1
|
Maryewski XA, Krasilnikov MS, Straková P, Holoubek J, Frčková T, Panina IS, Krylov NA, Gvozdev DA, Denisov VS, Semenov AN, Lotosh NY, Selishcheva AA, Chistov AA, Gulyak EL, Kozhemyakin GL, Korshun VA, Efremov RG, Ustinov AV, Růžek D, Eyer L, Alferova VA. Membrane-Active Singlet Oxygen Photogenerators as a Paradigm for Broad-Spectrum Antivirals: The Case of Halogenated (BOron)-DIPYrromethenes. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39772406 DOI: 10.1021/acsami.4c17482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Enveloped viruses, such as flaviviruses and coronaviruses, are pathogens of significant medical concern that cause severe infections in humans. Some photosensitizers are known to possess virucidal activity against enveloped viruses, targeting their lipid bilayer. Here we report a series of halogenated difluoroboron-dipyrromethene (BODIPYs) photosensitizers with strong virus-inactivating activity. Our structure-activity relationship analysis revealed that BODIPY scaffolds with a heavy halogen atom demonstrate significant efficacy against both tick-borne encephalitis virus (TBEV; Flaviviridae family) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2; Coronaviridae family) along with high singlet oxygen quantum yields. Moreover, select compounds also inactivated other enveloped viruses, such as herpes simplex virus type 1 and monkeypox virus. The nature and length of the alkyl side chain notably influenced the virus-inactivating activity of BODIPY molecules. Furthermore, molecular dynamics studies highlighted the critical importance of the positioning of the chromophore moiety within the lipid bilayer. As membrane-targeting photosensitizers, BODIPYs interact directly with virus particles, causing damage to the viral envelope membranes. Thus, TBEV pretreated with BODIPY was completely noninfective for lab mice. Consequently, BODIPY-based photosensitizers hold potential either as broad-spectrum virus-inactivating antivirals against a variety of phylogenetically unrelated enveloped viruses or as potent inactivators of viruses for the development of vaccines for preventing life-threatening emerging viral diseases.
Collapse
Affiliation(s)
- Xenia A Maryewski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maxim S Krasilnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Petra Straková
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic
- Laboratory of Clinical Immunology and Immunology of Infectious Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic
| | - Jiří Holoubek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Tereza Frčková
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Irina S Panina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Daniil A Gvozdev
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119991 Moscow, Russia
| | - Vladislav S Denisov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Alexey N Semenov
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119991 Moscow, Russia
- Dynamics of Fluids, Department of Experimental Physics, Saarland University, Campus E2 6, 66123 Saarbrücken, Germany
| | - Natalia Y Lotosh
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119991 Moscow, Russia
| | - Alla A Selishcheva
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119991 Moscow, Russia
| | - Alexey A Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Evgeny L Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Grigory L Kozhemyakin
- Research Institute for Systems Biology and Medicine, 18 Nauchny proezd, 117246 Moscow, Russia
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Alexey V Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Daniel Růžek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Luděk Eyer
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Vera A Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
2
|
Ulrich D, Hensel A, Classen N, Hafezi W, Sendker J, Kühn J. Aescin Inhibits Herpes simplex Virus Type 1 Induced Membrane Fusion. PLANTA MEDICA 2024; 90:1156-1166. [PMID: 39442532 DOI: 10.1055/a-2441-6570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Infections with Herpes simplex virus can cause severe ocular diseases and encephalitis. The present study aimed to investigate potential inhibitors of fusion between HSV-1 and the cellular membrane of the host cell. Fusion and entry of HSV-1 into the host cell is mimicked by a virus-free eukaryotic cell culture system by co-expression of the HSV-1 glycoproteins gD, gH, gL, and gB in presence of a gD receptor, resulting in excessive membrane fusion and polykaryocyte formation. A microscopic read-out was used for the screening of potential inhibitors, whereas luminometric quantification of cell-cell fusion was used in a reporter fusion assay. HSV-1 gB was tagged at its C-terminus with mCherry to express mCherry-gB in both assay systems for the visualization of the polykaryocyte formation. Reporter protein expression of SEAP was regulated by a Tet-On 3 G system. The saponin mixture aescin was identified as the specific inhibitor (IC50 7.4 µM, CC50 24.3 µM, SI 3.3) of membrane fusion. A plaque reduction assay on Vero cells reduced HSV-1 entry into cells and HSV-1 cell-to-cell spread significantly; 15 µM aescin decreased relative plaque counts to 41%, and 10 µM aescin resulted in a residual plaque size of 11% (HSV-1 17 syn+) and 2% (HSV-1 ANG path). Release of the HSV-1 progeny virus was reduced by one log step in the presence of 15 µM aescin. Virus particle integrity was mainly unaffected. Analytical investigation of aescin by UHPLC-MS revealed aescin IA and -IB and isoaescin IA and -IB as the main compounds with different functional activities. Aescin IA had the lowest IC50, the highest CC50, and an SI of > 4.6.
Collapse
Affiliation(s)
- Diana Ulrich
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Nica Classen
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Wali Hafezi
- Institute of Virology Münster (IVM), University of Münster, Münster, Germany
| | - Jandirk Sendker
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Joachim Kühn
- Institute of Virology Münster (IVM), University of Münster, Münster, Germany
| |
Collapse
|
3
|
Mengist HM, Denman P, Frost C, Sng JDJ, Logan S, Yarlagadda T, Spann KM, Barner L, Fairfull-Smith KE, Short KR, Boase NR. High-Throughput Synthesis and Evaluation of Antiviral Copolymers for Enveloped Respiratory Viruses. Biomacromolecules 2024; 25:7377-7391. [PMID: 39367828 DOI: 10.1021/acs.biomac.4c01049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
COVID-19 made apparent the devastating impact viral pandemics have had on global health and order. Development of broad-spectrum antivirals to provide early protection upon the inevitable emergence of new viral pandemics is critical. In this work, antiviral polymers are discovered using a combination of high-throughput polymer synthesis and antiviral screening, enabling diverse polymer compositions to be explored. Amphipathic polymers, with ionizable tertiary amine groups, are the most potent antivirals, effective against influenza virus and SARS-CoV-2, with minimal cytotoxicity. It is hypothesized that these polymers interact with the viral membrane as they showed no activity against a nonenveloped virus (rhinovirus). The switchable chemistry of the polymers during endosomal acidification was evaluated using lipid monolayers, indicating that a complex synergy between hydrophobicity and ionization drives polymer-membrane interactions. This new high-throughput methodology can be adapted to continue to engineer the potency of the lead candidates or develop antiviral polymers against other emerging viral classes.
Collapse
Affiliation(s)
| | - Paul Denman
- Centre for Materials Science, Queensland University of Technology, Brisbane 4000, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Charlotte Frost
- Centre for Materials Science, Queensland University of Technology, Brisbane 4000, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Julian D J Sng
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Saskia Logan
- Centre for Materials Science, Queensland University of Technology, Brisbane 4000, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane 4000, Australia
| | - Kirsten M Spann
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane 4000, Australia
| | - Leonie Barner
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Kathryn E Fairfull-Smith
- Centre for Materials Science, Queensland University of Technology, Brisbane 4000, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Nathan Rb Boase
- Centre for Materials Science, Queensland University of Technology, Brisbane 4000, Australia
- School of Chemistry and Physics, Queensland University of Technology, Brisbane 4000, Australia
| |
Collapse
|
4
|
Ostroumova OS, Efimova SS. Lipid-Centric Approaches in Combating Infectious Diseases: Antibacterials, Antifungals and Antivirals with Lipid-Associated Mechanisms of Action. Antibiotics (Basel) 2023; 12:1716. [PMID: 38136750 PMCID: PMC10741038 DOI: 10.3390/antibiotics12121716] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
One of the global challenges of the 21st century is the increase in mortality from infectious diseases against the backdrop of the spread of antibiotic-resistant pathogenic microorganisms. In this regard, it is worth targeting antibacterials towards the membranes of pathogens that are quite conservative and not amenable to elimination. This review is an attempt to critically analyze the possibilities of targeting antimicrobial agents towards enzymes involved in pathogen lipid biosynthesis or towards bacterial, fungal, and viral lipid membranes, to increase the permeability via pore formation and to modulate the membranes' properties in a manner that makes them incompatible with the pathogen's life cycle. This review discusses the advantages and disadvantages of each approach in the search for highly effective but nontoxic antimicrobial agents. Examples of compounds with a proven molecular mechanism of action are presented, and the types of the most promising pharmacophores for further research and the improvement of the characteristics of antibiotics are discussed. The strategies that pathogens use for survival in terms of modulating the lipid composition and physical properties of the membrane, achieving a balance between resistance to antibiotics and the ability to facilitate all necessary transport and signaling processes, are also considered.
Collapse
Affiliation(s)
- Olga S. Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia;
| | | |
Collapse
|
5
|
Mikhnovets IE, Holoubek J, Panina IS, Kotouček J, Gvozdev DA, Chumakov SP, Krasilnikov MS, Zhitlov MY, Gulyak EL, Chistov AA, Nikitin TD, Korshun VA, Efremov RG, Alferova VA, Růžek D, Eyer L, Ustinov AV. Alkyl Derivatives of Perylene Photosensitizing Antivirals: Towards Understanding the Influence of Lipophilicity. Int J Mol Sci 2023; 24:16483. [PMID: 38003673 PMCID: PMC10671050 DOI: 10.3390/ijms242216483] [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: 09/06/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Amphipathic perylene derivatives are broad-spectrum antivirals against enveloped viruses that act as fusion inhibitors in a light-dependent manner. The compounds target the lipid bilayer of the viral envelope using the lipophilic perylene moiety and photogenerating singlet oxygen, thereby causing damage to unsaturated lipids. Previous studies show that variation of the polar part of the molecule is important for antiviral activity. Here, we report modification of the lipophilic part of the molecule, perylene, by the introduction of 4-, 8-, and 12-carbon alkyls into position 9(10) of the perylene residue. Using Friedel-Crafts acylation and Wolff-Kishner reduction, three 3-acetyl-9(10)-alkylperylenes were synthesized from perylene and used to prepare 9 nucleoside and 12 non-nucleoside amphipathic derivatives. These compounds were characterized as fluorophores and singlet oxygen generators, as well as tested as antivirals against herpes virus-1 (HSV-1) and vesicular stomatitis virus (VSV), both known for causing superficial skin/mucosa lesions and thus serving as suitable candidates for photodynamic therapy. The results suggest that derivatives with a short alkyl chain (butyl) have strong antiviral activity, whereas the introduction of longer alkyl substituents (n = 8 and 12) to the perylenyethynyl scaffold results in a dramatic reduction of antiviral activity. This phenomenon is likely attributable to the increased lipophilicity of the compounds and their ability to form insoluble aggregates. Moreover, molecular dynamic studies revealed that alkylated perylene derivatives are predominately located closer to the middle of the bilayer compared to non-alkylated derivatives. The predicted probability of superficial positioning correlated with antiviral activity, suggesting that singlet oxygen generation is achieved in the subsurface layer of the membrane, where the perylene group is more accessible to dissolved oxygen.
