Open
Abstract
Protein-protein and protein-nucleic acid interactions are often considered difficult drug targets because the surfaces involved lack obvious druggable pockets. Cryptic pockets could present opportunities for targeting these interactions, but identifying and exploiting these pockets remains challenging. Here, we apply a general pipeline for identifying cryptic pockets to the interferon inhibitory domain (IID) of Ebola virus viral protein 35 (VP35). VP35 plays multiple essential roles in Ebola's replication cycle but lacks pockets that present obvious utility for drug design. Using adaptive sampling simulations and machine learning algorithms, we predict VP35 harbors a cryptic pocket that is allosterically coupled to a key dsRNA-binding interface. Thiol labeling experiments corroborate the predicted pocket and mutating the predicted allosteric network supports our model of allostery. Finally, covalent modifications that mimic drug binding allosterically disrupt dsRNA binding that is essential for immune evasion. Based on these results, we expect this pipeline will be applicable to other proteins.
Collapse
Affiliation(s)
- Matthew A Cruz
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Thomas E Frederick
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Upasana L Mallimadugula
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sukrit Singh
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Neha Vithani
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Maxwell I Zimmerman
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Justin R Porter
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Katelyn E Moeder
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gregory R Bowman
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Center for the Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
10
|
Corona A, Wycisk K, Talarico C, Manelfi C, Milia J, Cannalire R, Esposito F, Gribbon P, Zaliani A, Iaconis D, Beccari AR, Summa V, Nowotny M, Tramontano E. Natural Compounds Inhibit SARS-CoV-2 nsp13 Unwinding and ATPase Enzyme Activities. ACS Pharmacol Transl Sci 2022; 5:226-239. [PMID: 35434533 PMCID: PMC9003574 DOI: 10.1021/acsptsci.1c00253] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Indexed: 12/27/2022]
Abstract
![]()
SARS-CoV-2 infection
is still spreading worldwide, and new antiviral
therapies are an urgent need to complement the approved vaccine preparations.
SARS-CoV-2 nps13 helicase is a validated drug target participating
in the viral replication complex and possessing two associated activities:
RNA unwinding and 5′-triphosphatase. In the search of SARS-CoV-2
direct antiviral agents, we established biochemical assays for both
SARS-CoV-2 nps13-associated enzyme activities and screened both in silico and in vitro a small in-house
library of natural compounds. Myricetin, quercetin, kaempferol, and
flavanone were found to inhibit the SARS-CoV-2 nps13 unwinding activity
at nanomolar concentrations, while licoflavone C was shown to block
both SARS-CoV-2 nps13 activities at micromolar concentrations. Mode
of action studies showed that all compounds are nsp13 noncompetitive
inhibitors versus ATP, while computational studies suggested that
they can bind both nucleotide and 5′-RNA nsp13 binding sites,
with licoflavone C showing a unique pattern of interaction with nsp13
amino acid residues. Overall, we report for the first time natural
flavonoids as selective inhibitors of SARS-CoV-2 nps13 helicase with
low micromolar activity.
Collapse
Affiliation(s)
- Angela Corona
- Dipartimento di Scienze della vita e dell'ambiente, Università degli Studi di Cagliari, Cittadella Universitaria di Monserrato, SS-554, 09042 Monserrato, Cagliari, Italy
| | - Krzysztof Wycisk
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, Warsaw 02-109, Poland
| | - Carmine Talarico
- Dompé Farmaceutici SpA, via Campo di Pile, 67100 L'Aquila, Italy
| | - Candida Manelfi
- Dompé Farmaceutici SpA, via Campo di Pile, 67100 L'Aquila, Italy
| | - Jessica Milia
- Dipartimento di Scienze della vita e dell'ambiente, Università degli Studi di Cagliari, Cittadella Universitaria di Monserrato, SS-554, 09042 Monserrato, Cagliari, Italy
| | - Rolando Cannalire
- Department of Pharmacy, University of Napoli "Federico II", via D. Montesano 49, Napoli 80131, Italy
| | - Francesca Esposito
- Dipartimento di Scienze della vita e dell'ambiente, Università degli Studi di Cagliari, Cittadella Universitaria di Monserrato, SS-554, 09042 Monserrato, Cagliari, Italy
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany.,Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany.,Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Daniela Iaconis
- Dompé Farmaceutici SpA, via Campo di Pile, 67100 L'Aquila, Italy
| | - Andrea R Beccari
- Dompé Farmaceutici SpA, via Campo di Pile, 67100 L'Aquila, Italy
| | - Vincenzo Summa
- Department of Pharmacy, University of Napoli "Federico II", via D. Montesano 49, Napoli 80131, Italy
| | - Marcin Nowotny
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology, Ks. Trojdena 4, Warsaw 02-109, Poland
| | - Enzo Tramontano
- Dipartimento di Scienze della vita e dell'ambiente, Università degli Studi di Cagliari, Cittadella Universitaria di Monserrato, SS-554, 09042 Monserrato, Cagliari, Italy
| |
Collapse
|
11
|
