1
|
Mosidze E, Franci G, Dell'Annunziata F, Capuano N, Colella M, Salzano F, Galdiero M, Bakuridze A, Folliero V. Silver Nanoparticle-Mediated Antiviral Efficacy against Enveloped Viruses: A Comprehensive Review. GLOBAL CHALLENGES (HOBOKEN, NJ) 2025; 9:2400380. [PMID: 40352632 PMCID: PMC12065099 DOI: 10.1002/gch2.202400380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/19/2025] [Indexed: 05/14/2025]
Abstract
Viral infections continue to pose a significant challenge to global health, with increasing resistance to conventional antiviral therapies highlighting the urgent need for alternative treatment strategies. Silver nanoparticles (AgNPs) have attracted attention as broad-spectrum antiviral agents due to their unique physicochemical properties and ability to target multiple stages of viral infection. This review provides a comprehensive analysis of the antiviral mechanisms of AgNPs, highlighting their efficacy against clinically relevant enveloped viruses such as influenza, herpes simplex, hepatitis B, and coronaviruses. How key nanoparticle characteristics, including size, shape, surface functionalization, and synthesis methods, influence their antiviral performance is examined. Studies indicate that AgNPs exert their effects through direct interactions with viral particles, inhibition of viral adhesion, and entry into host cells with disruption of viral replication. Furthermore, their potential applications in therapeutic formulations, antiviral coatings, and nanomedicine-based strategies are explored. Despite their promise, challenges regarding cytotoxicity, stability, and large-scale production must be addressed to ensure their safe and effective clinical use. This review highlights the transformative potential of AgNPs in antiviral therapy and highlights the need for further investigation to facilitate their clinical translation in the fight against emerging and drug-resistant viral infections.
Collapse
Affiliation(s)
- Ekaterine Mosidze
- Department of Pharmaceutical Technology33 Vazha‐Pshavela AveTbilisi0178Georgia
| | - Gianluigi Franci
- Department of MedicineSurgery and Dentistry “Scuola Medica Salernitana”University of SalernoVia S. Allende 43Baronissi84081Italy
| | - Federica Dell'Annunziata
- Department of MedicineSurgery and Dentistry “Scuola Medica Salernitana”University of SalernoVia S. Allende 43Baronissi84081Italy
| | - Nicoletta Capuano
- Department of MedicineSurgery and Dentistry “Scuola Medica Salernitana”University of SalernoVia S. Allende 43Baronissi84081Italy
| | - Marica Colella
- Microbiology and Virology Unit, Interdisciplinary Department of MedicineUniversity of Bari “Aldo Moro”Piazza G. Cesare 11Bari70124Italy
- Department of Theoretical and Applied Sciences (DiSTA)eCampus UniversityNovedrate22060Italy
| | - Flora Salzano
- Department of MedicineSurgery and Dentistry “Scuola Medica Salernitana”University of SalernoVia S. Allende 43Baronissi84081Italy
| | - Massimiliano Galdiero
- Department of Experimental MedicineUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Aliosha Bakuridze
- Department of Pharmaceutical Technology33 Vazha‐Pshavela AveTbilisi0178Georgia
| | - Veronica Folliero
- Department of MedicineSurgery and Dentistry “Scuola Medica Salernitana”University of SalernoVia S. Allende 43Baronissi84081Italy
| |
Collapse
|
2
|
Lourenco Nogueira C, Boegel SJ, Shukla M, Ngo W, Hui A, Jones LW, Aucoin MG. Antiviral activity of contact lens care solutions and rub-and-rinse regimen against adenovirus. Optom Vis Sci 2025; 102:156-166. [PMID: 39903204 PMCID: PMC11939091 DOI: 10.1097/opx.0000000000002231] [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: 02/06/2025] Open
Abstract
SIGNIFICANCE Although human adenoviruses are the leading cause of acute viral conjunctivitis, there is a lack of data surrounding the efficacy of contact lens care products against these viruses. PURPOSE This study investigates the antiviral activity of several commercially available contact lens care solutions against human adenovirus type 5 (Ad5). METHODS Six contact lens care solutions (Biotrue, Boston Simplus, OPTI-FREE Puremoist, Clear Care, cleadew, and cleadew GP) were investigated. Quantitative suspensions tests were conducted on Ad5 solutions after interaction with the different contact lens care solutions for 4 or 6 hours. For the hydrogen peroxide solution (Clear Care), interaction times of 0, 2, 4, and 6 hours prior to neutralization were also investigated. Finally, the impact of rubbing and rinsing of Ad5 contaminated contact lenses with the solutions was studied. RESULTS Solutions based on povidone-iodine demonstrated a more than 3-log reduction in virus after 4 and 6 hours of incubation. In contrast, hydrogen peroxide only demonstrated a 0.52-log reduction after 6 hours of incubation. Increasing the contact time with hydrogen peroxide increased Ad5 inactivation, with a 2.18-log reduction after 6 hours of incubation with the solution prior to neutralization. Nonoxidative systems did not demonstrate a significant log reduction after 4 hours of incubation. However, rubbing and rinsing of contact lenses using the nonoxidative systems reduced the virus counts from contaminated contact lenses to below the limit of quantification. CONCLUSIONS Povidone-iodine solutions have a significant effect on reducing the viability of Ad5. Hydrogen peroxide care solutions are effective only if unneutralized contact time is increased. Nonoxidative systems can be effective in combating contaminated lenses only with the addition of a rub-and-rinse step.
Collapse
Affiliation(s)
- Christiane Lourenco Nogueira
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Scott Joseph Boegel
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Manish Shukla
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, Waterloo, Ontario, Canada
| | - William Ngo
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Alex Hui
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, Waterloo, Ontario, Canada
- School of Optometry and Vision Science, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Lyndon W. Jones
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Marc G. Aucoin
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
3
|
Doyle CA, Busey GW, Iobst WH, Kiessling V, Renken C, Doppalapudi H, Stremska ME, Manjegowda MC, Arish M, Wang W, Naphade S, Kennedy J, Bloyet LM, Thompson CE, Rothlauf PW, Stipes EJ, Whelan SPJ, Tamm LK, Kreutzberger AJB, Sun J, Desai BN. Endosomal fusion of pH-dependent enveloped viruses requires ion channel TRPM7. Nat Commun 2024; 15:8479. [PMID: 39353909 PMCID: PMC11445543 DOI: 10.1038/s41467-024-52773-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
The majority of viruses classified as pandemic threats are enveloped viruses which enter the cell through receptor-mediated endocytosis and take advantage of endosomal acidification to activate their fusion machinery. Here we report that the endosomal fusion of low pH-requiring viruses is highly dependent on TRPM7, a widely expressed TRP channel that is located on the plasma membrane and in intracellular vesicles. Using several viral infection systems expressing the envelope glycoproteins of various viruses, we find that loss of TRPM7 protects cells from infection by Lassa, LCMV, Ebola, Influenza, MERS, SARS-CoV-1, and SARS-CoV-2. TRPM7 ion channel activity is intrinsically necessary to acidify virus-laden endosomes but is expendable for several other endosomal acidification pathways. We propose a model wherein TRPM7 ion channel activity provides a countercurrent of cations from endosomal lumen to cytosol necessary to sustain the pumping of protons into these virus-laden endosomes. This study demonstrates the possibility of developing a broad-spectrum, TRPM7-targeting antiviral drug to subvert the endosomal fusion of low pH-dependent enveloped viruses.
Collapse
Affiliation(s)
- Catherine A Doyle
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Gregory W Busey
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Wesley H Iobst
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Volker Kiessling
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Chloe Renken
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Hansa Doppalapudi
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Marta E Stremska
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Mohan C Manjegowda
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Mohd Arish
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Weiming Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Nikegen Inc., Shanghai, China
| | - Shardul Naphade
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Joel Kennedy
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Louis-Marie Bloyet
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Cassandra E Thompson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul W Rothlauf
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Program in Virology, Harvard Medical School, Boston, MA, USA
| | - Eric J Stipes
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lukas K Tamm
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Alex J B Kreutzberger
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Boston Children's Hospital, Boston, MA, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Bimal N Desai
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA.