Collapse
Affiliation(s)
- Igor E. Mikhnovets
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Jiří Holoubek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic (D.R.); (L.E.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Irina S. Panina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Jan Kotouček
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic;
| | - Daniil A. Gvozdev
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia;
| | - Stepan P. Chumakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Maxim S. Krasilnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Mikhail Y. Zhitlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Evgeny L. Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Timofei D. Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Roman G. Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Daniel Růžek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic (D.R.); (L.E.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Luděk Eyer
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic (D.R.); (L.E.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| |
Collapse
|
6
|
Straková P, Bednář P, Kotouček J, Holoubek J, Fořtová A, Svoboda P, Štefánik M, Huvarová I, Šimečková P, Mašek J, Gvozdev DA, Mikhnovets IE, Chistov AA, Nikitin TD, Krasilnikov MS, Ustinov AV, Alferova VA, Korshun VA, Růžek D, Eyer L. Antiviral activity of singlet oxygen-photogenerating perylene compounds against SARS-CoV-2: Interaction with the viral envelope and photodynamic virion inactivation. Virus Res 2023; 334:199158. [PMID: 37339718 PMCID: PMC10307035 DOI: 10.1016/j.virusres.2023.199158] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/09/2023] [Accepted: 06/18/2023] [Indexed: 06/22/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has prompted great interest in novel broad-spectrum antivirals, including perylene-related compounds. In the present study, we performed a structure-activity relationship analysis of a series of perylene derivatives, which comprised a large planar perylene residue, and structurally divergent polar groups connected to the perylene core by a rigid ethynyl or thiophene linker. Most of the tested compounds did not exhibit significant cytotoxicity towards multiple cell types susceptible to SARS-CoV-2 infection, and did not change the expressions of cellular stress-related genes under normal light conditions. These compounds showed nanomolar or sub-micromolar dose-dependent anti-SARS-CoV-2 activity, and also suppressed the in vitro replication of feline coronavirus (FCoV), also termed feline infectious peritonitis virus (FIPV). Perylene compounds exhibited high affinity for liposomal and cellular membranes, and efficiently intercalated into the envelopes of SARS-CoV-2 virions, thereby blocking the viral-cell fusion machinery. Furthermore, the studied compounds were demonstrated to be potent photosensitizers, generating reactive oxygen species (ROS), and their anti-SARS-CoV-2 activities were considerably enhanced after irradiation with blue light. Our results indicated that photosensitization is the major mechanism underlying the anti-SARS-CoV-2 activity of perylene derivatives, with these compounds completely losing their antiviral potency under red light. Overall, perylene-based compounds are broad-spectrum antivirals against multiple enveloped viruses, with antiviral action based on light-induced photochemical damage (ROS-mediated, likely singlet oxygen-mediated), causing impairment of viral membrane rheology.
Collapse
Affiliation(s)
- Petra Straková
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-370 05 České Budějovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Petr Bednář
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-370 05 České Budějovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic; Faculty of Science, University of South Bohemia, Ceske Budejovice, CZ-37005, Czech Republic
| | - Jan Kotouček
- Department of Pharmacology and Toxicology, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic
| | - Jiří Holoubek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-370 05 České Budějovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Andrea Fořtová
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Pavel Svoboda
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-370 05 České Budějovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic; Department of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, CZ-612 42 Brno, Czech Republic
| | - Michal Štefánik
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
| | - Ivana Huvarová
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic
| | - Pavlína Šimečková
- Department of Pharmacology and Toxicology, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic
| | - Josef Mašek
- Department of Pharmacology and Toxicology, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic
| | - Daniil A Gvozdev
- Department of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Igor E Mikhnovets
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Alexey A Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Timofei D Nikitin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Maxim S Krasilnikov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Alexey V Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Vera A Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Daniel Růžek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-370 05 České Budějovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Luděk Eyer
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, CZ-621 00 Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-370 05 České Budějovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic.
| |
Collapse
|
7
|
Mariewskaya KA, Gvozdev DA, Chistov AA, Straková P, Huvarová I, Svoboda P, Kotouček J, Ivanov NM, Krasilnikov MS, Zhitlov MY, Pak AM, Mikhnovets IE, Nikitin TD, Korshun VA, Alferova VA, Mašek J, Růžek D, Eyer L, Ustinov AV. Membrane-Targeting Perylenylethynylphenols Inactivate Medically Important Coronaviruses via the Singlet Oxygen Photogeneration Mechanism. Molecules 2023; 28:6278. [PMID: 37687107 PMCID: PMC10488391 DOI: 10.3390/molecules28176278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Perylenylethynyl derivatives have been recognized as broad-spectrum antivirals that target the lipid envelope of enveloped viruses. In this study, we present novel perylenylethynylphenols that exhibit nanomolar or submicromolar antiviral activity against Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) and feline infectious peritonitis virus (FIPV) in vitro. Perylenylethynylphenols incorporate into viral and cellular membranes and block the entry of the virus into the host cell. Furthermore, these compounds demonstrate an ability to generate singlet oxygen when exposed to visible light. The rate of singlet oxygen production is positively correlated with antiviral activity, confirming that the inhibition of fusion is primarily due to singlet-oxygen-induced damage to the viral envelope. The unique combination of a shape that affords affinity to the lipid bilayer and the capacity to generate singlet oxygen makes perylenylethynylphenols highly effective scaffolds against enveloped viruses. The anticoronaviral activity of perylenylethynylphenols is strictly light-dependent and disappears in the absence of daylight (under red light). Moreover, these compounds exhibit negligible cytotoxicity, highlighting their significant potential for further exploration of the precise antiviral mechanism and the broader scope and limitations of this compound class.
Collapse
Affiliation(s)
- Kseniya A. Mariewskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Daniil A. Gvozdev
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia;
| | - Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Petra Straková
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (P.S.); (I.H.); (P.S.); (D.R.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-625 00 Brno, Czech Republic
| | - Ivana Huvarová
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (P.S.); (I.H.); (P.S.); (D.R.)
| | - Pavel Svoboda
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (P.S.); (I.H.); (P.S.); (D.R.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-625 00 Brno, Czech Republic
- Department of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackého tř. 1946/1, CZ-612 42 Brno, Czech Republic
| | - Jan Kotouček
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (J.K.); (J.M.)
| | - Nikita M. Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Maxim S. Krasilnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Mikhail Y. Zhitlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Alexandra M. Pak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Igor E. Mikhnovets
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Timofei D. Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| | - Josef Mašek
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (J.K.); (J.M.)
| | - Daniel Růžek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (P.S.); (I.H.); (P.S.); (D.R.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-625 00 Brno, Czech Republic
| | - Luděk Eyer
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic; (P.S.); (I.H.); (P.S.); (D.R.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-625 00 Brno, Czech Republic
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.A.C.); (N.M.I.); (M.S.K.); (M.Y.Z.); (A.M.P.); (I.E.M.); (T.D.N.); (V.A.A.); (A.V.U.)
| |
Collapse
|
8
|
Kamzeeva PN, Aralov AV, Alferova VA, Korshun VA. Recent Advances in Molecular Mechanisms of Nucleoside Antivirals. Curr Issues Mol Biol 2023; 45:6851-6879. [PMID: 37623252 PMCID: PMC10453654 DOI: 10.3390/cimb45080433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
The search for new drugs has been greatly accelerated by the emergence of new viruses and drug-resistant strains of known pathogens. Nucleoside analogues (NAs) are a prospective class of antivirals due to known safety profiles, which are important for rapid repurposing in the fight against emerging pathogens. Recent improvements in research methods have revealed new unexpected details in the mechanisms of action of NAs that can pave the way for new approaches for the further development of effective drugs. This review accounts advanced techniques in viral polymerase targeting, new viral and host enzyme targeting approaches, and prodrug-based strategies for the development of antiviral NAs.
Collapse
Affiliation(s)
| | | | | | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (P.N.K.); (A.V.A.); (V.A.A.)
| |
Collapse
|
9
|
Nowakowski M, Wiśniewska-Becker A, Czapla-Masztafiak J, Szlachetko J, Budziak A, Polańska Ż, Pietralik-Molińska Z, Kozak M, Kwiatek WM. Cr(vi) permanently binds to the lipid bilayer in an inverted hexagonal phase throughout the reduction process. RSC Adv 2023; 13:18854-18863. [PMID: 37350866 PMCID: PMC10282592 DOI: 10.1039/d2ra07851a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/16/2023] [Indexed: 06/24/2023] Open
Abstract
Cr(vi) is a harmful, carcinogenic agent with a high permeability rate throughout the lipid membranes. In an intracellular environment and during interactions with cellular membranes, it undergoes an instant reduction to lower oxidation states throughout radical states, recognized as the most dangerous factor for cells. The cellular membrane is the most visible cellular organelle in the interior and exterior of a cell. In this study, liposomes and non-lamellar inverted hexagonal phase lipid structures based on phosphoethanolamine (PE) were used as model cellular bilayers because of their simple composition, preparation procedure, and the many other properties of natural systems. The lipid membranes were subjected to 0.075 mM Cr(vi) for 15 min, after which the Cr content was removed via dialysis. This way, the remaining Cr content could be studied qualitatively and quantitatively. Using the combined XRF/XAS/EPR approach, we revealed that some Cr content (Cr(iii) and Cr(vi)) was still present in the samples even after long-term dialysis at a temperature significantly above the phase transition for the chosen liposome. The amount of bound Cr increased with increasing PE and -C[double bond, length as m-dash]C- bond content in lipid mixtures. Internal membrane order decreased in less fluid membranes, while in more liquified ones, internal order was only slightly changed after subjecting them to the Cr(vi) agent. The results suggest that the inverted hexagonal phase of lipid structures is much more sensitive to oxidation than the lamellar lipid phase, which can play an important role in the strong cytotoxicity of Cr(vi).
Collapse
Affiliation(s)
- Michal Nowakowski
- Institute of Nuclear Physics Polish Academy of Sciences PL-31342 Krakow Poland
| | - Anna Wiśniewska-Becker
- Jagiellonian University in Krakow, Faculty of Biochemistry, Biophysics and Biotechnology PL-30387 Krakow Poland
| | | | - Jakub Szlachetko
- Solaris National Synchrotron Radiation Centre, Jagiellonian University 30-392 Krakow Poland
| | - Andrzej Budziak
- AGH University of Science and Technology, Faculty of Energy and Fuels Krakow Poland
| | - Żaneta Polańska
- Adam Mickiewicz University in Poznan, Faculty of Physics PL-61-614 Poznan Poland
| | | | - Maciej Kozak
- Adam Mickiewicz University in Poznan, Faculty of Physics PL-61-614 Poznan Poland
| | - Wojciech M Kwiatek
- Institute of Nuclear Physics Polish Academy of Sciences PL-31342 Krakow Poland
| |
Collapse
|
10
|
Eyer L, Seley-Radtke K, Ruzek D. New directions in the experimental therapy of tick-borne encephalitis. Antiviral Res 2023; 210:105504. [PMID: 36574904 DOI: 10.1016/j.antiviral.2022.105504] [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: 11/17/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Tick-borne encephalitis (TBE) is a potentially fatal disease common in much of Europe and Asia. There is no specific therapy for the treatment of TBE patients. However, several efforts are being made to develop small molecules that specifically interfere with the life cycle of TBE virus. In particular, recently various nucleoside analogues that can inhibit the viral replicase, such as the RNA-dependent RNA polymerase or viral methyltransferases, have been explored. In addition, human or chimeric (i.e., structural chimeras that combine mouse variable domains with human constant domains) monoclonal antibodies with promising potential for post-exposure prophylaxis or early therapy have been developed. This review summarizes the latest directions and experimental approaches that may be used to combat TBE in humans.