Liu CH, Hu YT, Wong SH, Lin LT. Therapeutic Strategies against Ebola Virus Infection. Viruses 2022; 14:v14030579. [PMID: 35336986 PMCID: PMC8954160 DOI: 10.3390/v14030579] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Since the 2014–2016 epidemic, Ebola virus (EBOV) has spread to several countries and has become a major threat to global health. EBOV is a risk group 4 pathogen, which imposes significant obstacles for the development of countermeasures against the virus. Efforts have been made to develop anti-EBOV immunization and therapeutics, with three vaccines and two antibody-based therapeutics approved in recent years. Nonetheless, the high fatality of Ebola virus disease highlights the need to continuously develop antiviral strategies for the future management of EBOV outbreaks in conjunction with vaccination programs. This review aims to highlight potential EBOV therapeutics and their target(s) of inhibition, serving as a summary of the literature to inform readers of the novel candidates available in the continued search for EBOV antivirals.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yee-Tung Hu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Shu Hui Wong
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Correspondence:
| |
Collapse
|
12
|
Agraharam G, Girigoswami A, Girigoswami K. Myricetin: a Multifunctional Flavonol in Biomedicine. CURRENT PHARMACOLOGY REPORTS 2022; 8:48-61. [PMID: 35036292 PMCID: PMC8743163 DOI: 10.1007/s40495-021-00269-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVEIW The root cause of many diseases like CVD, cancer, and aging is free radicals which exert their effect by interfering with different metabolic pathways. The sources of free radicals can be exogenous, like UV rays from sunlight, and endogenous due to different metabolic by-products.In our body, there are defense mechanisms present, such as antioxidant enzymes and antioxidant molecules to combat these free radicals, but if there is an overload of these free radicals in our body, the defense system may not be sufficient to neutralize these free radicals. In such situations, we are exposed to a chronic low dose of oxidants creating oxidative stress, which is responsible for eliciting different diseases. RECENT FINDINGS Pubmed and Google Scholar are the search engines used to sort out relevant papers on myricetin and its role in combating many diseases. Myricetin is present in many fruits and vegetables and is a known antioxidant. It can elevate the antioxidant enzyme levels; reduces the lipid peroxidation; and is known to protect against cancer. In the case of myocardial dysfunction, myricetin has been shown to suppress the inflammatory cytokines and reduced the mortality rate. Myricetin has also been found to reduce platelet aggregation and control the viral infections by interfering in the DNA replication pathways. SUMMARY In this paper, we have briefly reviewed about the different type and site of free radicals and the role of myricetin in addressing the ROS and different diseases.
Collapse
Affiliation(s)
- Gopikrishna Agraharam
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103 Tamilnadu India
| | - Agnishwar Girigoswami
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103 Tamilnadu India
| | - Koyeli Girigoswami
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103 Tamilnadu India
| |
Collapse
|
13
|
Corona A, Fanunza E, Salata C, Morwitzer MJ, Distinto S, Zinzula L, Sanna C, Frau A, Daino GL, Quartu M, Taglialatela-Scafati O, Rigano D, Reid S, Mirazimi A, Tramontano E. Cynarin blocks Ebola virus replication by counteracting VP35 inhibition of interferon-beta production. Antiviral Res 2022; 198:105251. [DOI: 10.1016/j.antiviral.2022.105251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 11/02/2022]
|
14
|
INMI1 Zika Virus NS4B Antagonizes the Interferon Signaling by Suppressing STAT1 Phosphorylation. Viruses 2021; 13:v13122448. [PMID: 34960717 PMCID: PMC8705506 DOI: 10.3390/v13122448] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
The evasion of the Interferon response has important implications in Zika virus (ZIKV) disease. Mutations in ZIKV viral protein NS4B, associated with modulation of the interferon (IFN) system, have been linked to increased pathogenicity in animal models. In this study, we unravel ZIKV NS4B as antagonist of the IFN signaling cascade. Firstly, we reported the genomic characterization of NS4B isolated from a strain of the 2016 outbreak, ZIKV Brazil/2016/INMI1, and we predicted its membrane topology. Secondly, we analyzed its phylogenetic correlation with other flaviviruses, finding a high similarity with dengue virus 2 (DEN2) strains; in particular, the highest conservation was found when NS4B was aligned with the IFN inhibitory domain of DEN2 NS4B. Hence, we asked whether ZIKV NS4B was also able to inhibit the IFN signaling cascade, as reported for DEN2 NS4B. Our results showed that ZIKV NS4B was able to strongly inhibit the IFN stimulated response element and the IFN-γ-activated site transcription, blocking IFN-I/-II responses. mRNA expression levels of the IFN stimulated genes ISG15 and OAS1 were also strongly reduced in presence of NS4B. We found that the viral protein was acting by suppressing the STAT1 phosphorylation and consequently blocking the nuclear transport of both STAT1 and STAT2.