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
4
|
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
|
5
|
Pageau G, Levasseur M, Paniconi T, Jubinville E, Goulet-Beaulieu V, Boivin G, Jean J. The possibility of spreading herpes simplex virus type 1 via food handling and sharing. J Appl Microbiol 2023; 134:lxad224. [PMID: 37827542 DOI: 10.1093/jambio/lxad224] [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: 06/01/2023] [Revised: 08/11/2023] [Accepted: 10/11/2023] [Indexed: 10/14/2023]
Abstract
AIMS Herpes simplex virus type 1 (HSV-1) is an enveloped virus that causes recurrent and incurable diseases in 67% of the world population. Although it is not listed as a foodborne virus, some studies have shown that it can be recovered from surfaces as well as food. METHODS AND RESULTS We investigated its persistence at -20°C, 4°C, 20°C, or 37°C for up to 7 days on stainless steel, aluminum, glass, polypropylene, cheddar cheese, sliced almond, and apple skin and in cola soft drink, orange juice, coffee, and milk, as well as its transferability from stainless steel to dry or moistened nitrile or latex gloves over time at typical ambient temperatures. Based on the plaque assay on Vero cells, HSV-1 persisted at least 24 h on all surfaces and at least 1 h on food matrices but was inactivated quickly in cola soft drink. Temperature and pH affected HSV-1 infectivity. Transfer of HSV-1 at a contact pressure of 1 kg cm2-1 for 10 s occurred only on latex, especially moistened. CONCLUSIONS Our data on the persistence of HSV-1 on food-related surfaces suggest that some risk may be associated with sharing foods with infected carriers.
Collapse
Affiliation(s)
- Gabrielle Pageau
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec G1V 0A6, Canada
| | - Marianne Levasseur
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec G1V 0A6, Canada
| | - Teresa Paniconi
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec G1V 0A6, Canada
| | - Eric Jubinville
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec G1V 0A6, Canada
| | - Valérie Goulet-Beaulieu
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec G1V 0A6, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Quebec City, Quebec G1V 4G2, Canada
| | - Julie Jean
- Department of Food Sciences, Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec G1V 0A6, Canada
| |
Collapse
|
6
|
Cheng Z, Shang J, Wang H, Yu L, Yuan Z, Zhang Y, Du Y, Tian J. Molecular imaging-guided extracellular vesicle-based drug delivery for precise cancer management: Current status and future perspectives. J Control Release 2023; 362:97-120. [PMID: 37625599 DOI: 10.1016/j.jconrel.2023.08.040] [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: 05/18/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Extracellular vesicles (EVs), the mediators of intercellular communication, have attracted the attention of researchers for the important roles they play in cancer treatment. Compared with other inorganic nano-materials, EVs possess the advantages of higher biocompatibility, better physiochemical stability, easier surface modification, and excellent biosafety. They can be used as an advanced drug delivery system with an improved therapeutic index for various therapeutic agents. Engineered EV-based imaging and therapeutic agents (engineered EVs) have emerged as useful tools in targeted cancer diagnosis and therapy. Non-invasive tracing of engineered EVs contributes to a better evaluation of their functions in cancer progression, in vivo dynamic biodistribution, therapeutic response, and drug-loading efficiency. Recent advances in real-time molecular imaging (MI), and innovative EV labeling strategies have led to the development of novel tools that can evaluate the pharmacokinetics of engineered EVs in cancer management, which may accelerate further clinical translation of novel EV-based drug delivery platforms. Herein, we review the latest advances in EVs, their characteristics, and current examples of EV-based targeted drug delivery for cancer. Then, we discuss the prominent applications of MI for tracing both natural and engineered EVs. Finally, we discuss the current challenges and considerations of EVs in targeted cancer treatment and the limitations of different MI modalities. In the coming decades, EV-based therapeutic applications for cancer with improved drug loading and targeting abilities will be developed, and better anti-cancer effects of drug delivery nanoplatform will be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jihuan Shang
- School of Clinical Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Huarong Wang
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China
| | - Leyi Yu
- Beijing Haidian Hospital, Beijing 100080, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China.
| | - Yinlong Zhang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, China; Science and Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
7
|
Cadena-Cruz C, Villarreal Camacho JL, De Ávila-Arias M, Hurtado-Gomez L, Rodriguez A, San-Juan-Vergara H. Respiratory syncytial virus entry mechanism in host cells: A general overview. Mol Microbiol 2023; 120:341-350. [PMID: 37537859 DOI: 10.1111/mmi.15133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 08/05/2023]
Abstract
Respiratory syncytial virus (RSV) is a virus that causes acute respiratory infections in neonates and older adults. To infect host cells, the attachment glycoprotein (G) interacts with a cell surface receptor. This interaction determines the specific cell types that are susceptible to infection. RSV possesses a type I fusion protein F. Type I fusion proteins are metastable when rearrangement of the prefusion F occurs; the fusion peptide is exposed transforming the protein into postfusion form. The transition between the prefusion form and its postfusion form facilitates the viral envelope and the host cell membrane to fuse, enabling the virus to enter the host cell. Understanding the entry mechanism employed by RSV is crucial for developing effective antiviral therapies. In this review, we will discuss the various types of viral fusion proteins and explore the potential entry mechanisms utilized by RSV. A deeper understanding of these mechanisms will provide valuable insights for the development of novel approaches to treat RSV infections.
Collapse
Affiliation(s)
- C Cadena-Cruz
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
- Facultad de Ciencias de la Salud, Programa de Medicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - J L Villarreal Camacho
- Facultad de Ciencias de la Salud, Programa de Medicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Marcio De Ávila-Arias
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | - Leidy Hurtado-Gomez
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | - Alexander Rodriguez
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | | |
Collapse
|
8
|
Parida VK, Saidulu D, Bhatnagar A, Gupta AK, Afzal MS. A critical assessment of SARS-CoV-2 in aqueous environment: Existence, detection, survival, wastewater-based surveillance, inactivation methods, and effective management of COVID-19. CHEMOSPHERE 2023; 327:138503. [PMID: 36965534 PMCID: PMC10035368 DOI: 10.1016/j.chemosphere.2023.138503] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/08/2023] [Accepted: 03/22/2023] [Indexed: 06/01/2023]
Abstract
In early January 2020, the causal agent of unspecified pneumonia cases detected in China and elsewhere was identified as a novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and was the major cause of the COVID-19 outbreak. Later, the World Health Organization (WHO) proclaimed the COVID-19 pandemic a worldwide public health emergency on January 30, 2020. Since then, many studies have been published on this topic. In the present study, bibliometric analysis has been performed to analyze the research hotspots of the coronavirus. Coronavirus transmission, detection methods, potential risks of infection, and effective management practices have been discussed in the present review. Identification and quantification of SARS-CoV-2 viral loads in various water matrices have been reviewed. It was observed that the viral shedding through urine and feces of COVID-19-infected patients might be a primary mode of SARS-CoV-2 transmission in water and wastewater. In this context, the present review highlights wastewater-based epidemiology (WBE)/sewage surveillance, which can be utilized as an effective tool for tracking the transmission of COVID-19. This review also emphasizes the role of different disinfection techniques, such as chlorination, ultraviolet irradiation, and ozonation, for the inactivation of coronavirus. In addition, the application of computational modeling methods has been discussed for the effective management of COVID-19.
Collapse
Affiliation(s)
- Vishal Kumar Parida
- School of Environmental Science and Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Duduku Saidulu
- Department of Civil Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Amit Bhatnagar
- Department of Separation Science, LUT School of Engineering Science, LUT University, Sammonkatu 12, Mikkeli FI-50130, Finland.
| | - Ashok Kumar Gupta
- Department of Civil Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| | - Mohammad Saud Afzal
- Department of Civil Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| |
Collapse
|
9
|
Tarannum T, Ahmed S. Recent development in antiviral surfaces: Impact of topography and environmental conditions. Heliyon 2023; 9:e16698. [PMID: 37260884 PMCID: PMC10227326 DOI: 10.1016/j.heliyon.2023.e16698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023] Open
Abstract
The transmission of viruses is largely dependent on contact with contaminated virus-laden communal surfaces. While frequent surface disinfection and antiviral coating techniques are put forth by researchers as a plan of action to tackle transmission in dire situations like the Covid-19 pandemic caused by SARS-CoV-2 virus, these procedures are often laborious, time-consuming, cost-intensive, and toxic. Hence, surface topography-mediated antiviral surfaces have been gaining more attention in recent times. Although bioinspired hydrophobic antibacterial nanopatterned surfaces mimicking the natural sources is a very prevalent and successful strategy, the antiviral prospect of these surfaces is yet to be explored. Few recent studies have explored the potential of nanopatterned antiviral surfaces. In this review, we highlighted surface properties that have an impact on virus attachment and persistence, particularly focusing and emphasizing on the prospect of the nanotextured surface with enhanced properties to be used as antiviral surface. In addition, recent developments in surface nanopatterning techniques depending on the nano-scaled dimensions have been discussed. The impacts of environments and surface topology on virus inactivation have also been reviewed.