Collapse
Affiliation(s)
- Ludek Eyer
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Katherine Seley-Radtke
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Daniel Ruzek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
11
|
Chistov AA, Chumakov SP, Mikhnovets IE, Nikitin TD, Slesarchuk NA, Uvarova VI, Rubekina AA, Nikolaeva YV, Radchenko EV, Khvatov EV, Orlov AA, Frolenko VS, Sukhorukov MV, Kolpakova ES, Shustova EY, Galochkina AV, Streshnev PP, Osipov EM, Sapozhnikova KA, Moiseenko AV, Brylev VA, Proskurin GV, Dokukin YS, Kutyakov SV, Aralov AV, Korshun VA, Strelkov SV, Palyulin VA, Ishmukhametov AA, Shirshin EA, Osolodkin DI, Shtro AA, Kozlovskaya LI, Alferova VA, Ustinov AV. 5-(Perylen-3-ylethynyl)uracil as an antiviral scaffold: Potent suppression of enveloped virus reproduction by 3-methyl derivatives in vitro. Antiviral Res 2023; 209:105508. [PMID: 36581049 DOI: 10.1016/j.antiviral.2022.105508] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Amphipathic nucleoside and non-nucleoside derivatives of pentacyclic aromatic hydrocarbon perylene are known as potent non-cytotoxic broad-spectrum antivirals. Here we report 3-methyl-5-(perylen-3-ylethynyl)-uracil-1-acetic acid and its amides, a new series of compounds based on a 5-(perylen-3-ylethynyl)-uracil scaffold. The compounds demonstrate pronounced in vitro activity against arthropod-borne viruses, namely tick-borne encephalitis virus (TBEV) and yellow fever virus (YFV), in plaque reduction assays with EC50 values below 1.9 and 1.3 nM, respectively, and Chikungunya virus (CHIKV) in cytopathic effect inhibition test with EC50 values below 3.2 μM. The compounds are active against respiratory viruses as well: severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) in cytopathic effect inhibition test and influenza A virus (IAV) in virus titer reduction experiments are inhibited - EC50 values below 51 nM and 2.2 μM, respectively. The activity stems from the presence of a hydrophobic perylene core, and all of the synthesized compounds exhibit comparable 1O2 generation rates. Nonetheless, activity can vary by orders of magnitude depending on the hydrophilic part of the molecule, suggesting a complex mode of action. A time-of-addition experiment and fluorescent imaging indicate that the compounds inhibit viral fusion in a dose-dependent manner. The localization of the compound in the lipid bilayers and visible damage to the viral envelope suggest the membrane as the primary target. Dramatic reduction of antiviral activity with limited irradiation or under treatment with antioxidants further cements the idea of photoinduced ROS-mediated viral envelope damage being the mode of antiviral action.
Collapse
Affiliation(s)
- Alexey A Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Stepan P Chumakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Igor E Mikhnovets
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Timofei D Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nikita A Slesarchuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Victoria I Uvarova
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia
| | - Anna A Rubekina
- Department of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Yulia V Nikolaeva
- Smorodintsev Research Institute of Influenza, St. Petersburg, 197376, Russia
| | - Eugene V Radchenko
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Evgeny V Khvatov
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia
| | - Alexey A Orlov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia; FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia; Skolkovo Institute of Science and Technology, 143026, Moscow Region, Russia
| | - Vasilisa S Frolenko
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov Moscow State Medical University, Moscow, 119991, Russia
| | - Maksim V Sukhorukov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia; FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia
| | - Ekaterina S Kolpakova
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia
| | - Elena Y Shustova
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia
| | | | - Philipp P Streshnev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Eugene M Osipov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000, Leuven, Belgium
| | | | | | - Vladimir A Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia; Lumiprobe RUS Ltd., Moscow, 121351, Russia
| | - Gleb V Proskurin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Yuri S Dokukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Sergey V Kutyakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Andrey V Aralov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Sergei V Strelkov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000, Leuven, Belgium
| | - Vladimir A Palyulin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Aydar A Ishmukhametov
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov Moscow State Medical University, Moscow, 119991, Russia
| | - Evgeny A Shirshin
- Department of Physics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Dmitry I Osolodkin
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov Moscow State Medical University, Moscow, 119991, Russia
| | - Anna A Shtro
- Smorodintsev Research Institute of Influenza, St. Petersburg, 197376, Russia
| | - Liubov I Kozlovskaya
- FSASI "Chumakov FSC R&D IBP RAS" (Institute of Poliomyelitis), Moscow, 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov Moscow State Medical University, Moscow, 119991, Russia.
| | - Vera A Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia.
| | - Alexey V Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia; Lumiprobe RUS Ltd., Moscow, 121351, Russia.
| |
Collapse
|
12
|
Epand RM. The scientific adventures of Richard Epand. Biophys Chem 2023; 292:106931. [PMID: 36434860 DOI: 10.1016/j.bpc.2022.106931] [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: 11/01/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
This essay summarizes the many areas of science that my career has contributed to. It attempts to highlight some of the innovative concepts that developed from this work. The discussion encompasses studies I undertook from graduate school to the present but it will not attempt to be comprehensive. I apologize to individuals whose work I omitted. Because of space I cannot acknowledge all the contributions from other individuals that made these achievements possible.
Collapse
Affiliation(s)
- Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Health Sciences Centre, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada.
| |
Collapse
|
13
|
Added Value of Biophysics to Study Lipid-Driven Biological Processes: The Case of Surfactins, a Class of Natural Amphiphile Molecules. Int J Mol Sci 2022; 23:ijms232213831. [PMID: 36430318 PMCID: PMC9693386 DOI: 10.3390/ijms232213831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
The role of membrane lipids is increasingly claimed to explain biological activities of natural amphiphile molecules. To decipher this role, biophysical studies with biomimetic membrane models are often helpful to obtain insights at the molecular and atomic levels. In this review, the added value of biophysics to study lipid-driven biological processes is illustrated using the case of surfactins, a class of natural lipopeptides produced by Bacillus sp. showing a broad range of biological activities. The mechanism of interaction of surfactins with biomimetic models showed to be dependent on the surfactins-to-lipid ratio with action as membrane disturber without membrane lysis at low and intermediate ratios and a membrane permeabilizing effect at higher ratios. These two mechanisms are relevant to explain surfactins' biological activities occurring without membrane lysis, such as their antiviral and plant immunity-eliciting activities, and the one involving cell lysis, such as their antibacterial and hemolytic activities. In both biological and biophysical studies, influence of surfactin structure and membrane lipids on the mechanisms was observed with a similar trend. Hence, biomimetic models represent interesting tools to elucidate the biological mechanisms targeting membrane lipids and can contribute to the development of new molecules for pharmaceutical or agronomic applications.
Collapse
|
14
|
Rubekina AA, Kamzeeva PN, Alferova VA, Shustova EY, Kolpakova ES, Yakovchuk EV, Karpova EV, Borodulina MO, Belyaev ES, Khrulev AA, Korshun VA, Shirshin EA, Kozlovskaya LI, Aralov AV. Hydrophobic Rose Bengal Derivatives Exhibit Submicromolar-to-Subnanomolar Activity against Enveloped Viruses. Biomolecules 2022; 12:1609. [PMID: 36358961 PMCID: PMC9687286 DOI: 10.3390/biom12111609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 09/10/2023] Open
Abstract
Rose Bengal (RB) is an anionic xanthene dye with multiple useful biological features, including photosensitization properties. RB was studied extensively as a photosensitizer, mostly for antibacterial and antitumor photodynamic therapy (PDT). The application of RB to virus inactivation is rather understudied, and no RB derivatives have been developed as antivirals. In this work, we used a synthetic approach based on a successful design of photosensitizing antivirals to produce RB derivatives for virus photoinactivation. A series of n-alkyl-substituted RB derivatives was synthesized and evaluated as antiviral photosensitizers. The compounds exhibited similar 1O2 generation rate and efficiency, but drastically different activities against SARS-CoV-2, CHIKV, and HIV; with comparable cytotoxicity for different cell lines. Submicromolar-to-subnanomolar activities and high selectivity indices were detected for compounds with C4-6 alkyl (SARS-CoV-2) and C6-8 alkyl (CHIKV) chains. Spectrophotometric assessment demonstrates low aqueous solubility for C8-10 congeners and a significant aggregation tendency for the C12 derivative, possibly influencing its antiviral efficacy. Initial evaluation of the synthesized compounds makes them promising for further study as viral inactivators for vaccine preparations.