Collapse
|
15
|
Fakhri S, Mohammadi Pour P, Piri S, Farzaei MH, Echeverría J. Modulating Neurological Complications of Emerging Infectious Diseases: Mechanistic Approaches to Candidate Phytochemicals. Front Pharmacol 2021; 12:742146. [PMID: 34764869 PMCID: PMC8576094 DOI: 10.3389/fphar.2021.742146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/23/2021] [Indexed: 12/02/2022] Open
Abstract
Growing studies are revealing the critical manifestations of influenza, dengue virus (DENV) infection, Zika virus (ZIKV) disease, and Ebola virus disease (EVD) as emerging infectious diseases. However, their corresponding mechanisms of major complications headed for neuronal dysfunction are not entirely understood. From the mechanistic point of view, inflammatory/oxidative mediators are activated during emerging infectious diseases towards less cell migration, neurogenesis impairment, and neuronal death. Accordingly, the virus life cycle and associated enzymes, as well as host receptors, cytokine storm, and multiple signaling mediators, are the leading players of emerging infectious diseases. Consequently, chemokines, interleukins, interferons, carbohydrate molecules, toll-like receptors (TLRs), and tyrosine kinases are leading orchestrates of peripheral and central complications which are in near interconnections. Some of the resulting neuronal manifestations have attracted much attention, including inflammatory polyneuropathy, encephalopathy, meningitis, myelitis, stroke, Guillain-Barré syndrome (GBS), radiculomyelitis, meningoencephalitis, memory loss, headaches, cranial nerve abnormalities, tremor, and seizure. The complex pathophysiological mechanism behind the aforementioned complications urges the need for finding multi-target agents with higher efficacy and lower side effects. In recent decades, the natural kingdom has been highlighted as promising neuroprotective natural products in modulating several dysregulated signaling pathways/mediators. The present study provides neuronal manifestations of some emerging infectious diseases and underlying pathophysiological mechanisms. Besides, a mechanistic-based strategy is developed to introduce candidate natural products as promising multi-target agents in combating major dysregulated pathways towards neuroprotection in influenza, DENV infection, ZIKV disease, and EVD.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pardis Mohammadi Pour
- Department of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
16
|
Song H, Lee PG, Kim H, Lee UJ, Lee SH, Kim J, Kim BG. Polyphenol-Hydroxylating Tyrosinase Activity under Acidic pH Enables Efficient Synthesis of Plant Catechols and Gallols. Microorganisms 2021; 9:microorganisms9091866. [PMID: 34576760 PMCID: PMC8466947 DOI: 10.3390/microorganisms9091866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Tyrosinase is generally known as a melanin-forming enzyme, facilitating monooxygenation of phenols, oxidation of catechols into quinones, and finally generating biological melanin. As a homologous form of tyrosinase in plants, plant polyphenol oxidases perform the same oxidation reactions specifically toward plant polyphenols. Recent studies reported synthetic strategies for large scale preparation of hydroxylated plant polyphenols, using bacterial tyrosinases rather than plant polyphenol oxidase or other monooxygenases, by leveraging its robust monophenolase activity and broad substrate specificity. Herein, we report a novel synthesis of functional plant polyphenols, especially quercetin and myricetin from kaempferol, using screened bacterial tyrosinases. The critical bottleneck of the biocatalysis was identified as instability of the catechol and gallol under neutral and basic conditions. To overcome such instability of the products, the tyrosinase reaction proceeded under acidic conditions. Under mild acidic conditions supplemented with reducing agents, a bacterial tyrosinase from Bacillus megaterium (BmTy) displayed efficient consecutive two-step monophenolase activities producing quercetin and myricetin from kaempferol. Furthermore, the broad substrate specificity of BmTy toward diverse polyphenols enabled us to achieve the first biosynthesis of tricetin and 3'-hydroxyeriodictyol from apigenin and naringenin, respectively. These results suggest that microbial tyrosinase is a useful biocatalyst to prepare plant polyphenolic catechols and gallols with high productivity, which were hardly achieved by using other monooxygenases such as cytochrome P450s.