Collapse
Affiliation(s)
- Tanjina Tarannum
- Department of Chemical Engineering, Bangladesh University of Engineering and Technology, Dhaka-1000. Bangladesh
| | - Shoeb Ahmed
- Department of Chemical Engineering, Bangladesh University of Engineering and Technology, Dhaka-1000. Bangladesh
| |
Collapse
|
10
|
Antiviral Peptides in Antimicrobial Surface Coatings—From Current Techniques to Potential Applications. Viruses 2023; 15:v15030640. [PMID: 36992349 PMCID: PMC10051592 DOI: 10.3390/v15030640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
The transmission of pathogens through contact with contaminated surfaces is an important route for the spread of infections. The recent outbreak of COVID-19 highlights the necessity to attenuate surface-mediated transmission. Currently, the disinfection and sanitization of surfaces are commonly performed in this regard. However, there are some disadvantages associated with these practices, including the development of antibiotic resistance, viral mutation, etc.; hence, a better strategy is necessary. In recent years, peptides have been studied to be utilized as a potential alternative. They are part of the host immune defense and have many potential in vivo applications in drug delivery, diagnostics, immunomodulation, etc. Additionally, the ability of peptides to interact with different molecules and membrane surfaces of microorganisms has made it possible to exploit them in ex vivo applications such as antimicrobial (antibacterial and antiviral) coatings. Although antibacterial peptide coatings have been studied extensively and proven to be effective, antiviral coatings are a more recent development. Therefore, this study aims to highlight antiviral coating strategies and the current practices and application of antiviral coating materials in personal protective equipment, healthcare devices, and textiles and surfaces in public settings. Here, we have presented a review on potential techniques to incorporate peptides in current surface coating strategies that will serve as a guide for developing cost-effective, sustainable and coherent antiviral surface coatings. We further our discussion to highlight some challenges of using peptides as a surface coating material and to examine future perspectives.
Collapse
|
11
|
Karade SS, Franco EJ, Rojas AC, Hanrahan KC, Kolesnikov A, Yu W, MacKerell AD, Hill DC, Weber DJ, Brown AN, Treston AM, Mariuzza RA. Structure-Based Design of Potent Iminosugar Inhibitors of Endoplasmic Reticulum α-Glucosidase I with Anti-SARS-CoV-2 Activity. J Med Chem 2023; 66:2744-2760. [PMID: 36762932 PMCID: PMC10278443 DOI: 10.1021/acs.jmedchem.2c01750] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Enveloped viruses depend on the host endoplasmic reticulum (ER) quality control (QC) machinery for proper glycoprotein folding. The endoplasmic reticulum quality control (ERQC) enzyme α-glucosidase I (α-GluI) is an attractive target for developing broad-spectrum antivirals. We synthesized 28 inhibitors designed to interact with all four subsites of the α-GluI active site. These inhibitors are derivatives of the iminosugars 1-deoxynojirimycin (1-DNJ) and valiolamine. Crystal structures of ER α-GluI bound to 25 1-DNJ and three valiolamine derivatives revealed the basis for inhibitory potency. We established the structure-activity relationship (SAR) and used the Site Identification by Ligand Competitive Saturation (SILCS) method to develop a model for predicting α-GluI inhibition. We screened the compounds against SARS-CoV-2 in vitro to identify those with greater antiviral activity than the benchmark α-glucosidase inhibitor UV-4. These host-targeting compounds are candidates for investigation in animal models of SARS-CoV-2 and for testing against other viruses that rely on ERQC for correct glycoprotein folding.
Collapse
Affiliation(s)
- Sharanbasappa S. Karade
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Evelyn J. Franco
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA
| | - Ana C. Rojas
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA
| | - Kaley C. Hanrahan
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA
| | - Alexander Kolesnikov
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Wenbo Yu
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Computer-Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
- Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Alexander D. MacKerell
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Computer-Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
- Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | | | - David J. Weber
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Ashley N. Brown
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA
| | - Anthony M. Treston
- Emergent BioSolutions, Gaithersburg, MD 20879, USA
- Current address: Treadwell Therapeutics, Toronto M5G 2M9, Canada
| | - Roy A. Mariuzza
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
12
|
Choi W, Kim YH, Min J. Inhibition of Enveloped Virus Surrogate Phi6 Infection Using Yeast-Derived Vacuoles. Microbiol Spectr 2023; 11:e0266122. [PMID: 36688634 PMCID: PMC9927162 DOI: 10.1128/spectrum.02661-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The periodic emergence of infectious disease poses a serious threat to human life. Among the causative agents, including pathogenic bacteria and fungi, enveloped viruses have caused global pandemics. In the last 10 years, outbreaks of severe acute respiratory syndrome coronavirus 2 disease, severe acute respiratory syndrome, and Middle East respiratory syndrome have all been caused by enveloped viruses. Among several paths of secondary transmission, inhalation of aerosols containing saliva with sputum droplets from infected patients is the major path. To prevent these infectious diseases, mass use of antiviral agents is essential. The yeast-derived vacuole is a small organelle in which hydrolytic enzymes are concentrated. It is an intracellular organ with an excellent ability to process old organelles and bacteria and viruses that have invaded from the outside and can be present in sufficient quantity to be called a kind of enzyme bomb. We confirmed the inhibition of virus infection and structural collapse by vacuole treatment. Among several enzymes, proteases affected Phi6 infectivity. This study tried to isolate these vacuoles from yeast and use them as an antiviral agent for virus treatment, which is a recent issue. We confirmed that viral infectivity was inactivated, and structure collapsed through vacuole treatment. This paper is meaningful in that extracellularly isolated yeast-derived vacuoles are a first attempt to utilize vacuoles for viral treatment. IMPORTANCE The study assesses the vacuoles isolated from the yeast Saccharomyces cerevisiae as green antiviral agents to decrease the concerns about massive use of chemical antiviral agents and its side effects. To prevent the spreading of infectious diseases, personal or public use of antiviral agents is encouraged. The concern about the active compounds of these chemical antiviral agents has grown. Active compounds of antiviral agents have potential side effects on human health and the environment. Our proposed approach suggests effective and green antivirus material from a nonhazardous yeast strain. Also, large-scale production using a fermentation process can allow cost-effectiveness. The results showed sufficient reduced infectivity by vacuole treatment. The exposed vacuole can play the roles of both enzyme bomb to the virus and renewable nutrient source in the ecosystem.
Collapse
Affiliation(s)
- Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Jeonbuk, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, Cheongju, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Jeonbuk, South Korea
| |
Collapse
|
13
|
Bizzoca ME, Leuci S, Mignogna MD, Muzio EL, Caponio VCA, Muzio LL. Natural compounds may contribute in preventing SARS-CoV-2 infection: a narrative review. FOOD SCIENCE AND HUMAN WELLNESS 2022; 11:1134-1142. [PMID: 38621001 PMCID: PMC9160299 DOI: 10.1016/j.fshw.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronavirus pandemic infection is the most important health issue worldwide. Coronavirus disease 2019 is a contagious disease characterized by severe acute respiratory syndrome coronavirus 2. To date, excluding the possibility of vaccination, against SARS-CoV-2 infection it is possible to act only with supportive care and non-virus-specific treatments in order to improve the patient's symptoms. Pharmaceutical industry is investigating effects of medicinal plants, phytochemical extracts and aromatic herbs to find out natural substances which may act as antiviral drugs. Several studies have revealed how these substances may interfere with the viral life cycle, viral entry, replication, assembly or discharge, as well as virus-specific host targets or stimulating the host immune system, reducing oxidative stress and inflammatory response. A natural compound can be used as a prophylaxis by people professionally exposed to the risk of contagion and/or positive patients not in intensive care. The aim of this paper is to perform a narrative review of current literature in order to summarize the most studied natural compounds and their modes of action.
Collapse
Affiliation(s)
- Maria Eleonora Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
| | - Stefania Leuci
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Naples 80131, Italy
| | - Michele Davide Mignogna
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Naples 80131, Italy
| | - Eleonora Lo Muzio
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | | | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
- C.I.N.B.O. (Consorzio Interuniversitario Nazionale per la Bio-Oncologia), Chieti 66100, Italy
| |
Collapse
|
14
|
Mejía-Méndez JL, Vazquez-Duhalt R, Hernández LR, Sánchez-Arreola E, Bach H. Virus-like Particles: Fundamentals and Biomedical Applications. Int J Mol Sci 2022; 23:8579. [PMID: 35955711 PMCID: PMC9369363 DOI: 10.3390/ijms23158579] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
Nanotechnology is a fast-evolving field focused on fabricating nanoscale objects for industrial, cosmetic, and therapeutic applications. Virus-like particles (VLPs) are self-assembled nanoparticles whose intrinsic properties, such as heterogeneity, and highly ordered structural organization are exploited to prepare vaccines; imaging agents; construct nanobioreactors; cancer treatment approaches; or deliver drugs, genes, and enzymes. However, depending upon the intrinsic features of the native virus from which they are produced, the therapeutic performance of VLPs can vary. This review compiles the recent scientific literature about the fundamentals of VLPs with biomedical applications. We consulted different databases to present a general scenario about viruses and how VLPs are produced in eukaryotic and prokaryotic cell lines to entrap therapeutic cargo. Moreover, the structural classification, morphology, and methods to functionalize the surface of VLPs are discussed. Finally, different characterization techniques required to examine the size, charge, aggregation, and composition of VLPs are described.