Collapse
Affiliation(s)
- Anna A. Rubekina
- Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskie Gory, 119234 Moscow, Russia
| | - Polina N. Kamzeeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Gause Institute of New Antibiotics, Russian Academy of Sciences, Bolshaya Pirogovskaya 11, 119021 Moscow, Russia
| | - Elena Yu. Shustova
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Ekaterina S. Kolpakova
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Elizaveta V. Yakovchuk
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Evgenia V. Karpova
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Maria O. Borodulina
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Evgeny S. Belyaev
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Science, 119071 Moscow, Russia
| | - Alexei A. Khrulev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Evgeny A. Shirshin
- Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskie Gory, 119234 Moscow, Russia
- Laboratory of Clinical Biophotonics, Biomedical Science and Technology Park, I.M. Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia
| | - Liubov I. Kozlovskaya
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia
| | - Andrey V. Aralov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
15
|
On the Sensitivity of the Virion Envelope to Lipid Peroxidation. Microbiol Spectr 2022; 10:e0300922. [PMID: 36125312 PMCID: PMC9603946 DOI: 10.1128/spectrum.03009-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Emerging viruses are a public health threat best managed with broad spectrum antivirals. Common viral structures, like capsids or virion envelopes, have been proposed as targets for broadly active antiviral drugs. For example, a number of lipoperoxidators have been proposed to preferentially affect viral infectivity by targeting metabolically inactive enveloped virions while sparing metabolically active cells. However, this presumed preferential virion sensitivity to lipoperoxidation remains untested. To test whether virions are indeed more sensitive to lipoperoxidation than are cells, we analyzed the effects of two classic generic lipoperoxidators: lipophilic 2,2'-azobis(2,4-dimethylvaleronitrile) (AMVN) and hydrophilic 2,2'-azobis(2-methylpropionamidine) dihydrochloride (AAPH) on Vero and human foreskin fibroblasts (HFF) cell viability, HSV-1 plaquing efficiency, and virion and cell lipoperoxidation. Cells or virions were incubated with the lipoperoxidators at 37°C for 2 h or incubated in atmospheric O2, and dose responses (half maximal cytotoxic and effective concentration [CC50 and EC50]) were evaluated by three or four parameter regression. The HSV-1 virions were slightly more sensitive to lipoperoxidators than were the cells (selectivity index [SI], 3.3 to 7.4). The effects of the lipophilic AMVN on both cell and virion viability directly correlated with the extent of membrane lipoperoxidation as evaluated by two different probes, C11-Bodipy and liperfluo. Moreover, the hydrophilic AAPH-induced virion inactivation at lower concentrations than did lipoperoxidation. Known lipoperoxidators inhibit infectivity via lipoperoxidation-independent mechanisms. Antioxidants protected against a loss of viral infectivity by less than 5-fold. Carrier bovine serum albumin (BSA) protected against both peroxidators to a similar extent when present together with the lipoperoxidating agents, suggesting that BSA quenches them as expected. Virions incubated in atmospheric oxidative conditions suffered losses of infectivity that were similar to those of chemically peroxidated virions, and they were protected by water soluble vitamin C and BSA with no evident lipoperoxidation, indicating predominant peroxidative damage to nonlipid virion components. Thus, lipoperoxidation is not a mechanism by which to specifically inhibit the infectivity of enveloped viruses, and the effects of known lipoperoxidators on virion infectivity are not solely mediated by lipoperoxidation. IMPORTANCE Small molecules that induce lipoperoxidation have been proposed repeatedly as potential antiviral drugs based on a presumed unique sensitivity of virions to this type of damage. Several small molecules that inactivate virions without affecting cells have been proposed to act primarily by inducing lipoperoxidation. However, the preferential sensitivity of virions to lipoperoxidators had not been experimentally evaluated. Using two of the best characterized small molecule lipoperoxidators, which are widely considered to be the prototypical water soluble and liposoluble lipoperoxidators, we show that lipoperoxidators have no preference for virions over cells. Moreover, they also inactivate virions by mechanisms other than the induction of lipoperoxidation. Therefore, the general induction of lipoperoxidation is not a path by which to develop antivirals. Moreover, molecules with specific antiviral activity which are not cytotoxic and have no preference to localize to virions over cells are unlikely to act primarily by inducing lipoperoxidation.
Collapse
|
16
|
Shtro AA, Garshinina AV, Alferova VA, Kamzeeva PN, Volok VP, Kolpakova ES, Nikitin TD, Chistov AA, Belyaev ES, Korshun VA, Kozlovskaya LI, Aralov AV. Cationic Perylene Antivirals with Aqueous Solubility for Studies In Vivo. Pharmaceuticals (Basel) 2022; 15:1178. [PMID: 36297288 PMCID: PMC9610897 DOI: 10.3390/ph15101178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 08/26/2023] Open
Abstract
Perylene-based compounds are attracting significant attention due to their high broad-spectrum antiviral activity against enveloped viruses. Despite unambiguous results of in vitro studies and high selectivity index, the poor water solubility of these compounds prevented in vivo evaluation of their antiviral properties. In this work, we synthesized a series of compounds with a perylene pharmacophore bearing positively charged substituents to improve the aqueous solubility of this unique type of antivirals. Three types of charged groups were introduced: (1) quaternary morpholinium salts (3a-b); (2) a 2'-O-l-valinyl-uridine hydrochloride residue (8), and (3) a 3-methylbenzothiazolium cation (10). The synthesized compounds were evaluated based both on antiviral properties in vitro (CHIKV, SARS-CoV-2, and IAV) and on solubility in aqueous media. Compound 10 has the greatest aqueous solubility, making it preferable for pre-evaluation by intragastrical administration in a mouse model of lethal influenza pneumonia. The results indicate that the introduction of a positively charged group is a viable strategy for the design of drug candidates with a perylene scaffold for in vivo studies.
Collapse
Affiliation(s)
- Anna A. Shtro
- Smorodintsev Research Institute of Influenza, 197376 Saint Petersburg, Russia
| | | | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Gause Institute of New Antibiotics, 119021 Moscow, Russia
| | - Polina N. Kamzeeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Viktor P. Volok
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Ekaterina S. Kolpakova
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
| | - Timofei D. Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Evgeny S. Belyaev
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Science, 119071 Moscow, Russia
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Liubov I. Kozlovskaya
- Chumakov Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov Moscow State Medical University, 119991 Moscow, Russia
| | - Andrey V. Aralov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
17
|
Perylene as a controversial antiviral scaffold. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2022. [DOI: 10.1016/bs.armc.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
18
|
Entry Inhibitors of Hepatitis C Virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:207-222. [DOI: 10.1007/978-981-16-8702-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Liu Y, Zhang X, Qi W, Yang Y, Liu Z, An T, Wu X, Chen J. Prevention and Control Strategies of African Swine Fever and Progress on Pig Farm Repopulation in China. Viruses 2021; 13:2552. [PMID: 34960821 PMCID: PMC8704102 DOI: 10.3390/v13122552] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 01/26/2023] Open
Abstract
African swine fever (ASF) is a devastating disease in domestic and wild pigs. Since the first outbreak of ASF in August 2018 in China, the disease has spread throughout the country with an unprecedented speed, causing heavy losses to the pig and related industries. As a result, strategies for managing the disease are urgently needed. This paper summarizes the important aspects of three key elements about African swine fever virus (ASFV) transmission, including the sources of infection, transmission routes, and susceptible animals. It overviews the relevant prevention and control strategies, focusing on the research progress of ASFV vaccines, anti-ASFV drugs, ASFV-resistant pigs, efficient disinfection, and pig farm biosecurity. We then reviewed the key technical points concerning pig farm repopulation, which is critical to the pork industry. We hope to not only provide a theoretical basis but also practical strategies for effective dealing with the ASF epidemic and restoration of pig production.
Collapse
Affiliation(s)
- Yuanjia Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.L.); (Z.L.)
| | - Xinheng Zhang
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.W.)
| | - Wenbao Qi
- Research Center for African Swine Fever Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China;
| | - Yaozhi Yang
- Heilongjiang Dabeinong Agriculture and Animal Husbandry Food Company Limited, Harbin 150028, China;
| | - Zexin Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.L.); (Z.L.)
| | - Tongqing An
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China;
| | - Xiuhong Wu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.W.)
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.L.); (Z.L.)
| |
Collapse
|
20
|
Gentili V, Turrin G, Marchetti P, Rizzo S, Schiuma G, Beltrami S, Cristofori V, Illuminati D, Compagnin G, Trapella C, Rizzo R, Bortolotti D, Fantinati A. Synthesis and biological evaluation of novel rhodanine-based structures with antiviral activity towards HHV-6 virus. Bioorg Chem 2021; 119:105518. [PMID: 34861628 DOI: 10.1016/j.bioorg.2021.105518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/04/2021] [Accepted: 11/23/2021] [Indexed: 11/02/2022]
Abstract
An increased awareness of diseases associated with Human herpesvirus 6 (HHV-6) infection or reactivation has resulted in a growing interest in the evaluation of the best treatment options available for the clinical management of HHV-6 disease. However, no compound has yet been approved exclusively for HHV-6 infection treatment. For this reason, the identification of anti-HHV6 compounds provides a valuable opportunity for developing efficient antiviral therapies. A possible target for antiviral drugs is the virus-cell fusion step. In this study, we synthetized potential fusion intermediates inhibitors based on the rhodanine structure. The obtained derivatives were tested for cytotoxicity and for antiviral activity in human cells infected with HHV6. Level of infection was monitored by viral DNA quantification at different time points up to 7 days post infection. Among the synthetized derivatives, 9e showed a significative inhibitory effect on viral replication that lasted over 7 days, probably attributable to the particular combination of hydrophilic and hydrophobic substituents to the rhodanine moiety. Our results support the use of these amphipathic fusion inhibitors for the treatment of HHV-6 infections.
Collapse
Affiliation(s)
- Valentina Gentili
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Giulia Turrin
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Paolo Marchetti
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Sabrina Rizzo
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Giovanna Schiuma
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Silvia Beltrami
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Virginia Cristofori
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Davide Illuminati
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Greta Compagnin
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Claudio Trapella
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy.
| | - Roberta Rizzo
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy.
| | - Daria Bortolotti
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| | - Anna Fantinati
- University of Ferrara, Department of Chemical, Pharmaceutical and Agricultural Sciences, Via Fossato di Mortara, 17, 44121 Ferrara, Italy
| |
Collapse
|
21
|
Mariewskaya KA, Tyurin AP, Chistov AA, Korshun VA, Alferova VA, Ustinov AV. Photosensitizing Antivirals. Molecules 2021; 26:3971. [PMID: 34209713 PMCID: PMC8271894 DOI: 10.3390/molecules26133971] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/22/2021] [Accepted: 06/27/2021] [Indexed: 12/23/2022] Open
Abstract
Antiviral action of various photosensitizers is already summarized in several comprehensive reviews, and various mechanisms have been proposed for it. However, a critical consideration of the matter of the area is complicated, since the exact mechanisms are very difficult to explore and clarify, and most publications are of an empirical and "phenomenological" nature, reporting a dependence of the antiviral action on illumination, or a correlation of activity with the photophysical properties of the substances. Of particular interest is substance-assisted photogeneration of highly reactive singlet oxygen (1O2). The damaging action of 1O2 on the lipids of the viral envelope can probably lead to a loss of the ability of the lipid bilayer of enveloped viruses to fuse with the lipid membrane of the host cell. Thus, lipid bilayer-affine 1O2 photosensitizers have prospects as broad-spectrum antivirals against enveloped viruses. In this short review, we want to point out the main types of antiviral photosensitizers with potential affinity to the lipid bilayer and summarize the data on new compounds over the past three years. Further understanding of the data in the field will spur a targeted search for substances with antiviral activity against enveloped viruses among photosensitizers able to bind to the lipid membranes.
Collapse
Affiliation(s)
- Kseniya A. Mariewskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.P.T.); (A.A.C.); (V.A.K.)
- Higher Chemical College of the Russian Academy of Sciences, Mendeleev University of Chemical Technology, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Anton P. Tyurin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.P.T.); (A.A.C.); (V.A.K.)
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia
| | - Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.P.T.); (A.A.C.); (V.A.K.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.P.T.); (A.A.C.); (V.A.K.)
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.P.T.); (A.A.C.); (V.A.K.)
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.M.); (A.P.T.); (A.A.C.); (V.A.K.)
| |
Collapse
|
22
|
Lu S, Pan X, Chen D, Xie X, Wu Y, Shang W, Jiang X, Sun Y, Fan S, He J. Broad-spectrum antivirals of protoporphyrins inhibit the entry of highly pathogenic emerging viruses. Bioorg Chem 2021; 107:104619. [PMID: 33450541 PMCID: PMC7784547 DOI: 10.1016/j.bioorg.2020.104619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/18/2020] [Accepted: 12/27/2020] [Indexed: 12/19/2022]
Abstract
PPIX possesses broad antiviral activities in vitro against a panel of enveloped viruses. PPIX interacts with the lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. PPIX shows the antiviral effect in vivo by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus.