Collapse
Affiliation(s)
- Hanbit Song
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Pyung-Gang Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
- Institute of Engineering Research, Seoul National University, Seoul 08826, Korea
| | - Hyun Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Uk-Jae Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Sang-Hyuk Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Joonwon Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Byung-Gee Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (H.S.); (P.-G.L.); (H.K.); (U.-J.L.); (S.-H.L.); (J.K.)
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
- Bio-MAX/N-Bio Institute, Seoul National University, Seoul 08826, Korea
- Institute for Sustainable Development (ISD), Seoul National University, Seoul 08826, Korea
- Correspondence: ; Tel.: +82-2-880-6774; Fax: +82-2-876-8945
| |
Collapse
|
17
|
Fanunza E, Carletti F, Quartu M, Grandi N, Ermellino L, Milia J, Corona A, Capobianchi MR, Ippolito G, Tramontano E. Zika virus NS2A inhibits interferon signaling by degradation of STAT1 and STAT2. Virulence 2021; 12:1580-1596. [PMID: 34338586 PMCID: PMC8331042 DOI: 10.1080/21505594.2021.1935613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The Interferon (IFN) response is crucial to restrain pathogenic infections. Investigations into flavivirus-host interactions reported that the high virulence is linked to innate immune evasion. Zika Virus (ZIKV) has developed diversified strategies to evade the innate immune system. We report that the viral protein NS2A counteracts the IFN response by strongly suppressing the IFN signaling. NS2A targets transcription factors STAT1 and STAT2, to impede their nuclear localization, thereby suppressing the transcription of ISRE promoter and IFN-stimulated genes. We found that NS2A promotes degradation of STAT1 and STAT2. Treatment of NS2A transfected cells with MG132 restores the levels of both transcription factors, suggesting the involvement of the proteasome system. Given the impact that the IFN antagonism has on flavivirus virulence, the knowledge gained by characterizing the mechanism through which ZIKV evades the IFN response paves the ground for new strategies to attenuate the pathogenesis and to develop countermeasures against effective pharmacological targets.
Collapse
Affiliation(s)
- Elisa Fanunza
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Fabrizio Carletti
- Laboratory of Virology, National Institute for Infectious Diseases, L.Spallanzani͟ IRCCS, Rome, Italy
| | - Marina Quartu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Nicole Grandi
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Laura Ermellino
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Jessica Milia
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases, L.Spallanzani͟ IRCCS, Rome, Italy
| | - Giuseppe Ippolito
- Laboratory of Virology, National Institute for Infectious Diseases, L.Spallanzani͟ IRCCS, Rome, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| |
Collapse
|
18
|
Joshi RS, Jagdale SS, Bansode SB, Shankar SS, Tellis MB, Pandya VK, Chugh A, Giri AP, Kulkarni MJ. Discovery of potential multi-target-directed ligands by targeting host-specific SARS-CoV-2 structurally conserved main protease. J Biomol Struct Dyn 2021; 39:3099-3114. [PMID: 32329408 PMCID: PMC7212545 DOI: 10.1080/07391102.2020.1760137] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has resulted in the current COVID-19 pandemic. Worldwide this disease has infected over 2.5 million individuals with a mortality rate ranging from 5 to 10%. There are several efforts going on in the drug discovery to control the SARS-CoV-2 viral infection. The main protease (MPro) plays a critical role in viral replication and maturation, thus can serve as the primary drug target. To understand the structural evolution of MPro, we have performed phylogenetic and Sequence Similarity Network analysis, that depicted divergence of Coronaviridae MPro in five clusters specific to viral hosts. This clustering was corroborated with the comparison of MPro structures. Furthermore, it has been observed that backbone and binding site conformations are conserved despite variation in some of the residues. These attributes can be exploited to repurpose available viral protease inhibitors against SARS-CoV-2 MPro. In agreement with this, we performed screening of ∼7100 molecules including active ingredients present in the Ayurvedic anti-tussive medicines, anti-viral phytochemicals and synthetic anti-virals against SARS-CoV-2 MPro as the primary target. We identified several natural molecules like δ-viniferin, myricitrin, taiwanhomoflavone A, lactucopicrin 15-oxalate, nympholide A, afzelin, biorobin, hesperidin and phyllaemblicin B that strongly binds to SARS-CoV-2 MPro. Intrestingly, these molecules also showed strong binding with other potential targets of SARS-CoV-2 infection like viral receptor human angiotensin-converting enzyme 2 (hACE-2) and RNA dependent RNA polymerase (RdRp). We anticipate that our approach for identification of multi-target-directed ligand will provide new avenues for drug discovery against SARS-CoV-2 infection.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rakesh S. Joshi