Collapse
Affiliation(s)
- Jorge L. Mejía-Méndez
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología UNAM, Km 107 Carretera Tijuana-Ensenada, Ensenada 22860, Baja California, Mexico;
| | - Luis R. Hernández
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Eugenio Sánchez-Arreola
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
15
|
Adcock AF, Wang P, Ferguson IS, Obu SC, Sun YP, Yang L. Inactivation of Vesicular Stomatitis Virus with Light-Activated Carbon Dots and Mechanistic Implications. ACS APPLIED BIO MATERIALS 2022; 5:3158-3166. [PMID: 35797334 DOI: 10.1021/acsabm.2c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The prevention of viral transmission is an important step to address the spread of viral infections. Using the enveloped vesicular stomatitis virus (VSV) as a model, this study explored the antiviral functions of the specifically designed and prepared carbon dots (CDots). The CDots were prepared using small carbon nanoparticles with surface functionalization-passivation by oligomeric polyethylenimine (PEI). The results indicated that the PEI-CDots were readily activated by visible light to effectively and efficiently inactivate VSVs under various combinations of experimental conditions (viral titer, dot concentration, and treatment time). The photodynamically induced viral structural protein degradation and genomic RNA degradation were observed, suggesting the mechanistic origins, leading to the inactivation of virus. The results suggested CDots as a class of promising broad-spectrum antiviral agents for disinfection of viruses.
Collapse
Affiliation(s)
- Audrey F Adcock
- Biomanufacturing Research Institute and Technology Enterprise (BRITE) and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina 27707, United States
| | - Ping Wang
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Isaiah S Ferguson
- Biomanufacturing Research Institute and Technology Enterprise (BRITE) and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina 27707, United States
| | - Somtochukwu C Obu
- Biomanufacturing Research Institute and Technology Enterprise (BRITE) and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina 27707, United States
| | - Ya-Ping Sun
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Liju Yang
- Biomanufacturing Research Institute and Technology Enterprise (BRITE) and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina 27707, United States
| |
Collapse
|
16
|
Karade SS, Kolesnikov A, Treston AM, Mariuzza RA. Identification of Endoplasmic Reticulum α-Glucosidase I from a Thermophilic Fungus as a Platform for Structure-Guided Antiviral Drug Design. Biochemistry 2022; 61:822-832. [PMID: 35476408 DOI: 10.1021/acs.biochem.2c00092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
All viruses depend on host cell proteins for replication. Denying viruses' access to the function of critical host proteins can result in antiviral activity against multiple virus families. In particular, small-molecule drug candidates which inhibit the α-glucosidase enzymes of the endoplasmic reticulum (ER) translation quality control (QC) pathway have demonstrated broad-spectrum antiviral activities and low risk for development of viral resistance. However, antiviral drug discovery focused on the ERQC enzyme α-glucosidase I (α-GluI) has been hampered by difficulties in obtaining crystal structures of complexes with inhibitors. We report here the identification of an orthologous enzyme from a thermophilic fungus, Chaetomium thermophilum (Ct), as a robust surrogate for mammalian ER α-GluI and a platform for inhibitor design. Previously annotated only as a hypothetical protein, the Ct protein was validated as a bona fide α-glucosidase by comparing its crystal structure to that of mammalian α-GluI, by demonstrating enzymatic activity on the unusual α-d-Glcp-(1 → 2)-α-d-Glcp-(1 → 3) substrate glycan, and by showing that well-known inhibitors of mammalian α-GluI (1-DNJ, UV-4, UV-5) also inhibit Ct α-GluI. Crystal structures of Ct α-GluI in complex with three such inhibitors (UV-4, UV-5, EB-0159) revealed extensive interactions with all four enzyme subsites and provided insights into the catalytic mechanism. Identification of ER Ct α-GluI as a surrogate for mammalian α-GluI will accelerate the structure-guided discovery of broad-spectrum antivirals. This study also highlights Ct as a source of thermostable eukaryotic proteins, such as ER α-Glu I, that lack orthologs in bacterial or archaeal thermophiles.
Collapse
Affiliation(s)
- Sharanbasappa S Karade
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, United States
| | - Alexander Kolesnikov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, United States
| | | | - Roy A Mariuzza
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
17
|
Karade SS, Hill ML, Kiappes JL, Manne R, Aakula B, Zitzmann N, Warfield KL, Treston AM, Mariuzza RA. N-Substituted Valiolamine Derivatives as Potent Inhibitors of Endoplasmic Reticulum α-Glucosidases I and II with Antiviral Activity. J Med Chem 2021; 64:18010-18024. [PMID: 34870992 DOI: 10.1021/acs.jmedchem.1c01377] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Most enveloped viruses rely on the host cell endoplasmic reticulum (ER) quality control (QC) machinery for proper folding of glycoproteins. The key ER α-glucosidases (α-Glu) I and II of the ERQC machinery are attractive targets for developing broad-spectrum antivirals. Iminosugars based on deoxynojirimycin have been extensively studied as ER α-glucosidase inhibitors; however, other glycomimetic compounds are less established. Accordingly, we synthesized a series of N-substituted derivatives of valiolamine, the iminosugar scaffold of type 2 diabetes drug voglibose. To understand the basis for up to 100,000-fold improved inhibitory potency, we determined high-resolution crystal structures of mouse ER α-GluII in complex with valiolamine and 10 derivatives. The structures revealed extensive interactions with all four α-GluII subsites. We further showed that N-substituted valiolamines were active against dengue virus and SARS-CoV-2 in vitro. This study introduces valiolamine-based inhibitors of the ERQC machinery as candidates for developing potential broad-spectrum therapeutics against the existing and emerging viruses.
Collapse
Affiliation(s)
- Sharanbasappa S Karade
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, United States
| | - Michelle L Hill
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, U.K
| | - J L Kiappes
- Department of Chemistry, University College, London WC1H 0AJ, U.K
| | - Rajkumar Manne
- Sai Life Sciences Ltd., Hyderabad, 500032 Telangana, India
| | | | - Nicole Zitzmann
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, U.K
| | - Kelly L Warfield
- Emergent BioSolutions, Gaithersburg, Maryland 20879, United States
| | | | - Roy A Mariuzza
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
18
|
Anti-Herpes Simplex Virus Efficacy of Silk Cocoon, Silkworm Pupa and Non-Sericin Extracts. Antibiotics (Basel) 2021; 10:antibiotics10121553. [PMID: 34943765 PMCID: PMC8698825 DOI: 10.3390/antibiotics10121553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/25/2022] Open
Abstract
Herpes simplex virus (HSV) infections are prevalent worldwide and are the cause of life- threatening diseases. Standard treatment with antiviral drugs, such as acyclovir, could prevent serious complications; however, resistance has been reported specifically among immunocompromised patients. Therefore, the development of an alternative approach is needed. The silk cocoon derived from silkworm, Bombyx mori, has been recognized for its broad-spectrum biological activity, including antiviral activity; however, its effects against HSV infection are unknown. In this study, we investigated the inhibitory effects of silk extracts derived from the cocoon shell, silk cocoon, silkworm pupa and non-sericin extract, on blocking HSV-1 and HSV-2 binding to host cells, resulting in the inhibition of the virus infection in Vero cells. Non-sericin extract demonstrated the greatest effectiveness on inhibiting HSV-1 and HSV-2 binding activity. Moreover, the virucidal effect to inactivate HSV-1 and HSV-2 was determined and revealed that non-sericin extract also exerted the highest potential activity. Using the treatment of non-sericin extract in HSV-2-infected HeLa cells could significantly lower the HSV-induced cell death and prevent inflammation via lowering the inflammatory cytokine gene expression. The non-sericin extract was analyzed for its bioactive compounds in which gallic acid, flavonoid and xanthophyll were identified, and might have partially contributed to its antiviral activity. The finding in our study suggested the potential of silk extract as an alternative therapeutic treatment for HSV infection.