Severe emerging and re-emerging viral infections such as Lassa fever, Avian influenza (AI), and COVID-19 caused by SARS-CoV-2 urgently call for new strategies for the development of broad-spectrum antivirals targeting conserved components in the virus life cycle. Viral lipids are essential components, and viral-cell membrane fusion is the required entry step for most unrelated enveloped viruses. In this paper, we identified a porphyrin derivative of protoporphyrin IX (PPIX) that showed broad antiviral activities in vitro against a panel of enveloped pathogenic viruses including Lassa virus (LASV), Machupo virus (MACV), and SARS-CoV-2 as well as various subtypes of influenza A viral strains with IC50 values ranging from 0.91 ± 0.25 μM to 1.88 ± 0.34 μM. A mechanistic study using influenza A/Puerto Rico/8/34 (H1N1) as a testing strain showed that PPIX inhibits the infection in the early stage of virus entry through biophysically interacting with the hydrophobic lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. In addition, the preliminary antiviral activities of PPIX were further assessed by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus. The results showed that compared with the control group without drug treatment, the survival rate and mean survival time of the mice treated with PPIX were apparently prolonged. These data encourage us to conduct further investigations using PPIX as a lead compound for the rational design of lipid-targeting antivirals for the treatment of infection with enveloped viruses.
Collapse
Affiliation(s)
- Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Daiwei Chen
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xi Xie
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiaming Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Yuan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Sheng Fan
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China.
| |
Collapse
|
23
|
Dadhich R, Kapoor S. Various Facets of Pathogenic Lipids in Infectious Diseases: Exploring Virulent Lipid-Host Interactome and Their Druggability. J Membr Biol 2020; 253:399-423. [PMID: 32833058 PMCID: PMC7443855 DOI: 10.1007/s00232-020-00135-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Lipids form an integral, structural, and functional part of all life forms. They play a significant role in various cellular processes such as membrane fusion, fission, endocytosis, protein trafficking, and protein functions. Interestingly, recent studies have revealed their more impactful and critical involvement in infectious diseases, starting with the manipulation of the host membrane to facilitate pathogenic entry. Thereafter, pathogens recruit specific host lipids for the maintenance of favorable intracellular niche to augment their survival and proliferation. In this review, we showcase the lipid-mediated host pathogen interplay in context of life-threatening viral and bacterial diseases including the recent SARS-CoV-2 infection. We evaluate the emergent lipid-centric approaches adopted by these pathogens, while delineating the alterations in the composition and organization of the cell membrane within the host, as well as the pathogen. Lastly, crucial nexus points in their interaction landscape for therapeutic interventions are identified. Lipids act as critical determinants of bacterial and viral pathogenesis by altering the host cell membrane structure and functions.
Collapse
Affiliation(s)
- Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India.
- Wadhwani Research Centre for Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
24
|
Nikolayeva YV, Ulashchik EA, Chekerda EV, Galochkina AV, Slesarchuk NA, Chistov AA, Nikitin TD, Korshun VA, Shmanai VV, Ustinov AV, Shtro AA. 5-(Perylen-3-ylethynyl)uracil Derivatives Inhibit Reproduction of Respiratory Viruses. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020; 46:315-320. [PMID: 32834709 PMCID: PMC7305479 DOI: 10.1134/s1068162020030139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 01/18/2023]
Abstract
In this work, we describe the synthesis of 5-(perylen-3-ylethynyl)uridine and its ability to effectively inhibit the replication of respiratory disease pathogens in cell culture, namely: influenza A virus (IVA); type 3 parainfluenza virus (PIV-3); and human respiratory syncytial virus (RSV). Related known compounds were also analyzed: 5-(perylen-3-ylethynyl)-2'-deoxy-uridine; 5-(perylen-3-ylethynyl)-arabino-uridine; and 1-carboxymethyl-3-pivaloyloxymethyl-5-(perylen-3-ylethynyl)uracil.
Collapse
Affiliation(s)
- Y. V. Nikolayeva
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| | - E. A. Ulashchik
- Institute of Physical Organic Chemistry of the NAS Belarus, 220072 Minsk, Belarus
| | - E. V. Chekerda
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| | - A. V. Galochkina
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| | - N. A. Slesarchuk
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Chemistry, Moscow State University, 119991 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
| | - A. A. Chistov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
| | - T. D. Nikitin
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Chemistry, Moscow State University, 119991 Moscow, Russia
| | - V. A. Korshun
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
- Gause Institute of New Antibiotics, 119021 Moscow, Russia
| | - V. V. Shmanai
- Institute of Physical Organic Chemistry of the NAS Belarus, 220072 Minsk, Belarus
| | - A. V. Ustinov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
| | - A. A. Shtro
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| |
Collapse
|
25
|
Chen D, Lu S, Yang G, Pan X, Fan S, Xie X, Chen Q, Li F, Li Z, Wu S, He J. The seafood Musculus senhousei shows anti-influenza A virus activity by targeting virion envelope lipids. Biochem Pharmacol 2020; 177:113982. [PMID: 32305436 PMCID: PMC7162792 DOI: 10.1016/j.bcp.2020.113982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/14/2020] [Indexed: 12/17/2022]
Abstract
Pyropheophorbide a (PPa) was isolated from the seafood of M. senhousei. PPa shows a potent activity against a broad panel of influenza A viral strains. The mechanism of PPa is to block the entry of virus in the early stage of infection. The target of PPa may be that of lipid bilayer of the enveloped viruses.
Marine environments are known to be a new source of structurally diverse bioactive molecules. In this paper, we identified a porphyrin derivative of Pyropheophorbide a (PPa) from the mussel Musculus senhousei (M. senhousei) that showed broad anti-influenza A virus activity in vitro against a panel of influenza A viral strains. The analysis of the mechanism of action indicated that PPa functions in the early stage of virus infection by interacting with the lipid bilayer of the virion, resulting in an alteration of membrane-associated functions, thereby blocking the entry of enveloped viruses into host cells. In addition, the anti-influenza A virus activity of PPa was further assessed in mice infected with the influenza A virus. The survival rate and mean survival time of mice were apparently prolonged compared with the control group which was not treated with the drug. Therefore, PPa and its derivatives may represent lead compounds for controlling influenza A virus infection.
Collapse
Affiliation(s)
- Daiwei Chen
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Guang Yang
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xiaoyan Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Sheng Fan
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Xi Xie
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Qi Chen
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Fangfang Li
- Guangdong Provincial Key Laboratory of Emergency Test for Dangerous Chemicals, China National Analytical Center, Guangzhou 510070, PR China
| | - Zhonghuang Li
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shaohua Wu
- Key Laboratory for Microbial Resources of the Ministry of Education, Yunnan Institute of Microbiology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China.
| |
Collapse
|
26
|
Slesarchuk NA, Khvatov EV, Chistov AA, Proskurin GV, Nikitin TD, Lazarevich AI, Ulanovskaya AA, Ulashchik EA, Orlov AA, Jegorov AV, Ustinov AV, Tyurin AP, Shmanai VV, Ishmukhametov AA, Korshun VA, Osolodkin DI, Kozlovskaya LI, Aralov AV. Simplistic perylene-related compounds as inhibitors of tick-borne encephalitis virus reproduction. Bioorg Med Chem Lett 2020; 30:127100. [PMID: 32199731 DOI: 10.1016/j.bmcl.2020.127100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/28/2020] [Accepted: 03/07/2020] [Indexed: 12/12/2022]
Abstract
Rigid amphipathic fusion inhibitors are potent broad-spectrum antivirals based on the perylene scaffold, usually decorated with a hydrophilic group linked via ethynyl or triazole. We have sequentially simplified these structures by removing sugar moiety, then converting uridine to aniline, then moving to perylenylthiophenecarboxylic acids and to perylenylcarboxylic acid. All these polyaromatic compounds, as well as antibiotic heliomycin, still showed pronounced activity against tick-borne encephalitis virus (TBEV) with limited toxicity in porcine embryo kidney (PEK) cell line. 5-(Perylen-3-yl)-2-thiophenecarboxylic acid (5a) showed the highest antiviral activity with 50% effective concentration of approx. 1.6 nM.
Collapse
Affiliation(s)
- Nikita A Slesarchuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia; Department of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117312, Russia
| | - Evgeny V Khvatov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; FSBSI «Chumakov FSC R&D IBP RAS», Moscow 108819, Russia
| | - Alexey A Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Department of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117312, Russia
| | - Gleb V Proskurin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia
| | - Timofei D Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Anastasiya I Lazarevich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Mendeleev University of Chemical Technology, Moscow 125047, Russia
| | - Angelina A Ulanovskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Mendeleev University of Chemical Technology, Moscow 125047, Russia
| | | | | | - Artjom V Jegorov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia
| | - Alexey V Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Department of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117312, Russia.
| | - Anton P Tyurin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Department of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117312, Russia; Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Vadim V Shmanai
- Institute of Physico-Organic Chemistry, Minsk 220072, Belarus
| | - Aydar A Ishmukhametov
- FSBSI «Chumakov FSC R&D IBP RAS», Moscow 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia; Department of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 117312, Russia; Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Dmitry I Osolodkin
- Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia; FSBSI «Chumakov FSC R&D IBP RAS», Moscow 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Liubov I Kozlovskaya
- FSBSI «Chumakov FSC R&D IBP RAS», Moscow 108819, Russia; Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Andrey V Aralov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow 117997, Russia
| |
Collapse
|
27
|
Synthetic surfactin analogues have improved anti-PEDV properties. PLoS One 2019; 14:e0215227. [PMID: 30973929 PMCID: PMC6459484 DOI: 10.1371/journal.pone.0215227] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 03/29/2019] [Indexed: 01/15/2023] Open
Abstract
Surfactin has antiviral activity against various enveloped viruses by inhibiting viral membrane fusion. However, the potential utility of surfactin as an antiviral drug is limited by its cytotoxicity. In this study, 10 surfactin analogues were obtained by chemical synthesis and evaluated to determine their anti-PEDV activities, hemolytic activities, and critical micelle concentrations. The main goal of our study was to develop a safer drug; a surfactin analogue with high anti-PEDV activity and low hemolytic activity. Compared with surfactin, one of the analogues we developed, SLP5, has lower hemolytic activity, with the same antiviral activity. The selectivity index of SLP5 is 52, while the SI for surfactin is 4, in other words, the safe and effective concentration range of SLP5 is 12 times greater than that of surfactin. Like surfactin, SLP5 has a direct antiviral effect on PEDV. Structurally, SLP5 is a linear lipopeptide with three carboxyl groups. Surfactin derivatives similar to SLP5 could be obtained by lactone bond hydrolyzation of surfactin, as well as total synthesis.