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shounak S. Jagdale
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
| | - Sneha B. Bansode
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
| | - S. Shiva Shankar
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Meenakshi B. Tellis
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
- Department of Botany, Savitribai Phule Pune University, Pune, India
| | | | | | - Ashok P. Giri
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mahesh J. Kulkarni
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
19
|
Ma H, Song X, Huang P, Zhang W, Ling X, Yang X, Wu W, Xu H, Wang W. Myricetin protects natural killer cells from arsenite induced DNA damage by attenuating oxidative stress and retaining poly(ADP-Ribose) polymerase 1 activity. Mutat Res 2021; 865:503337. [PMID: 33865543 DOI: 10.1016/j.mrgentox.2021.503337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/30/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
Environmental exposure to arsenite (As+3) is known to induce immunotoxicity. Natural killer (NK) cells are innate lymphoid cells act as professional killers of tumor cells. Our previous report indicated that 500 ppb As+3 drinking water exposure induced significant DNA damage in the NK cells of C57BL/6 mice. Myricetin is a plant-derived flavonoid known as a strong antioxidant. In this study, daily administration of myricetin at 20 mg/kg was found to alleviate the cell population decrease and DNA damage in the NK cells of BALB/c mice exposed to 500 and 1000 ppb As+3 via drinking water. Oxidative stress and poly(ADP-ribose) polymerase 1 (PARP-1) inhibition were induced by As+3 at 1 and 2 μM in isolated mouse NK cells in vitro, which were attenuated by 20 μM myricetin. The mitigatory effect of myricetin on the PARP-1 inhibition in NK cells treated with As+3 was also found to be the result of its prevention of the zinc loss induced by As+3 on PARP-1. Collectively, these results demonstrated, for the first time, that myricetin could protect NK cells from As+3 induced DNA through attenuating oxidative stress and retaining PARP-1 activity, indicating that myricetin may be utilized for the prevention of the immunotoxicity induced by As+3 in NK cells.
Collapse
Affiliation(s)
- Huijuan Ma
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China
| | - Xiaodong Song
- Medical Laboratory Department, Hua Shan Hospital North, Fudan University, Shanghai, 201907, China
| | - Ping Huang
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China
| | - Weiwei Zhang
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China
| | - Xinyue Ling
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China
| | - Xiaoning Yang
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China
| | - Wenwei Wu
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China
| | - Huan Xu
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China.
| | - Wei Wang
- East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai, 200237, China; East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai, 200237, China; Department of Pharmacology and Toxicology and BIO5 Institute, University of Arizona, Tucson, AZ, 85721-0207, USA.
| |
Collapse
|
20
|
Christy MP, Uekusa Y, Gerwick L, Gerwick WH. Natural Products with Potential to Treat RNA Virus Pathogens Including SARS-CoV-2. JOURNAL OF NATURAL PRODUCTS 2021; 84:161-182. [PMID: 33352046 PMCID: PMC7771248 DOI: 10.1021/acs.jnatprod.0c00968] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Indexed: 05/03/2023]
Abstract
Three families of RNA viruses, the Coronaviridae, Flaviviridae, and Filoviridae, collectively have great potential to cause epidemic disease in human populations. The current SARS-CoV-2 (Coronaviridae) responsible for the COVID-19 pandemic underscores the lack of effective medications currently available to treat these classes of viral pathogens. Similarly, the Flaviviridae, which includes such viruses as Dengue, West Nile, and Zika, and the Filoviridae, with the Ebola-type viruses, as examples, all lack effective therapeutics. In this review, we present fundamental information concerning the biology of these three virus families, including their genomic makeup, mode of infection of human cells, and key proteins that may offer targeted therapies. Further, we present the natural products and their derivatives that have documented activities to these viral and host proteins, offering hope for future mechanism-based antiviral therapeutics. By arranging these potential protein targets and their natural product inhibitors by target type across these three families of virus, new insights are developed, and crossover treatment strategies are suggested. Hence, natural products, as is the case for other therapeutic areas, continue to be a promising source of structurally diverse new anti-RNA virus therapeutics.