Collapse
|
19
|
Park S, Tae H, Cho NJ. Biophysical Measurement Strategies for Antiviral Drug Development: Recent Progress in Virus-Mimetic Platforms Down to the Single Particle Level. Acc Chem Res 2021; 54:3204-3214. [PMID: 34346210 DOI: 10.1021/acs.accounts.1c00300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The rapid growth in the global human population has increased the prevalence of emerging infectious diseases, which poses a major risk to public health. In search of effective clinical solutions, the acquisition of knowledge and understanding of biomolecular processes associated with viral pathogens represents a prerequisite. In this context, biophysical engineering approaches are particularly promising since they can resolve biomolecular interactions systematically by circumventing the complexities associated with experiments involving natural biological systems. The engineering approaches encompass the design and construction of biomimetic platforms that simulate the physiological system. This approach enables us to characterize, measure, and quantitatively analyze biomolecular interactions.In this Account, we summarize biophysical measurements that our group has successfully adopted to develop broad-spectrum antiviral drugs based on the lipid envelope antiviral disruption (LEAD) strategy, targeting the structural integrity of the outer viral membrane to abrogate viral infectivity. We particularly focus on the engineering aspects related to the design and construction of the tethered lipid vesicle platform, which closely mimics the viral membrane. We first outline the development of the LEAD agents screening platform that integrates soft matter design components with biomaterials and surface functionalization strategies to facilitate parallel measurements tracking peptide-induced destabilization of nanoscale, virus-mimicking vesicles with tunable size and composition. Then, we describe how this platform can be effectively employed to gain insights into the membrane curvature dependency of certain peptides. The fundamental knowledge acquired through this systematic process is crucial in the identification and subsequent development of antiviral drug candidates. In particular, we highlight the development of curvature-sensitive α-helical (AH) peptides as a broad-spectrum antiviral agent that has been demonstrated as an effective therapeutic treatment against multiple enveloped viruses. Also, we introduce a tethered cluster of vesicles to mimic clusters of enveloped viruses, exhibiting higher infectivity levels in the biological system. Then, we discuss key considerations, including experimental artifacts, namely dye leakage and imaging-related photobleaching, and corresponding corrective measures to improve the accuracy of quantitative interpretation. With the ongoing development and application of the tethered lipid vesicle platform, there is a compelling opportunity to explore fundamental biointerfacial science and develop a new class of broad-spectrum antiviral agents to prepare for the future membrane-enveloped viral pandemics.
Collapse
Affiliation(s)
- Soohyun Park
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Hyunhyuk Tae
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| |
Collapse
|
20
|
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
|
21
|
Li G, Su B, Fu P, Bai Y, Ding G, Li D, Wang J, Yang G, Chu B. NPC1-regulated dynamic of clathrin-coated pits is essential for viral entry. SCIENCE CHINA-LIFE SCIENCES 2021; 65:341-361. [PMID: 34047913 PMCID: PMC8160554 DOI: 10.1007/s11427-021-1929-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022]
Abstract
Viruses utilize cellular lipids and manipulate host lipid metabolism to ensure their replication and spread. Therefore, the identification of lipids and metabolic pathways that are suitable targets for antiviral development is crucial. Using a library of compounds targeting host lipid metabolic factors and testing them for their ability to block pseudorabies virus (PRV) and vesicular stomatitis virus (VSV) infection, we found that U18666A, a specific inhibitor of Niemann-Pick C1 (NPC1), is highly potent in suppressing the entry of diverse viruses including pseudotyped severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). NPC1 deficiency markedly attenuates viral growth by decreasing cholesterol abundance in the plasma membrane, thereby inhibiting the dynamics of clathrin-coated pits (CCPs), which are indispensable for clathrin-mediated endocytosis. Significantly, exogenous cholesterol can complement the dynamics of CCPs, leading to efficient viral entry and infectivity. Administration of U18666A improves the survival and pathology of PRV- and influenza A virus-infected mice. Thus, our studies demonstrate a unique mechanism by which NPC1 inhibition achieves broad antiviral activity, indicating a potential new therapeutic strategy against SARS-CoV-2, as well as other emerging viruses.
Collapse
Affiliation(s)
- Guoli Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
| | - Bingqian Su
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
| | - Pengfei Fu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
| | - Yilin Bai
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Guangxu Ding
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
| | - Dahua Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Guoyu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China.
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Beibei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China.
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
22
|
Fu Y, Jaarsma AH, Kuipers OP. Antiviral activities and applications of ribosomally synthesized and post-translationally modified peptides (RiPPs). Cell Mol Life Sci 2021; 78:3921-3940. [PMID: 33532865 PMCID: PMC7853169 DOI: 10.1007/s00018-021-03759-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/15/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022]
Abstract
The emergence and re-emergence of viral epidemics and the risks of antiviral drug resistance are a serious threat to global public health. New options to supplement or replace currently used drugs for antiviral therapy are urgently needed. The research in the field of ribosomally synthesized and post-translationally modified peptides (RiPPs) has been booming in the last few decades, in particular in view of their strong antimicrobial activities and high stability. The RiPPs with antiviral activity, especially those against enveloped viruses, are now also gaining more interest. RiPPs have a number of advantages over small molecule drugs in terms of specificity and affinity for targets, and over protein-based drugs in terms of cellular penetrability, stability and size. Moreover, the great engineering potential of RiPPs provides an efficient way to optimize them as potent antiviral drugs candidates. These intrinsic advantages underscore the good therapeutic prospects of RiPPs in viral treatment. With the aim to highlight the underrated antiviral potential of RiPPs and explore their development as antiviral drugs, we review the current literature describing the antiviral activities and mechanisms of action of RiPPs, discussing the ongoing efforts to improve their antiviral potential and demonstrate their suitability as antiviral therapeutics. We propose that antiviral RiPPs may overcome the limits of peptide-based antiviral therapy, providing an innovative option for the treatment of viral disease.
Collapse
Affiliation(s)
- Yuxin Fu
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Ate H Jaarsma
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
- Department of Environmental Science, Aarhus University, 4000, Roskilde, Denmark
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
23
|
Mohan SV, Hemalatha M, Kopperi H, Ranjith I, Kumar AK. SARS-CoV-2 in environmental perspective: Occurrence, persistence, surveillance, inactivation and challenges. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 405:126893. [PMID: 32901196 PMCID: PMC7471803 DOI: 10.1016/j.cej.2020.126893] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 05/03/2023]
Abstract
The unprecedented global spread of the severe acute respiratory syndrome (SARS) caused by SARS-CoV-2 is depicting the distressing pandemic consequence on human health, economy as well as ecosystem services. So far novel coronavirus (CoV) outbreaks were associated with SARS-CoV-2 (2019), middle east respiratory syndrome coronavirus (MERS-CoV, 2012), and SARS-CoV-1 (2003) events. CoV relates to the enveloped family of Betacoronavirus (βCoV) with positive-sense single-stranded RNA (+ssRNA). Knowing well the persistence, transmission, and spread of SARS-CoV-2 through proximity, the faecal-oral route is now emerging as a major environmental concern to community transmission. The replication and persistence of CoV in the gastrointestinal (GI) tract and shedding through stools is indicating a potential transmission route to the environment settings. Despite of the evidence, based on fewer reports on SARS-CoV-2 occurrence and persistence in wastewater/sewage/water, the transmission of the infective virus to the community is yet to be established. In this realm, this communication attempted to review the possible influx route of the enteric enveloped viral transmission in the environmental settings with reference to its occurrence, persistence, detection, and inactivation based on the published literature so far. The possibilities of airborne transmission through enteric virus-laden aerosols, environmental factors that may influence the viral transmission, and disinfection methods (conventional and emerging) as well as the inactivation mechanism with reference to the enveloped virus were reviewed. The need for wastewater epidemiology (WBE) studies for surveillance as well as for early warning signal was elaborated. This communication will provide a basis to understand the SARS-CoV-2 as well as other viruses in the context of the environmental engineering perspective to design effective strategies to counter the enteric virus transmission and also serves as a working paper for researchers, policy makers and regulators.