Collapse
|
28
|
Chistov AA, Orlov AA, Streshnev PP, Slesarchuk NA, Aparin IO, Rathi B, Brylev VA, Kutyakov SV, Mikhura IV, Ustinov AV, Westman G, Palyulin VA, Jain N, Osolodkin DI, Kozlovskaya LI, Korshun VA. Compounds based on 5-(perylen-3-ylethynyl)uracil scaffold: High activity against tick-borne encephalitis virus and non-specific activity against enterovirus A. Eur J Med Chem 2019; 171:93-103. [PMID: 30909022 DOI: 10.1016/j.ejmech.2019.03.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/17/2019] [Accepted: 03/12/2019] [Indexed: 12/26/2022]
Abstract
Rigid amphipathic fusion inhibitors (RAFIs) are potent antivirals based on a perylene core linked with a nucleoside moiety. Sugar-free analogues of RAFIs, 5-(perylen-3-ylethynyl)uracil-1-acetic acid 1 and its amides 2, were synthesized using combined protection group strategy. Compounds 1 and 2 appeared to have low toxicity on porcine embryo kidney (PEK) or rhabdomiosarcoma (RD) cells together with remarkable activity against enveloped tick-borne encephalitis virus (TBEV): EC50 values vary from 0.077 μM to subnanomolar range. Surprisingly, 3-pivaloyloxymethyl (Pom) protected precursors 7 and 8 showed even more pronounced activity. All the compounds showed no activity against several non-enveloped enteroviruses, except 4-hydroxybutylamides 2d,g, which inhibited the reproduction of enterovirus A71 with EC50 50-100 μM, with a non-specific mode of action. The results suggest that the carbohydrate moiety of RAFI nucleosides does not play a crucial role in their antiviral action, and biological activity of the 5-(perylen-3-ylethynyl)uracil scaffold can be effectively modulated by substituents in positions 1 and 3. The high antiviral activity of these new compounds, coupled with low toxicity advocate their potential role in antiviral therapy.
Collapse
Affiliation(s)
- Alexey A Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Alexey A Orlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia; FSBSI "Chumakov FSC R&D IBP RAS", 8 bd 1 Poselok Instituta Poliomielita, Poselenie Moskovsky, Moscow 108819, Russia; Department of Chemistry, Lomonosov Moscow State University, Leninskie gory 1 bd 3, Moscow 119992, Russia
| | - Philipp P Streshnev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Nikita A Slesarchuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Ilya O Aparin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia; Skolkovo Institute of Science and Technology, Skolkovo, Moscow 143026, Russia
| | - Brijesh Rathi
- Department of Chemistry, Laboratory for Translational Chemistry and Drug Discovery, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
| | - Vladimir A Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia; Biotech Innovations Ltd, Leninskie gory 1 bd 75, Moscow 119992, Russia
| | - Sergey V Kutyakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Irina V Mikhura
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Alexey V Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Gunnar Westman
- Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Vladimir A Palyulin
- Department of Chemistry, Lomonosov Moscow State University, Leninskie gory 1 bd 3, Moscow 119992, Russia
| | - Nidhi Jain
- Department of Chemistry, Indian Institute of Technology, Delhi, India
| | - Dmitry I Osolodkin
- FSBSI "Chumakov FSC R&D IBP RAS", 8 bd 1 Poselok Instituta Poliomielita, Poselenie Moskovsky, Moscow 108819, Russia; Department of Chemistry, Lomonosov Moscow State University, Leninskie gory 1 bd 3, Moscow 119992, Russia; Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Liubov I Kozlovskaya
- FSBSI "Chumakov FSC R&D IBP RAS", 8 bd 1 Poselok Instituta Poliomielita, Poselenie Moskovsky, Moscow 108819, Russia; Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia.
| |
Collapse
|
29
|
Tick-borne encephalitis in Europe and Russia: Review of pathogenesis, clinical features, therapy, and vaccines. Antiviral Res 2019; 164:23-51. [PMID: 30710567 DOI: 10.1016/j.antiviral.2019.01.014] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/10/2018] [Accepted: 01/22/2019] [Indexed: 02/07/2023]
Abstract
Tick-borne encephalitis (TBE) is an illness caused by tick-borne encephalitis virus (TBEV) infection which is often limited to a febrile illness, but may lead to very aggressive downstream neurological manifestations. The disease is prevalent in forested areas of Europe and northeastern Asia, and is typically caused by infection involving one of three TBEV subtypes, namely the European (TBEV-Eu), the Siberian (TBEV-Sib), or the Far Eastern (TBEV-FE) subtypes. In addition to the three main TBEV subtypes, two other subtypes; i.e., the Baikalian (TBEV-Bkl) and the Himalayan subtype (TBEV-Him), have been described recently. In Europe, TBEV-Eu infection usually results in only mild TBE associated with a mortality rate of <2%. TBEV-Sib infection also results in a generally mild TBE associated with a non-paralytic febrile form of encephalitis, although there is a tendency towards persistent TBE caused by chronic viral infection. TBE-FE infection is considered to induce the most severe forms of TBE. Importantly though, viral subtype is not the sole determinant of TBE severity; both mild and severe cases of TBE are in fact associated with infection by any of the subtypes. In keeping with this observation, the overall TBE mortality rate in Russia is ∼2%, in spite of the fact that TBEV-Sib and TBEV-FE subtypes appear to be inducers of more severe TBE than TBEV-Eu. On the other hand, TBEV-Sib and TBEV-FE subtype infections in Russia are associated with essentially unique forms of TBE rarely seen elsewhere if at all, such as the hemorrhagic and chronic (progressive) forms of the disease. For post-exposure prophylaxis and TBE treatment in Russia and Kazakhstan, a specific anti-TBEV immunoglobulin is currently used with well-documented efficacy, but the use of specific TBEV immunoglobulins has been discontinued in Europe due to concerns regarding antibody-enhanced disease in naïve individuals. Therefore, new treatments are essential. This review summarizes available data on the pathogenesis and clinical features of TBE, plus different vaccine preparations available in Europe and Russia. In addition, new treatment possibilities, including small molecule drugs and experimental immunotherapies are reviewed. The authors caution that their descriptions of approved or experimental therapies should not be considered to be recommendations for patient care.
Collapse
|
30
|
Sapozhnikova KA, Slesarchuk NA, Orlov AA, Khvatov EV, Radchenko EV, Chistov AA, Ustinov AV, Palyulin VA, Kozlovskaya LI, Osolodkin DI, Korshun VA, Brylev VA. Ramified derivatives of 5-(perylen-3-ylethynyl)uracil-1-acetic acid and their antiviral properties. RSC Adv 2019; 9:26014-26023. [PMID: 35531032 PMCID: PMC9070374 DOI: 10.1039/c9ra06313g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/14/2019] [Indexed: 01/03/2023] Open
Abstract
The propargylamide of N3-Pom-protected 5-(perylen-3-ylethynyl)uracil acetic acid, a universal precursor, was used in a CuAAC click reaction for the synthesis of several derivatives, including three ramified molecules with high activities against tick-borne encephalitis virus (TBEV). Pentaerythritol-based polyazides were used for the assembly of molecules containing 2⋯4 antiviral 5-(perylen-3-ylethynyl)uracil scaffolds, the first examples of polyvalent perylene antivirals. Cluster compounds showed enhanced absorbance, however, their fluorescence was reduced due to self-quenching. Due to the solubility issues, Pom group removal succeeded only for compounds with one peryleneethynyluracil unit. Four compounds, including one ramified cluster 9f, showed remarkable 1⋯3 nM EC50 values against TBEV in cell culture. Ramified clusters of antiviral perylenylethynyl scaffold were prepared using CuAAC reaction of 5-(perylen-3-ylethynyl)-3-pivaloyloxymethyl-1-(propargylamidomethyl)uracil with azides. Compounds inhibited TBEV reproduction at nanomolar concentrations.![]()
Collapse
Affiliation(s)
| | - Nikita A. Slesarchuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry
- Moscow 117997
- Russia
- Department of Chemistry
- Lomonosov Moscow State University
| | - Alexey A. Orlov
- Department of Chemistry
- Lomonosov Moscow State University
- Moscow 119991
- Russia
- FSBSI "Chumakov FSC R&D IBP RAS"
| | - Evgeny V. Khvatov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry
- Moscow 117997
- Russia
- FSBSI "Chumakov FSC R&D IBP RAS"
- Moscow 108819
| | | | - Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry
- Moscow 117997
- Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry
- Moscow 117997
- Russia
- Biotech Innovations Ltd
- Moscow 119992
| | | | - Liubov I. Kozlovskaya
- FSBSI "Chumakov FSC R&D IBP RAS"
- Moscow 108819
- Russia
- Sechenov First Moscow State Medical University
- Moscow 119991
| | - Dmitry I. Osolodkin
- Department of Chemistry
- Lomonosov Moscow State University
- Moscow 119991
- Russia
- FSBSI "Chumakov FSC R&D IBP RAS"
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry
- Moscow 117997
- Russia
- Department of Biology and Biotechnology
- National Research University Higher School of Economics
| | - Vladimir A. Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry
- Moscow 117997
- Russia
- Biotech Innovations Ltd
- Moscow 119992
| |
Collapse
|
31
|
Yuan L, Zhang S, Wang Y, Li Y, Wang X, Yang Q. Surfactin Inhibits Membrane Fusion during Invasion of Epithelial Cells by Enveloped Viruses. J Virol 2018; 92:e00809-18. [PMID: 30068648 PMCID: PMC6189506 DOI: 10.1128/jvi.00809-18] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/25/2018] [Indexed: 01/15/2023] Open
Abstract
Because membrane fusion is a crucial step in the process by which enveloped viruses invade host cells, membrane fusion inhibitors can be effective drugs against enveloped viruses. We found that surfactin from Bacillus subtilis can suppress the proliferation of porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) in epithelial cells at a relatively low concentration range (15 to 50 μg/ml), without cytotoxicity or viral membrane disruption. Membrane fusion inhibition experiments demonstrate that surfactin treatment significantly reduces the rate at which the virus fuses to the cell membrane. Thermodynamic experiments show that the incorporation of small amounts of surfactin hinders the formation of negative curvature by lamellar-phase lipids, suggesting that surfactin acts a membrane fusion inhibitor. A fluorescent lipopeptide similar to surfactin was synthesized, and its ability to insert into the viral membrane was confirmed by spectroscopy. In vivo experiments have shown that oral administration of surfactin to piglets protects against PEDV infection. In conclusion, our study indicates that surfactin is a membrane fusion inhibitor with activity against enveloped viruses. As the first reported naturally occurring wedge lipid membrane fusion inhibitor, surfactin is likely to be a prototype for the development of a broad range of novel antiviral drugs.IMPORTANCE Membrane fusion inhibitors are a rapidly emerging class of antiviral drugs that inhibit the infection process of enveloped viruses. They can be classified, on the basis of the viral components targeted, as fusion protein targeting or membrane lipid targeting. Lipid-targeting membrane fusion inhibitors have a broader antiviral spectrum and are less likely to select for drug-resistant mutations. Here we show that surfactin is a membrane fusion inhibitor and has a strong antiviral effect. The insertion of surfactin into the viral envelope lipids reduces the probability of viral fusion. We also demonstrate that oral administration of surfactin protects piglets from PEDV infection. Surfactin is the first naturally occurring wedge lipid membrane fusion inhibitor that has been identified and may be effective against many viruses beyond the scope of this study. Understanding its mechanism of action provides a foundation for the development of novel antiviral agents.