Collapse
Affiliation(s)
- Mitchell P. Christy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - Yoshinori Uekusa
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Lena Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - William H. Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
21
|
Sanna C, Maxia A, Fenu G, Loi MC. So Uncommon and so Singular, but Underexplored: An Updated Overview on Ethnobotanical Uses, Biological Properties and Phytoconstituents of Sardinian Endemic Plants. PLANTS (BASEL, SWITZERLAND) 2020; 9:E958. [PMID: 32751394 PMCID: PMC7465485 DOI: 10.3390/plants9080958] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
The last decades have recorded an increase of plant-based drug discovery processes. Indeed, natural products possess a superior chemical diversity as compared to synthetic ones, leading to a renewal in searching for new therapeutic agents from the plant kingdom. In particular, since the structural variety of natural compounds reflects the biodiversity of their source organisms, regions of the world with high biodiversity and endemism deserve particular interest. In this context, Sardinia Island (Italy), with 290 endemic taxa (12% of the total flora), is expected to provide unique and structurally diverse phytochemicals for drug development. Several research groups built up a large program dedicated to the analysis of Sardinian endemic species, highlighting their peculiar features, both in respect of phytochemical and biological profiles. On this basis, the aim of this review is to provide an up-to-date and comprehensive overview on ethnobotanical uses, biological properties and phytoconstituents of Sardinian endemic plants in order to support their beneficial potential and to provide input for future investigations. We documented 152 articles published from 1965 to June 2020 in which a broad range of biological activities and the identification of previously undescribed compounds have been reported, supporting their great value as sources of therapeutic agents.
Collapse
Affiliation(s)
- Cinzia Sanna
- Department of Life and Environmental Sciences, University of Cagliari, Via Sant’Ignazio da Laconi 13, 09123 Cagliari, Italy; (A.M.); (G.F.); (M.C.L.)
- Co.S.Me.Se—Consorzio per lo Studio dei Metaboliti Secondari, Via Sant’Ignazio da Laconi 13, 09123 Cagliari, Italy
| | - Andrea Maxia
- Department of Life and Environmental Sciences, University of Cagliari, Via Sant’Ignazio da Laconi 13, 09123 Cagliari, Italy; (A.M.); (G.F.); (M.C.L.)
- Co.S.Me.Se—Consorzio per lo Studio dei Metaboliti Secondari, Via Sant’Ignazio da Laconi 13, 09123 Cagliari, Italy
| | - Giuseppe Fenu
- Department of Life and Environmental Sciences, University of Cagliari, Via Sant’Ignazio da Laconi 13, 09123 Cagliari, Italy; (A.M.); (G.F.); (M.C.L.)
| | - Maria Cecilia Loi
- Department of Life and Environmental Sciences, University of Cagliari, Via Sant’Ignazio da Laconi 13, 09123 Cagliari, Italy; (A.M.); (G.F.); (M.C.L.)
| |
Collapse
|
22
|
Wilson BAP, Thornburg CC, Henrich CJ, Grkovic T, O'Keefe BR. Creating and screening natural product libraries. Nat Prod Rep 2020; 37:893-918. [PMID: 32186299 PMCID: PMC8494140 DOI: 10.1039/c9np00068b] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Covering: up to 2020The National Cancer Institute of the United States (NCI) has initiated a Cancer Moonshot program entitled the NCI Program for Natural Product Discovery. As part of this effort, the NCI is producing a library of 1 000 000 partially purified natural product fractions which are being plated into 384-well plates and provided to the research community free of charge. As the first 326 000 of these fractions have now been made available, this review seeks to describe the general methods used to collect organisms, extract those organisms, and create a prefractionated library. Importantly, this review also details both cell-based and cell-free bioassay methods and the adaptations necessary to those methods to productively screen natural product libraries. Finally, this review briefly describes post-screen dereplication and compound purification and scale up procedures which can efficiently identify active compounds and produce sufficient quantities of natural products for further pre-clinical development.
Collapse
Affiliation(s)
- Brice A P Wilson
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, USA.
| | | | | | | | | |
Collapse
|
23
|
Quercetin Blocks Ebola Virus Infection by Counteracting the VP24 Interferon-Inhibitory Function. Antimicrob Agents Chemother 2020; 64:AAC.00530-20. [PMID: 32366711 DOI: 10.1128/aac.00530-20] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/28/2020] [Indexed: 01/03/2023] Open
Abstract
Ebola virus (EBOV) is among the most devastating pathogens causing fatal hemorrhagic fever in humans. The epidemics from 2013 to 2016 resulted in more than 11,000 deaths, and another outbreak is currently ongoing. Since there is no FDA-approved drug so far to fight EBOV infection, there is an urgent need to focus on drug discovery. Considering the tight correlation between the high EBOV virulence and its ability to suppress the type I interferon (IFN-I) system, identifying molecules targeting viral protein VP24, one of the main virulence determinants blocking the IFN response, is a promising novel anti-EBOV therapy approach. Hence, in the effort to find novel EBOV inhibitors, a screening of a small set of flavonoids was performed; it showed that quercetin and wogonin can suppress the VP24 effect on IFN-I signaling inhibition. The mechanism of action of the most active compound, quercetin, showing a half-maximal inhibitory concentration (IC50) of 7.4 μM, was characterized to significantly restore the IFN-I signaling cascade, blocked by VP24, by directly interfering with the VP24 binding to karyopherin-α and thus restoring P-STAT1 nuclear transport and IFN gene transcription. Quercetin significantly blocked viral infection, specifically targeting EBOV VP24 anti-IFN-I function. Overall, quercetin is the first identified inhibitor of the EBOV VP24 anti-IFN function, representing a molecule interacting with a viral binding site that is very promising for further drug development aiming to block EBOV infection at the early steps.