Collapse
Key Words
- (h+), Photoholes
- +ssRNA, Positive Sense Single-Stranded RNA
- A-WWTS, Algal-WWTS
- ACE2, Angiotensin-converting enzyme 2
- AH, Absolute Humidity
- AOPs, Advanced Oxidation Processes
- ASP, Activate Sludge Process
- Aerosols
- BCoV, Bovine Enteric Coronavirus)
- BSL, Biosafety Level
- BVDV1, Bovine Viral Diarrhea Virus Type 1
- BVDV2, Bovine Viral Diarrhea Virus Type 2
- BoRv, Bovine Rotavirus Group A
- CCA, Carbon Covered Alumina
- CNT, Carbon Nanotubes
- COVID-19
- COVID-19, Coronavirus Disease 2019
- CRFK, Crandell Reese feline kidney cell line (CRFK)
- CVE, Coxsackievirus B5
- ClO2, Chlorine dioxide
- Cl−, Chlorine
- Cys, Cysteine
- DBP, Disinfection by-products
- DBT, L2 and Delayed Brain Tumor Cell Cultures
- DMEM, Dulbecco’s Modified Eagle Medium
- DNA, deoxyribose nucleic acid
- Disinfection
- E gene, Envelope protein gene
- EV, Echovirus 11
- Enteric virus
- Enveloped virus
- FC, Free Chlorine
- FFP3, Filtering Face Piece
- FIPV, Feline infectious peritonitis virus
- GI, Gastrointestinal tract
- H2O2, Hydrogen Peroxide
- H3N2, InfluenzaA
- H6N2, Avian influenza virus
- HAV, Hepatitis A virus (HAV)
- HAdV, Human Adenovirus
- HCoV, Human CoV
- HEV, Hepatitis E virus
- HKU1, Human CoV1
- ICC-PCR, Integrated Cell Culture with PCR
- JCV, JCV polyomavirus
- MALDI-TOF MS, Mass Spectrometry
- MBR, Membrane Bioreactor (MBR)
- MERS-CoV, Middle East Respiratory Syndrome Coronavirus
- MHV, Murine hepatitis virus
- MNV-1, Murine Norovirus
- MWCNTs, Multiwalled Carbon Nanotubes
- Met, Methionine
- N gene, Nucleocapsid protein gene
- NCoV, Novel coronavirus
- NGS, Next generation sequencing
- NTP, Non-Thermal Plasma
- O2, Singlet Oxygen
- O3, Ozone
- ORF, Open Reading Frame
- PAA, Para Acetic Acid
- PCR, Polymerase Chain Reaction
- PEC, Photoelectrocatalytical
- PEG, Polyethylene Glycol
- PFU, Plaque Forming Unit
- PMMoV, Pepper Mild Mottle Virus
- PMR, Photocatalytic Membrane Reactors
- PPE, Personal Protective Equipment
- PTAF, Photocatalytic Titanium Apatite Filter
- PV-1, Polivirus-1
- PV-3, Poliovirus 3
- PVDF, Polyvinylidene Fluoride
- Qβ, bacteriophages
- RH, Relative Humidity
- RNA, Ribose nucleic acid
- RONS, Reactive Oxygen and/or Nitrogen Species
- RT-PCR, Real Time Polymerase Chain Reaction
- RVA, Rotaviruses A
- SARS-CoV-1, Severe Acute Respiratory Syndrome Coronavirus 1
- SARS-CoV-2, Severe Acute Respiratory Syndrome Coronavirus 2
- SBR, Sequential Batch Reactor
- SODIS, Solar water disinfection
- STP, Sewage Treatment Plant
- Sewage
- T90, First order reaction time required for completion of 90%
- T99.9, First order reaction time required for completion of 99.9%
- TGEV, Porcine Coronavirus Transmissible Gastroenteritis Virus
- TGEV, Transmissible Gastroenteritis
- Trp, Tryptophan
- Tyr, Tyrosine
- US-EPA, United States Environmental Protection Agency
- UV, Ultraviolet
- WBE, Wastewater-Based Epidemiology
- WWT, Wastewater Treatment
- WWTPs, Wastewater Treatment Plants
- dPCR, Digital PCR
- ds, Double Stranded
- dsDNA, Double Stranded DNA
- log10, logarithm with base 10
- qRT-PCR, quantitative RT-PCR
- ss, Single Stranded
- ssDNA, Single Stranded DNA
- ssRNA, Single Stranded RNA
- αCoV, Alphacoronavirus
- βCoV, Betacoronavirus
Collapse
Affiliation(s)
- S Venkata Mohan
- Bioengineering and Environmental Sciences Lab, Department of Energy and Environmental Engineering (DEEE), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Campus, Hyderabad 500007, India
| | - Manupati Hemalatha
- Bioengineering and Environmental Sciences Lab, Department of Energy and Environmental Engineering (DEEE), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Campus, Hyderabad 500007, India
| | - Harishankar Kopperi
- Bioengineering and Environmental Sciences Lab, Department of Energy and Environmental Engineering (DEEE), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
| | - I Ranjith
- Bioengineering and Environmental Sciences Lab, Department of Energy and Environmental Engineering (DEEE), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
| | - A Kiran Kumar
- Bioengineering and Environmental Sciences Lab, Department of Energy and Environmental Engineering (DEEE), CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Dispensary, Hyderabad 500007, India
| |
Collapse
|
24
|
Yoon BK, Jeon WY, Sut TN, Cho NJ, Jackman JA. Stopping Membrane-Enveloped Viruses with Nanotechnology Strategies: Toward Antiviral Drug Development and Pandemic Preparedness. ACS NANO 2021; 15:125-148. [PMID: 33306354 DOI: 10.1021/acsnano.0c07489] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Membrane-enveloped viruses are a leading cause of viral epidemics, and there is an outstanding need to develop broad-spectrum antiviral strategies to treat and prevent enveloped virus infections. In this review, we critically discuss why the lipid membrane surrounding enveloped virus particles is a promising antiviral target and cover the latest progress in nanotechnology research to design and evaluate membrane-targeting virus inhibition strategies. These efforts span diverse topics such as nanomaterials, self-assembly, biosensors, nanomedicine, drug delivery, and medical devices and have excellent potential to support the development of next-generation antiviral drug candidates and technologies. Application examples in the areas of human medicine and agricultural biosecurity are also presented. Looking forward, research in this direction is poised to strengthen capabilities for virus pandemic preparedness and demonstrates how nanotechnology strategies can help to solve global health challenges related to infectious diseases.
Collapse
Affiliation(s)
- Bo Kyeong Yoon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Won-Yong Jeon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Tun Naw Sut
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Joshua A Jackman
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
25
|
Guha S, Bhaumik SR. Viral regulation of mRNA export with potentials for targeted therapy. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1864:194655. [PMID: 33246183 DOI: 10.1016/j.bbagrm.2020.194655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/15/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
Eukaryotic gene expression begins with transcription in the nucleus to synthesize mRNA (messenger RNA), which is subsequently exported to the cytoplasm for translation to protein. Like transcription and translation, mRNA export is an important regulatory step of eukaryotic gene expression. Various factors are involved in regulating mRNA export, and thus gene expression. Intriguingly, some of these factors interact with viral proteins, and such interactions interfere with mRNA export of the host cell, favoring viral RNA export. Hence, viruses hijack host mRNA export machinery for export of their own RNAs from nucleus to cytoplasm for translation to proteins for viral life cycle, suppressing host mRNA export (and thus host gene expression and immune/antiviral response). Therefore, the molecules that can impair the interactions of these mRNA export factors with viral proteins could emerge as antiviral therapeutic agents to suppress viral RNA transport and enhance host mRNA export, thereby promoting host gene expression and immune response. Thus, there has been a number of studies to understand how virus hijacks mRNA export machinery in suppressing host gene expression and promoting its own RNA export to the cytoplasm for translation to proteins required for viral replication/assembly/life cycle towards developing targeted antiviral therapies, as concisely described here.
Collapse
Affiliation(s)
- Shalini Guha
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | - Sukesh R Bhaumik
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA.
| |
Collapse
|
26
|
Hajjo R, Tropsha A. A Systems Biology Workflow for Drug and Vaccine Repurposing: Identifying Small-Molecule BCG Mimics to Reduce or Prevent COVID-19 Mortality. Pharm Res 2020; 37:212. [PMID: 33025261 PMCID: PMC7537965 DOI: 10.1007/s11095-020-02930-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Coronavirus disease 2019 (COVID-19) is expected to continue to cause worldwide fatalities until the World population develops 'herd immunity', or until a vaccine is developed and used as a prevention. Meanwhile, there is an urgent need to identify alternative means of antiviral defense. Bacillus Calmette-Guérin (BCG) vaccine that has been recognized for its off-target beneficial effects on the immune system can be exploited to boast immunity and protect from emerging novel viruses. METHODS We developed and employed a systems biology workflow capable of identifying small-molecule antiviral drugs and vaccines that can boast immunity and affect a wide variety of viral disease pathways to protect from the fatal consequences of emerging viruses. RESULTS Our analysis demonstrates that BCG vaccine affects the production and maturation of naïve T cells resulting in enhanced, long-lasting trained innate immune responses that can provide protection against novel viruses. We have identified small-molecule BCG mimics, including antiviral drugs such as raltegravir and lopinavir as high confidence hits. Strikingly, our top hits emetine and lopinavir were independently validated by recent experimental findings that these compounds inhibit the growth of SARS-CoV-2 in vitro. CONCLUSIONS Our results provide systems biology support for using BCG and small-molecule BCG mimics as putative vaccine and drug candidates against emergent viruses including SARS-CoV-2.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy - Computational Chemical Biology, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan.