Collapse
Affiliation(s)
- Lvfeng Yuan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Shuai Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Yongheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Xiaoqing Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University. Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
32
|
Proskurin GV, Orlov AA, Brylev VA, Kozlovskaya LI, Chistov AA, Karganova GG, Palyulin VA, Osolodkin DI, Korshun VA, Aralov AV. 3′-O-Substituted 5-(perylen-3-ylethynyl)-2′-deoxyuridines as tick-borne encephalitis virus reproduction inhibitors. Eur J Med Chem 2018; 155:77-83. [DOI: 10.1016/j.ejmech.2018.05.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/01/2018] [Accepted: 05/24/2018] [Indexed: 12/11/2022]
|
33
|
Henipavirus Infection: Natural History and the Virus-Host Interplay. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2018. [DOI: 10.1007/s40506-018-0155-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
34
|
Sepúlveda-Crespo D, de la Mata FJ, Gómez R, Muñoz-Fernández MA. Sulfonate-ended carbosilane dendrimers with a flexible scaffold cause inactivation of HIV-1 virions and gp120 shedding. NANOSCALE 2018; 10:8998-9011. [PMID: 29726564 DOI: 10.1039/c8nr01664j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Infection with human immunodeficiency virus type 1 (HIV-1) continues to be a global public health issue, especially in low-resource countries. Sexual transmission is responsible for the majority of HIV-1 infections worldwide. Women are more susceptible to HIV-1 acquisition than men and represent nearly 50% of the HIV-infected population. Topical vaginal microbicides that act at the earlier stages of infection offer a prevention strategy to reduce the acquisition of HIV-1. Dendrimers are nano-sized, radially symmetric molecules with a well-defined and monodisperse structure consisting of tree-like arms or branches. We perform a TZM.bl cell line-based screening of two families of carbosilane dendrimers (6 nanocompounds: G1-S12P, G2-S24P, G3-S48P, G1-C12P, G2-C24P and G3-C48P) that we have previously synthesized, containing 12, 24 or 48 sulfonate (or carboxylate) end-groups and a polyphenolic core. This work shows that second- and third-generation sulfonate-ended carbosilane dendrimers with a polyphenolic core (G2-S24P and G3-S48P, respectively) display low cytotoxicity (CC50 > 300 μM) with virucidal anti-R5-HIV-1 activity (EC50 < 50 nM; therapeutic index >6000) causing irreversible HIV-1 inactivation (80-90%) by loss of HIV-1 RNA (40%), gp120 shedding (70-80%) and p24 capsid protein release (45-60%). Herein, we demonstrate that sulfonate end-groups and a flexible scaffold from carbosilane dendrimers strongly influence their properties acting as potent virucides.
Collapse
Affiliation(s)
- Daniel Sepúlveda-Crespo
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain.
| | | | | | | |
Collapse
|
35
|
Buranda T, Gineste C, Wu Y, Bondu V, Perez D, Lake KR, Edwards BS, Sklar LA. A High-Throughput Flow Cytometry Screen Identifies Molecules That Inhibit Hantavirus Cell Entry. SLAS DISCOVERY 2018; 23:634-645. [PMID: 29608398 DOI: 10.1177/2472555218766623] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS), which infects more than 200,000 people worldwide. Sin Nombre virus (SNV) and Andes virus (ANDV) cause the most severe form of HCPS, with case fatality ratios of 30%-40%. There are no specific therapies or vaccines for SNV. Using high-throughput flow cytometry, we screened the Prestwick Chemical Library for small-molecule inhibitors of the binding interaction between UV-inactivated and fluorescently labeled SNVR18 particles, and decay-accelerating factor (DAF) expressed on Tanoue B cells. Eight confirmed hit compounds from the primary screen were investigated further in secondary screens that included infection inhibition, cytotoxicity, and probe interference. Antimycin emerged as a bona fide hit compound that inhibited cellular infection of the major HCPS (SNV)- and HCPS (Hantaan)-causing viruses. Confirming our assay's ability to detect active compounds, orthogonal testing of the hit compound showed that antimycin binds directly to the virus particle and blocks recapitulation of physiologic integrin activation caused by SNV binding to the integrin PSI domain.
Collapse
Affiliation(s)
- Tione Buranda
- 1 Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA, and in revised form Feb 15, 2018. Accepted for publication Mar 1, 2018.,2 Center for Infectious Diseases and Immunity, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Catherine Gineste
- 3 University of New Mexico Center for Molecular Discovery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Yang Wu
- 1 Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA, and in revised form Feb 15, 2018. Accepted for publication Mar 1, 2018
| | - Virginie Bondu
- 1 Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA, and in revised form Feb 15, 2018. Accepted for publication Mar 1, 2018
| | - Dominique Perez
- 1 Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA, and in revised form Feb 15, 2018. Accepted for publication Mar 1, 2018.,4 Department of Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Kaylin R Lake
- 5 Department of Biochemistry, University of New Mexico, Albuquerque, NM, USA
| | - Bruce S Edwards
- 1 Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA, and in revised form Feb 15, 2018. Accepted for publication Mar 1, 2018.,3 University of New Mexico Center for Molecular Discovery, University of New Mexico School of Medicine, Albuquerque, NM, USA.,4 Department of Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Larry A Sklar
- 1 Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA, and in revised form Feb 15, 2018. Accepted for publication Mar 1, 2018.,3 University of New Mexico Center for Molecular Discovery, University of New Mexico School of Medicine, Albuquerque, NM, USA.,4 Department of Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
36
|
Kozlovskaya LI, Golinets AD, Eletskaya AA, Orlov AA, Palyulin VA, Kochetkov SN, Alexandrova LA, Osolodkin DI. Selective Inhibition of Enterovirus A Species Members' Reproduction by Furano[2, 3- d]pyrimidine Nucleosides Revealed by Antiviral Activity Profiling against (+)ssRNA Viruses. ChemistrySelect 2018; 3:2321-2325. [PMID: 32328513 PMCID: PMC7169607 DOI: 10.1002/slct.201703052] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
The rational design of broad-spectrum antivirals requires data on antiviral activity of compounds against multiple viruses, which are often not available. We have developed a panel of (+)ssRNA viruses composed of Enterovirus and Flavivirus genera members allowing to study these activity spectra. Antiviral activity profiling of a set of nucleoside analogues revealed N 4-hydroxycytidine as an efficient inhibitor of replication of coxsackieviruses and other enteroviruses, but ineffective against tick-borne encephalitis virus. Furano[2, 3-d]pyrimidine nucleosides with n-pentyl or n-hexyl tails showed selective inhibition of Enterovirus A representatives. 5-(Tetradec-1-yn-1-yl)-uridine showed selective inhibition of tick-borne encephalitis virus at the micromolar level.
Collapse
Affiliation(s)
- Liubov I. Kozlovskaya
- Institute of Poliomielitis and Viral EncephalitidesFSBSI Chumakov FSC R&D IBP RASPoselok Instituta Poliomielita, 8 bd. 1, Poselenie MoskovskyMoscow 108819Russia
- Sechenov First Moscow State Medical University, Trubetskaya ul., 8Moscow 119991Russia
| | - Anastasia D. Golinets
- Institute of Poliomielitis and Viral EncephalitidesFSBSI Chumakov FSC R&D IBP RASPoselok Instituta Poliomielita, 8 bd. 1, Poselenie MoskovskyMoscow 108819Russia
- Sechenov First Moscow State Medical University, Trubetskaya ul., 8Moscow 119991Russia
| | - Anastasia A. Eletskaya
- Institute of Poliomielitis and Viral EncephalitidesFSBSI Chumakov FSC R&D IBP RASPoselok Instituta Poliomielita, 8 bd. 1, Poselenie MoskovskyMoscow 108819Russia
- Lomonosov Moscow State University, Leninskie Gory, 1Moscow 119991Russia
| | - Alexey A. Orlov
- Institute of Poliomielitis and Viral EncephalitidesFSBSI Chumakov FSC R&D IBP RASPoselok Instituta Poliomielita, 8 bd. 1, Poselenie MoskovskyMoscow 108819Russia
- Lomonosov Moscow State University, Leninskie Gory, 1Moscow 119991Russia
| | | | - Sergey N. Kochetkov
- Engelhargt Institute of Molecular BiologyRussian Academy of Sciences, Ul. Vavilova, 32Moscow 119991Russia
| | - Liudmila A. Alexandrova
- Engelhargt Institute of Molecular BiologyRussian Academy of Sciences, Ul. Vavilova, 32Moscow 119991Russia
| | - Dmitry I. Osolodkin
- Institute of Poliomielitis and Viral EncephalitidesFSBSI Chumakov FSC R&D IBP RASPoselok Instituta Poliomielita, 8 bd. 1, Poselenie MoskovskyMoscow 108819Russia
- Sechenov First Moscow State Medical University, Trubetskaya ul., 8Moscow 119991Russia
- Lomonosov Moscow State University, Leninskie Gory, 1Moscow 119991Russia
| |
Collapse
|
37
|
Carravilla P, Nieva JL. HIV antivirals: targeting the functional organization of the lipid envelope. Future Virol 2018. [DOI: 10.2217/fvl-2017-0114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Most of the surface of the lipid bilayer covering the human immunodeficiency virus type 1 (HIV-1) particle is directly accessible from the aqueous medium. Its peculiar chemical composition and physical properties appear to be critical for infection and, therefore, may comprise a target for selective antiviral activity. The HIV-1 membrane is enriched in raft-type lipids and also displays aminophospholipids on its external leaflet. We contend here that a great deal of membrane-active compounds described to block HIV-1 infection can do so by following a common mechanism of action: alteration of the lateral heterogeneity that supports the functional organization of the lipid envelope. The confirmation of this hypothesis could lay new foundations for the rational development of compounds with anti-HIV activity.
Collapse
Affiliation(s)
- Pablo Carravilla
- Biofisika Institute (CSIC, UPV/EHU) & Department of Biochemistry & Molecular Biology, University of the Basque Country (UPV/EHU), PO Box 644, 48080 Bilbao, Spain
| | - José L Nieva
- Biofisika Institute (CSIC, UPV/EHU) & Department of Biochemistry & Molecular Biology, University of the Basque Country (UPV/EHU), PO Box 644, 48080 Bilbao, Spain
| |
Collapse
|
38
|
Eyer L, Nencka R, de Clercq E, Seley-Radtke K, Růžek D. Nucleoside analogs as a rich source of antiviral agents active against arthropod-borne flaviviruses. Antivir Chem Chemother 2018; 26:2040206618761299. [PMID: 29534608 PMCID: PMC5890575 DOI: 10.1177/2040206618761299] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/30/2018] [Indexed: 12/27/2022] Open
Abstract
Nucleoside analogs represent the largest class of small molecule-based antivirals, which currently form the backbone of chemotherapy of chronic infections caused by HIV, hepatitis B or C viruses, and herpes viruses. High antiviral potency and favorable pharmacokinetics parameters make some nucleoside analogs suitable also for the treatment of acute infections caused by other medically important RNA and DNA viruses. This review summarizes available information on antiviral research of nucleoside analogs against arthropod-borne members of the genus Flavivirus within the family Flaviviridae, being primarily focused on description of nucleoside inhibitors of flaviviral RNA-dependent RNA polymerase, methyltransferase, and helicase/NTPase. Inhibitors of intracellular nucleoside synthesis and newly discovered nucleoside derivatives with high antiflavivirus potency, whose modes of action are currently not completely understood, have drawn attention. Moreover, this review highlights important challenges and complications in nucleoside analog development and suggests possible strategies to overcome these limitations.