Collapse
|
24
|
Fanunza E, Frau A, Corona A, Tramontano E. Insights into Ebola Virus VP35 and VP24 Interferon Inhibitory Functions and their Initial Exploitation as Drug Targets. Infect Disord Drug Targets 2020; 19:362-374. [PMID: 30468131 DOI: 10.2174/1871526519666181123145540] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 12/15/2022]
Abstract
Upon viral infection, the interferon (IFN) system triggers potent antiviral mechanisms limiting viral growth and spread. Hence, to sustain their infection, viruses evolved efficient counteracting strategies to evade IFN control. Ebola virus (EBOV), member of the family Filoviridae, is one of the most virulent and deadly pathogen ever faced by humans. The etiological agent of the Ebola Virus Disease (EVD), EBOV can be undoubtedly considered the perfect example of a powerful inhibitor of the host organism immune response activation. Particularly, the efficacious suppression of the IFN cascade contributes to disease progression and severity. Among the EBOVencoded proteins, the Viral Proteins 35 (VP35) and 24 (VP24) are responsible for the EBOV extreme virulence, representing the core of such inhibitory function through which EBOV determines its very effective shield to the cellular immune defenses. VP35 inhibits the activation of the cascade leading to IFN production, while VP24 inhibits the activation of the IFN-stimulated genes. A number of studies demonstrated that both VP35 and VP24 is validated target for drug development. Insights into the structural characteristics of VP35 and VP24 domains revealed crucial pockets exploitable for drug development. Considered the lack of therapy for EVD, restoring the immune activation is a promising approach for drug development. In the present review, we summarize the importance of VP35 and VP24 proteins in counteracting the host IFN cellular response and discuss their potential as druggable viral targets as a promising approach toward attenuation of EBOV virulence.
Collapse
Affiliation(s)
- Elisa Fanunza
- Department of Life and Environmental Sciences, University of Cagliari, Sardinia, Italy
| | - Aldo Frau
- Department of Life and Environmental Sciences, University of Cagliari, Sardinia, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Sardinia, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Sardinia, Italy.,Genetics and Biomedical Research Institute, National Research Council, Monserrato, Italy
| |
Collapse
|
25
|
Flego M, Frau A, Accardi L, Mallano A, Ascione A, Gellini M, Fanunza E, Vella S, Di Bonito P, Tramontano E. Intracellular human antibody fragments recognizing the VP35 protein of Zaire Ebola filovirus inhibit the protein activity. BMC Biotechnol 2019; 19:64. [PMID: 31488108 PMCID: PMC6727353 DOI: 10.1186/s12896-019-0554-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Ebola hemorrhagic fever is caused by the Ebola filovirus (EBOV), which is one of the most aggressive infectious agents known worldwide. The EBOV pathogenesis starts with uncontrolled viral replication and subversion of both the innate and adaptive host immune response. The multifunctional viral VP35 protein is involved in this process by exerting an antagonistic action against the early antiviral alpha/beta interferon (IFN-α/β) response, and represents a suitable target for the development of strategies to control EBOV infection. Phage display technology permits to select antibodies as single chain Fragment variable (scFv) from an artificial immune system, due to their ability to specifically recognize the antigen of interest. ScFv is ideal for genetic manipulation and to obtain antibody constructs useful for targeting either antigens expressed on cell surface or intracellular antigens if the scFv is expressed as intracellular antibody (intrabody) or delivered into the cells. RESULTS Monoclonal antibodies (mAb) in scFv format specific for the EBOV VP35 were isolated from the ETH-2 library of human recombinant antibodies by phage display technology. Five different clones were identified by sequencing, produced in E.coli and expressed in CHO mammalian cells to be characterized in vitro. All the selected scFvs were able to react with recombinant VP35 protein in ELISA, one of the scFvs being also able to react in Western Blot assay (WB). In addition, all scFvs were expressed in cell cytoplasm as intrabodies; a luciferase reporter gene inhibition assay performed in A549 cells showed that two of the scFvs can significantly hamper the inhibition of the IFN-β-induced RIG-I signaling cascade mediated by EBOV VP35. CONCLUSION Five antibodies in scFv format recognize an active form of EBOV VP35 in ELISA, while one antibody also recognizes VP35 in WB. Two of these scFvs were also able to interfere with the intracellular activity of VP35 in a cell system in vitro. These findings suggest that such antibodies in scFv format might be employed to develop therapeutic molecules able to hamper EBOV infections.