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy, UNC Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| |
Collapse
|
27
|
Hensel A, Bauer R, Heinrich M, Spiegler V, Kayser O, Hempel G, Kraft K. Challenges at the Time of COVID-19: Opportunities and Innovations in Antivirals from Nature. PLANTA MEDICA 2020; 86:659-664. [PMID: 32434254 PMCID: PMC7356065 DOI: 10.1055/a-1177-4396] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 05/03/2023]
Abstract
As viral infections are an increasing threat to human societies, the need for new therapeutic strategies is becoming even more obvious. As no vaccine is available for COVID-19, the development of directly acting antiviral agents and preventive strategies have to be considered. Nature provides a huge reservoir of anti-infectious compounds, from which we can deduce innovative ideas, therapies, and products. Anti-adhesive natural products interact with the receptor-mediated recognition and early interaction of viruses with the host cells, leading to a reduced internalisation of the virus and reduced infections (e.g., procyanidin-B-2-di-O-gallate against influenza and herpes virus). Lignans like podophyllotoxin and bicyclol show strong antiviral activities against different viruses, and essential oils can directly interact with viral membranes and reduce the host's inflammatory responses (e.g., 1,8-cineol). Echinacea extracts stimulate the immune system, and bioavailable alkamides are key players by interacting with immunomodulating cannabinoid receptors. COVID-19 and SARS-CoV-2 infections have, in part, successfully been treated in China by preparations from traditional Chinese medicine and, while it is too early to draw conclusions, some promising data are available. There is huge potential, but intensified research is needed to develop evidence-based medicines with a clearly defined chemical profile. Intensified research and development, and therefore funding, are needed for exploiting nature's reservoir against viral infections. Combined action for basic research, chemistry, pharmacognosy, virology, and clinical studies, but also supply chain, sustainable sourcing, and economic aspects have to be considered. This review calls for intensified innovative science on natural products for the patients and for a healthier world!
Collapse
Affiliation(s)
- Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Michael Heinrich
- Pharmacognosy and Phytotherapy, UCL School of Pharmacy, London, UK
| | - Verena Spiegler
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Oliver Kayser
- Technical Biochemistry, TU Dortmund University, Dortmund, Germany
| | - Georg Hempel
- Institute of Pharmaceutical and Medical Chemistry, Clinical Pharmacy, University of Münster, Münster, Germany
| | - Karin Kraft
- University Medicine Rostock, Chair of Complementary Medicine, Rostock, Germany
| |
Collapse
|
28
|
Hachim MY, Al Heialy S, Hachim IY, Halwani R, Senok AC, Maghazachi AA, Hamid Q. Interferon-Induced Transmembrane Protein (IFITM3) Is Upregulated Explicitly in SARS-CoV-2 Infected Lung Epithelial Cells. Front Immunol 2020; 11:1372. [PMID: 32595654 PMCID: PMC7301886 DOI: 10.3389/fimmu.2020.01372] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/28/2020] [Indexed: 12/01/2022] Open
Abstract
Current guidelines for COVID-19 management recommend the utilization of various repurposed drugs. Despite ongoing research toward the development of a vaccine against SARS-CoV-2, such a vaccine will not be available in time to contribute to the containment of the ongoing pandemic. Therefore, there is an urgent need to develop a framework for the rapid identification of novel targets for diagnostic and therapeutic interventions. We analyzed publicly available transcriptomic datasets of SARS-CoV infected humans and mammals to identify consistent differentially expressed genes then validated in SARS-CoV-2 infected epithelial cells transcriptomic datasets. Comprehensive toxicogenomic analysis of the identified genes to identify possible interactions with clinically proven drugs was carried out. We identified IFITM3 as an early upregulated gene, and valproic acid was found to enhance its mRNA expression as well as induce its antiviral action. These findings indicate that analysis of publicly available transcriptomic and toxicogenomic data represents a rapid approach for the identification of novel targets and molecules that can modify the action of such targets during the early phases of emerging infections like COVID-19.
Collapse
Affiliation(s)
- Mahmood Yaseen Hachim
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Saba Al Heialy
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Ibrahim Yaseen Hachim
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Abiola C. Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Azzam A. Maghazachi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
29
|
Cagno V, Tintori C, Civra A, Cavalli R, Tiberi M, Botta L, Brai A, Poli G, Tapparel C, Lembo D, Botta M. Novel broad spectrum virucidal molecules against enveloped viruses. PLoS One 2018; 13:e0208333. [PMID: 30532192 PMCID: PMC6285983 DOI: 10.1371/journal.pone.0208333] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/06/2018] [Indexed: 01/01/2023] Open
Abstract
Viral infections are an important cause of death worldwide. Unfortunately, there is still a lack of antiviral drugs or vaccines for a large number of viruses, and this represents a remarkable challenge particularly for emerging and re-emerging viruses. For this reason, the identification of broad spectrum antiviral compounds provides a valuable opportunity for developing efficient antiviral therapies. Here we report on a class of rhodanine and thiobarbituric derivatives displaying a broad spectrum antiviral activity against seven different enveloped viruses including an HSV-2 acyclovir resistant strain with favorable selectivity indexes. Due to their selective action on enveloped viruses and to their lipid oxidation ability, we hypothesize a mechanism on the viral envelope that affects the fluidity of the lipid bilayer, thus compromising the efficiency of virus-cell fusion and preventing viral entry.
Collapse
Affiliation(s)
- Valeria Cagno
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
- Department of Molecular Microbiology, University of Geneva, Geneva, Switzerland
| | - Cristina Tintori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Andrea Civra
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Marika Tiberi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lorenzo Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Annalaura Brai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Lead Discovery Siena S.r.l., Castelnuovo Berardenga, Siena, Italy
| | - Giulio Poli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Caroline Tapparel
- Department of Molecular Microbiology, University of Geneva, Geneva, Switzerland
| | - David Lembo
- Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Lead Discovery Siena S.r.l., Castelnuovo Berardenga, Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
30
|
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
|
31
|
Edagwa BJ, Gendelman HE. Antimicrobials: Broad-spectrum antivirals. NATURE MATERIALS 2018; 17:114-116. [PMID: 29358769 DOI: 10.1038/nmat5064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Affiliation(s)
- Benson J Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| |
Collapse
|
32
|
A Simple Platform for the Rapid Development of Antimicrobials. Sci Rep 2017; 7:17610. [PMID: 29242618 PMCID: PMC5730575 DOI: 10.1038/s41598-017-17941-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/29/2017] [Indexed: 11/09/2022] Open
Abstract
Recent infectious outbreaks highlight the need for platform technologies that can be quickly deployed to develop therapeutics needed to contain the outbreak. We present a simple concept for rapid development of new antimicrobials. The goal was to produce in as little as one week thousands of doses of an intervention for a new pathogen. We tested the feasibility of a system based on antimicrobial synbodies. The system involves creating an array of 100 peptides that have been selected for broad capability to bind and/or kill viruses and bacteria. The peptides are pre-screened for low cell toxicity prior to large scale synthesis. Any pathogen is then assayed on the chip to find peptides that bind or kill it. Peptides are combined in pairs as synbodies and further screened for activity and toxicity. The lead synbody can be quickly produced in large scale, with completion of the entire process in one week.
Collapse
|
33
|
Carravilla P, Cruz A, Martin-Ugarte I, Oar-Arteta IR, Torralba J, Apellaniz B, Pérez-Gil J, Requejo-Isidro J, Huarte N, Nieva JL. Effects of HIV-1 gp41-Derived Virucidal Peptides on Virus-like Lipid Membranes. Biophys J 2017; 113:1301-1310. [PMID: 28797705 DOI: 10.1016/j.bpj.2017.06.061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/22/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022] Open
Abstract
Membrane fusion induced by the envelope glycoprotein enables the intracellular replication of HIV-1; hence, this process constitutes a major target for antiretroviral compounds. It has been proposed that peptides having propensity to interact with membrane interfaces might exert broad antiviral activity against enveloped viruses. To test this hypothesis, in this contribution we have analyzed the antiviral effects of peptides derived from the membrane-proximal external region and the transmembrane domain of the envelope glycoprotein subunit gp41, which display different degrees of interfacial hydrophobicity. Our data support the virucidal activity of a region that combines hydrophobic-at-interface membrane-proximal external region aromatics with hydrophobic residues of the transmembrane domain, and contains the absolutely conserved 679LWYIK/R683 sequence, proposed to embody a "cholesterol recognition/interaction amino acid consensus" motif. We further sought to correlate the antiviral activity of these peptides and their effects on membranes that mimic lipid composition and biophysical properties of the viral envelope. The data revealed that peptides endowed with virucidal activity were membrane active and induced permeabilization and fusion of virus-like lipid vesicles. In addition, they modulated lipid packing and miscibility of laterally segregated liquid domains, two properties that depend on the high cholesterol content of the viral membrane. Thus, the overall experimental evidence is consistent with a pattern of HIV inhibition that involves direct alteration of the physical chemistry of the virus membrane. Furthermore, the sequence-dependent effects observed might guide the development of new virucidal peptides.