Collapse
Affiliation(s)
- Luděk Eyer
- Department of Virology, Veterinary Research Institute, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Radim Nencka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Erik de Clercq
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Daniel Růžek
- Department of Virology, Veterinary Research Institute, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
| |
Collapse
|
39
|
Rigid amphipathic fusion inhibitors demonstrate antiviral activity against African swine fever virus. J Gen Virol 2018; 99:148-156. [DOI: 10.1099/jgv.0.000991] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
40
|
Speerstra S, Chistov AA, Proskurin GV, Aralov AV, Ulashchik EA, Streshnev PP, Shmanai VV, Korshun VA, Schang LM. Antivirals acting on viral envelopes via biophysical mechanisms of action. Antiviral Res 2018; 149:164-173. [DOI: 10.1016/j.antiviral.2017.11.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/04/2023]
|
41
|
Sobhy H. A comparative review of viral entry and attachment during large and giant dsDNA virus infections. Arch Virol 2017; 162:3567-3585. [PMID: 28866775 PMCID: PMC5671522 DOI: 10.1007/s00705-017-3497-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/13/2017] [Indexed: 12/19/2022]
Abstract
Viruses enter host cells via several mechanisms, including endocytosis, macropinocytosis, and phagocytosis. They can also fuse at the plasma membrane and can spread within the host via cell-to-cell fusion or syncytia. The mechanism used by a given viral strain depends on its external topology and proteome and the type of cell being entered. This comparative review discusses the cellular attachment receptors and entry pathways of dsDNA viruses belonging to the families Adenoviridae, Baculoviridae, Herpesviridae and nucleocytoplasmic large DNA viruses (NCLDVs) belonging to the families Ascoviridae, Asfarviridae, Iridoviridae, Phycodnaviridae, and Poxviridae, and giant viruses belonging to the families Mimiviridae and Marseilleviridae as well as the proposed families Pandoraviridae and Pithoviridae. Although these viruses have several common features (e.g., topology, replication and protein sequence similarities) they utilize different entry pathways to infect wide-range of hosts, including humans, other mammals, invertebrates, fish, protozoa and algae. Similarities and differences between the entry methods used by these virus families are highlighted, with particular emphasis on viral topology and proteins that mediate viral attachment and entry. Cell types that are frequently used to study viral entry are also reviewed, along with other factors that affect virus-host cell interactions.
Collapse
Affiliation(s)
- Haitham Sobhy
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden.
| |
Collapse
|
42
|
Pentagalloylglucose, a highly bioavailable polyphenolic compound present in Cortex moutan, efficiently blocks hepatitis C virus entry. Antiviral Res 2017; 147:19-28. [PMID: 28923507 DOI: 10.1016/j.antiviral.2017.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 08/07/2017] [Accepted: 09/13/2017] [Indexed: 01/19/2023]
Abstract
Approximately 142 million people worldwide are infected with hepatitis C virus (HCV). Although potent direct acting antivirals are available, high costs limit access to treatment. Chronic hepatitis C virus infection remains a major cause of orthotopic liver transplantation. Moreover, re-infection of the graft occurs regularly. Antivirals derived from natural sources might be an alternative and cost-effective option to complement therapy regimens for global control of hepatitis C virus infection. We tested the antiviral properties of a mixture of different Chinese herbs/roots named Zhi Bai Di Huang Wan (ZBDHW) and its individual components on HCV. One of the ZBDHW components, Penta-O-Galloyl-Glucose (PGG), was further analyzed for its mode of action in vitro, its antiviral activity in primary human hepatocytes as well as for its bioavailability and hepatotoxicity in mice. ZBDHW, its component Cortex Moutan and the compound PGG efficiently block entry of HCV of all major genotypes and also of the related flavivirus Zika virus. PGG does not disrupt HCV virion integrity and acts primarily during virus attachment. PGG shows an additive effect when combined with the well characterized HCV inhibitor Daclatasvir. Analysis of bioavailability in mice revealed plasma levels above tissue culture IC50 after a single intraperitoneal injection. In conclusion, PGG is a pangenotypic HCV entry inhibitor with high bioavailability. The low cost and wide availability of this compound make it a promising candidate for HCV combination therapies, and also emerging human pathogenic flaviviruses like ZIKV.
Collapse
|
43
|
Aralov AV, Proskurin GV, Orlov AA, Kozlovskaya LI, Chistov AA, Kutyakov SV, Karganova GG, Palyulin VA, Osolodkin DI, Korshun VA. Perylenyltriazoles inhibit reproduction of enveloped viruses. Eur J Med Chem 2017; 138:293-299. [DOI: 10.1016/j.ejmech.2017.06.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/06/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022]
|
44
|
Chistov AA, Kutyakov SV, Guz AV, Mikhura IV, Ustinov AV, Korshun VA. Improved Large-scale Synthesis of 5-(Perylen-3-ylethynyl)-arabino-uridine (aUY11), the Broad-Spectrum Antiviral. ORG PREP PROCED INT 2017. [DOI: 10.1080/00304948.2017.1343040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | | | - Arsentiy V. Guz
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Irina V. Mikhura
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | | |
Collapse
|
45
|
Pietschmann T. Clinically Approved Ion Channel Inhibitors Close Gates for Hepatitis C Virus and Open Doors for Drug Repurposing in Infectious Viral Diseases. J Virol 2017; 91:e01914-16. [PMID: 27807238 PMCID: PMC5215334 DOI: 10.1128/jvi.01914-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection causes severe liver disease and affects ca. 146 million individuals. Novel directly acting antivirals targeting HCV have revolutionized treatment. However, high costs limit access to therapy. Recently, several related drugs used in humans to treat allergies or as neuroleptics emerged as potent HCV cell entry inhibitors. Insights into their antiviral modes of action may increase opportunities for drug repurposing in hepatitis C and possibly other important human viral infections.
Collapse
Affiliation(s)
- Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany, and German Centre for Infection Research, Hannover-Braunschweig, Germany
| |
Collapse
|
46
|
Chistov AA, Ivanov NM, Kutyakov SV, Ustinov AV, Glybin AV, Streshnev PP, Mikhura IV, Korshun VA. Fluorescent nucleosides with an elongated rigid linker: attaching perylene to a nucleobase via a one-pot desilylation/Sonogashira reaction. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.09.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
47
|
Hepatitis C virus cell entry: a target for novel antiviral strategies to address limitations of direct acting antivirals. Hepatol Int 2016; 10:741-8. [DOI: 10.1007/s12072-016-9724-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/16/2016] [Indexed: 12/12/2022]
|
48
|
Jackman JA, Lee J, Cho NJ. Nanomedicine for Infectious Disease Applications: Innovation towards Broad-Spectrum Treatment of Viral Infections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1133-9. [PMID: 26551316 DOI: 10.1002/smll.201500854] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 07/10/2015] [Indexed: 05/27/2023]
Abstract
Nanomedicine enables unique diagnostic and therapeutic capabilities to tackle problems in clinical medicine. As multifunctional agents with programmable properties, nanomedicines are poised to revolutionize treatment strategies. This promise is especially evident for infectious disease applications, for which the continual emergence, re-emergence, and evolution of pathogens has proven difficult to counter by conventional approaches. Herein, a conceptual framework is presented that envisions possible routes for the development of nanomedicines as superior broad-spectrum antiviral agents against enveloped viruses. With lipid membranes playing a critical role in the life cycle of medically important enveloped viruses including HIV, influenza, and Ebola, cellular and viral membrane interfaces are ideal elements to incorporate into broad-spectrum antiviral strategies. Examples are presented that demonstrate how nanomedicine strategies inspired by lipid membranes enable a wide range of targeting opportunities to gain control of critical stages in the virus life cycle through either direct or indirect approaches involving membrane interfaces. The capabilities can be realized by enabling new inhibitory functions or improving the function of existing drugs through nanotechnology-enabled solutions. With these exciting opportunities, due attention is also given to the clinical translation of nanomedicines for infectious disease applications, especially as pharmaceutical drug-discovery pipelines demand new routes of innovation.
Collapse
Affiliation(s)
- Joshua A Jackman
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, 637553, Singapore
| | - Jaywon Lee
- College of Business, Korea Advanced Institute of Science and Technology, 85 Hoegiro, Seoul, 130-722, Korea
| | - Nam-Joon Cho
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, 637553, Singapore
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, 637459, Singapore
| |
Collapse
|
49
|
Chistov AA, Kutyakov SV, Ustinov AV, Aparin IO, Glybin AV, Mikhura IV, Korshun VA. 2-Ethynylperylene and improved synthesis of 3-ethynylperylene. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.01.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
50
|
Rowse M, Qiu S, Tsao J, Yamauchi Y, Wang G, Luo M. Reduction of Influenza Virus Envelope's Fusogenicity by Viral Fusion Inhibitors. ACS Infect Dis 2016; 2:47-53. [PMID: 27622947 DOI: 10.1021/acsinfecdis.5b00109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
During cell entry of an enveloped virus, the viral membrane must be fused with the cellular membrane. The virus envelope has a unique structure consisting of viral proteins and a virus-specific lipid composition, whereas the host membrane has its own structure with host membrane proteins. Compound 136 was previously found to bind in close proximity to the viral envelope and inhibit influenza virus entry. We showed here that the 136-treated influenza virus still caused hemolysis. When liposomes were used as the target membrane for 136-treated viruses, aberrant fusion occurred; few liposomes fused per virion, and glycoproteins were not distributed evenly across fusion complexes. Additionally, large fusion aggregates did not form, and in some instances, neck-like structures were found. Based on previous results and hemolysis, fusion inhibition by 136 occurs post-scission but prior to lipid mixing.
Collapse
Affiliation(s)
- Michael Rowse
- Department of Microbiology, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, Alabama 35294, United States
| | - Shihong Qiu
- Department of Microbiology, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, Alabama 35294, United States
| | - Jun Tsao
- Department of Microbiology, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, Alabama 35294, United States
| | - Yohei Yamauchi
- Institute of Biochemistry, ETH Zurich, HPM E8.2, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Guoxin Wang
- Laboratory
of Structural Biology, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Ming Luo
- Laboratory
of Structural Biology, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Department of Chemistry College of Arts
and Sciences, Georgia State University, Atlanta, Georgia 30302, United States
| |
Collapse
|