Collapse
Affiliation(s)
- Michela Flego
- Istituto Superiore di Sanità (ISS), National Center for Global Health, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Aldo Frau
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SS554 09042 Monserrato, Cagliari, Italy
| | - Luisa Accardi
- Department of Infectious Diseases, Viral Hepatitis, Oncoviruses and Retroviruses (EVOR) unit, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Mallano
- Istituto Superiore di Sanità (ISS), National Center for Global Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandro Ascione
- Istituto Superiore di Sanità (ISS), National Center for Global Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Mara Gellini
- Istituto Superiore di Sanità (ISS), National Center for Global Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Elisa Fanunza
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SS554 09042 Monserrato, Cagliari, Italy
| | - Stefano Vella
- Istituto Superiore di Sanità (ISS), National Center for Global Health, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paola Di Bonito
- Department of Infectious Diseases, Viral Hepatitis, Oncoviruses and Retroviruses (EVOR) unit, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy.
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SS554 09042 Monserrato, Cagliari, Italy.
| |
Collapse
|
26
|
Di Palma F, Daino GL, Ramaswamy VK, Corona A, Frau A, Fanunza E, Vargiu AV, Tramontano E, Ruggerone P. Relevance of Ebola virus VP35 homo-dimerization on the type I interferon cascade inhibition. Antivir Chem Chemother 2019; 27:2040206619889220. [PMID: 31744306 PMCID: PMC6883671 DOI: 10.1177/2040206619889220] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/13/2019] [Indexed: 01/10/2023] Open
Abstract
Ebola virus high lethality relies on its ability to efficiently bypass the host innate antiviral response, which senses the viral dsRNA through the RIG-I receptor and induces type I interferon α/β production. In the bypassing action, the Ebola virus protein VP35 plays a pivotal role at multiple levels of the RIG-I cascade, masking the viral 5′-triphosphorylated dsRNA from RIG-I, and interacting with other cascade components. The VP35 type I interferon inhibition is exerted by the C-terminal domain, while the N-terminal domain, containing a coiled-coil region, is primarily required for oligomerization. However, mutations at key VP35 residues L90/93/107A (VP35-3m) in the coiled-coil region were reported to affect oligomerization and reduce type I interferon antagonism, indicating a possible but unclear role of homo-oligomerization on VP35 interaction with the RIG-I pathway components. In this work, we investigated the VP35 dimerization thermodynamics and its contribution to type I interferon antagonism by computational and biological methods. Focusing on the coiled-coil region, we combined coarse-grained and all-atom simulations on wild type VP35 and VP35-3m homo-dimerization. According to our results, wild type VP35 coiled-coil is able to self-assemble into dimers, while VP35-3m coiled-coil shows poor propensity to even dimerize. Free-energy calculations confirmed the key role of L90, L93 and L107 in stabilizing the coiled-coil homo-dimeric structure. In vitro type I interferon antagonism studies, using full-length wild type VP35 and VP35-3m, revealed that VP35 homo-dimerization is an essential preliminary step for dsRNA binding, which appears to be the main factor of the VP35 RIG-I cascade inhibition, while it is not essential to block the other steps.
Collapse
Affiliation(s)
- Francesco Di Palma
- Department of Physics, University of Cagliari, Cittadella
Universitaria, Monserrato, Italy
| | - Gian Luca Daino
- Department of Life and Environmental Sciences, University of
Cagliari, Cittadella Universitaria, Monserrato, Italy
| | | | - Angela Corona
- Department of Life and Environmental Sciences, University of
Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Aldo Frau
- Department of Life and Environmental Sciences, University of
Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Elisa Fanunza
- Department of Life and Environmental Sciences, University of
Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Attilio V Vargiu
- Department of Physics, University of Cagliari, Cittadella
Universitaria, Monserrato, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of
Cagliari, Cittadella Universitaria, Monserrato, Italy
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale
delle Ricerche (CNR), Monserrato, Italy
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, Cittadella
Universitaria, Monserrato, Italy
- Istituto Officina dei Materiali (CNR-IOM), UOS Cagliari SLACS,
Monserrato, Italy
| |
Collapse
|