Collapse
Affiliation(s)
- Pablo Carravilla
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Antonio Cruz
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid, Spain; Healthcare Research Institute of Hospital 12 de Octubre, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Itziar Martin-Ugarte
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Itziar R Oar-Arteta
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Johanna Torralba
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Beatriz Apellaniz
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Jesús Pérez-Gil
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid, Spain; Healthcare Research Institute of Hospital 12 de Octubre, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - José Requejo-Isidro
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Nerea Huarte
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - José L Nieva
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| |
Collapse
|
34
|
Khandelwal N, Chander Y, Rawat KD, Riyesh T, Nishanth C, Sharma S, Jindal N, Tripathi BN, Barua S, Kumar N. Emetine inhibits replication of RNA and DNA viruses without generating drug-resistant virus variants. Antiviral Res 2017. [PMID: 28624461 DOI: 10.1016/j.antiviral.2017.06.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
At a noncytotoxic concentration, emetine was found to inhibit replication of DNA viruses [buffalopoxvirus (BPXV) and bovine herpesvirus 1 (BHV-1)] as well as RNA viruses [peste des petits ruminants virus (PPRV) and Newcastle disease virus (NDV)]. Using the time-of-addition and virus step-specific assays, we showed that emetine treatment resulted in reduced synthesis of viral RNA (PPRV and NDV) and DNA (BPXV and BHV-1) as well as inhibiting viral entry (NDV and BHV-1). In addition, emetine treatment also resulted in decreased synthesis of viral proteins. In a cell free endogenous viral polymerase assay, emetine was found to significantly inhibit replication of NDV, but not BPXV genome, suggesting that besides directly inhibiting specific viral polymerases, emetine may also target other factors essentially required for efficient replication of the viral genome. Moreover, emetine was found to significantly inhibit BPXV-induced pock lesions on chorioallantoic membrane (CAM) along with associated mortality of embryonated chicken eggs. At a lethal dose 50 (LD50) of 126.49 ng/egg and at an effective concentration 50 (EC50) of 3.03 ng/egg, the therapeutic index of the emetine against BPXV was determined to be 41.74. Emetine was also found to significantly delay NDV-induced mortality in chicken embryos associated with reduced viral titers. Further, emetine-resistant mutants were not observed upon long-term (P = 25) sequential passage of BPXV and NDV in cell culture. Collectively, we have extended the effective antiviral activity of emetine against diverse groups of DNA and RNA viruses and propose that emetine could provide significant therapeutic value against some of these viruses without inducing an antiviral drug-resistant phenotype.
Collapse
Affiliation(s)
- Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Krishan Dutt Rawat
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Thachamvally Riyesh
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India
| | - Chikkahonnaiah Nishanth
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Shalini Sharma
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Naresh Jindal
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Bhupendra N Tripathi
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India.
| |
Collapse
|
35
|
González-González E, Alvarez MM, Márquez-Ipiña AR, Santiago GTD, Rodríguez-Martínez LM, Annabi N, Khademhosseini A. Anti-Ebola therapies based on monoclonal antibodies: current state and challenges ahead. Crit Rev Biotechnol 2017; 37:53-68. [PMID: 26611830 PMCID: PMC5568563 DOI: 10.3109/07388551.2015.1114465] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The 2014 Ebola outbreak, the largest recorded, took us largely unprepared, with no available vaccine or specific treatment. In this context, the World Health Organization declared that the humanitarian use of experimental therapies against Ebola Virus (EBOV) is ethical. In particular, an experimental treatment consisting of a cocktail of three monoclonal antibodies (mAbs) produced in tobacco plants and specifically directed to the EBOV glycoprotein (GP) was tested in humans, apparently with good results. Several mAbs with high affinity to the GP have been described. This review discusses our current knowledge on this topic. Particular emphasis is devoted to those mAbs that have been assayed in animal models or humans as possible therapies against Ebola. Engineering aspects and challenges for the production of anti-Ebola mAbs are also briefly discussed; current platforms for the design and production of full-length mAbs are cumbersome and costly.
Collapse
Affiliation(s)
- E González-González
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, Ave. Eugenio Garza Sada 2501 Sur Col. Tecnológico, CP 64849, Monterrey, Nuevo León, México
| | - MM Alvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, Ave. Eugenio Garza Sada 2501 Sur Col. Tecnológico, CP 64849, Monterrey, Nuevo León, México
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
| | - AR Márquez-Ipiña
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, Ave. Eugenio Garza Sada 2501 Sur Col. Tecnológico, CP 64849, Monterrey, Nuevo León, México
| | - G Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, Ave. Eugenio Garza Sada 2501 Sur Col. Tecnológico, CP 64849, Monterrey, Nuevo León, México
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
| | - LM Rodríguez-Martínez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, Ave. Eugenio Garza Sada 2501 Sur Col. Tecnológico, CP 64849, Monterrey, Nuevo León, México
| | - N Annabi
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115
| | - A Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, MA, USA
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
36
|
Caputo AT, Alonzi DS, Marti L, Reca IB, Kiappes JL, Struwe WB, Cross A, Basu S, Lowe ED, Darlot B, Santino A, Roversi P, Zitzmann N. Structures of mammalian ER α-glucosidase II capture the binding modes of broad-spectrum iminosugar antivirals. Proc Natl Acad Sci U S A 2016; 113:E4630-8. [PMID: 27462106 PMCID: PMC4987793 DOI: 10.1073/pnas.1604463113] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The biosynthesis of enveloped viruses depends heavily on the host cell endoplasmic reticulum (ER) glycoprotein quality control (QC) machinery. This dependency exceeds the dependency of host glycoproteins, offering a window for the targeting of ERQC for the development of broad-spectrum antivirals. We determined small-angle X-ray scattering (SAXS) and crystal structures of the main ERQC enzyme, ER α-glucosidase II (α-GluII; from mouse), alone and in complex with key ligands of its catalytic cycle and antiviral iminosugars, including two that are in clinical trials for the treatment of dengue fever. The SAXS data capture the enzyme's quaternary structure and suggest a conformational rearrangement is needed for the simultaneous binding of a monoglucosylated glycan to both subunits. The X-ray structures with key catalytic cycle intermediates highlight that an insertion between the +1 and +2 subsites contributes to the enzyme's activity and substrate specificity, and reveal that the presence of d-mannose at the +1 subsite renders the acid catalyst less efficient during the cleavage of the monoglucosylated substrate. The complexes with iminosugar antivirals suggest that inhibitors targeting a conserved ring of aromatic residues between the α-GluII +1 and +2 subsites would have increased potency and selectivity, thus providing a template for further rational drug design.
Collapse
Affiliation(s)
- Alessandro T Caputo
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Dominic S Alonzi
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Lucia Marti
- Institute of Sciences of Food Production, Consiglio Nazionale delle Ricerche Unit of Lecce, 73100 Lecce, Italy
| | - Ida-Barbara Reca
- Institute of Sciences of Food Production, Consiglio Nazionale delle Ricerche Unit of Lecce, 73100 Lecce, Italy
| | - J L Kiappes
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Weston B Struwe
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Alice Cross
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Souradeep Basu
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Edward D Lowe
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Benoit Darlot
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom; Ecole Nationale Supérieure de Chimie de Montpellier, 34296 Montpellier Cedex 5, France
| | - Angelo Santino
- Institute of Sciences of Food Production, Consiglio Nazionale delle Ricerche Unit of Lecce, 73100 Lecce, Italy
| | - Pietro Roversi
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom;
| | - Nicole Zitzmann
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom;
| |
Collapse
|
37
|
Abstract
Current treatments efficiently control chronic HBV infection but they do not lead to its elimination. Now, Ebert and colleagues have shown that cellular inhibitor of apoptosis proteins (cIAPs) prevent TNF-mediated killing of infected hepatocytes and that cIAP antagonists might lead to HBV cure by promoting death of infected cells.
Collapse
|
38
|
Abstract
Effective antivirals have been developed against specific viruses, such as HIV, Hepatitis C virus and influenza virus. This 'one bug-one drug' approach to antiviral drug development can be successful, but it may be inadequate for responding to an increasing diversity of viruses that cause significant diseases in humans. The majority of viral pathogens that cause emerging and re-emerging infectious diseases are membrane-enveloped viruses, which require the fusion of viral and cell membranes for virus entry. Therefore, antivirals that target the membrane fusion process represent new paradigms for broad-spectrum antiviral discovery. In this Review, we discuss the mechanisms responsible for the fusion between virus and cell membranes and explore how broad-spectrum antivirals target this process to prevent virus entry.
Collapse
Affiliation(s)
- Frederic Vigant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| |
Collapse
|