1
|
Alejandro B, Kim EJ, Hwang JY, Park JW, Smith M, Chung D. Genetic and phenotypic changes to Venezuelan equine encephalitis virus following treatment with β-D-N4-hydroxycytidine, an RNA mutagen. Sci Rep 2024; 14:25265. [PMID: 39448734 PMCID: PMC11502654 DOI: 10.1038/s41598-024-76788-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
The high mutation rate of RNA viruses provides viral populations with the ability to adapt to new environments but also makes them vulnerable to extinction due to the deleterious effects of mutations, which is the conceptual basis for the antiviral activity of RNA mutagens. However, there are still gaps in the quantitative understanding of the dynamics between the mutations induced by an RNA mutagen and its effects on viral fitness. To address this, we used Venezuelan Equine Encephalitis Virus (VEEV) and the potent RNA mutagen β-d-N4-hydroxycytidine (NHC) as a model to analyze virus replication competency and mutation frequency following treatment in the total and replication-competent viral populations separately. We found that NHC induced transition mutations in a concentration dependent manner in the total population, while the replication-competent population maintained itself within an increased, yet narrow, mutation spectrum. The incorporation of NHC mainly happened during the positive sense RNA synthesis of VEEV. A growth kinetic analysis of VEEV population treated with NHC pointed to a lower but more diverse distribution in mutational fitness, demonstrating that NHC-induced mutations negatively and broadly affect the fitness of the virus. Together, our study provides mechanistic insight into how RNA mutagens affect viral population landscape and the potential of RNA mutagens as an antiviral strategy for alphaviruses.
Collapse
Affiliation(s)
- Brian Alejandro
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| | - Eun Jung Kim
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| | - Jae Yeon Hwang
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Brown Cancer Center Bioinformatics Core, University of Louisville, Louisville, KY, USA
| | - Juw Won Park
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Brown Cancer Center Bioinformatics Core, University of Louisville, Louisville, KY, USA
| | - Melissa Smith
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Donghoon Chung
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
2
|
Ogbaini-Emovon E, Akpede G, Okogbenin S, Osagiede E, Tobin E, Asogun D, Okokhere P, Okonofua M, Akpede N, Akhideno P, Erameh C, Rafiu M, Azubuike C, Iraoya K, Iruolagbe C, Erohubie C, Ahmed D, Ediawe O, Okoguale J, Eifediyi R, Odia I, Agbukor J, Adomeh D, Odike MAC, Ovienria W, Elkanem A, Muoebenam EB, Ojide KC, Pallasch E, Müller J, Hinzmann J, Günther S, Pahlmann M, Thielebein A, Duraffour S, Oestereich L, Krumkamp R. Virus Load Kinetics in Lassa Fever Patients Treated With Ribavirin: A Retrospective Cohort Study From Southern Nigeria. Open Forum Infect Dis 2024; 11:ofae575. [PMID: 39450398 PMCID: PMC11500659 DOI: 10.1093/ofid/ofae575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Background The standard of care for Lassa fever is the use of ribavirin with supportive therapy. There is little information on the course of viremia and its relationship with clinical outcomes in patients treated with ribavirin. Methods We conducted a retrospective analysis of virologic and clinical parameters of 152 reverse transcription polymerase chain reaction-confirmed Lassa fever cases admitted and treated with ribavirin therapy. We describe the Lassa virus RNA kinetics in blood in relation to the clinical course of the patients. Results The overall mortality was 9%. The median duration (interquartile range [IQR]) of illness before admission was 8 (5-12) days. Median (IQR) Ct values on admission (t0 ) were lower among patients who died (21 [20-27]) than in those who survived (34 [30-37]; P < .01). The receiver operating characteristics curve of the association between outcome and Ct value at t0 had a high classification performance, with an AUC of 0.92 (95% CI, 0.86-0.98). The median time to viral clearance (IQR) was 10 (5-15) days. The viral load decreased steadily with the duration of treatment, and all survivors achieved viral clearance within 25 days of hospitalization. Conclusions Our study demonstrates that the Ct value on admission has prognostic value and Lassa fever patients treated with ribavirin typically clear the virus within 3-4 weeks of hospitalization. This kinetics has implications for the design of clinical case management and future clinical trial protocols.
Collapse
Affiliation(s)
- Ephraim Ogbaini-Emovon
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - George Akpede
- Department of Pediatrics, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Sylvanus Okogbenin
- Department of Obstetrics and Gynaecology, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Emmanuel Osagiede
- Department of Community, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Ekaete Tobin
- Department of Community, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Danny Asogun
- Department of Community, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Peter Okokhere
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Martha Okonofua
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Nosa Akpede
- Department of Community, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Peter Akhideno
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Cyril Erameh
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Mojeed Rafiu
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | | | - Kelly Iraoya
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Chris Iruolagbe
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Christian Erohubie
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Dazumi Ahmed
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Osahogie Ediawe
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Joseph Okoguale
- Department of Obstetrics and Gynaecology, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Reuben Eifediyi
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Ikponmwonsa Odia
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Jacqueline Agbukor
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Donatus Adomeh
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Maxy A C Odike
- Department of Histopathology, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Wilson Ovienria
- Department of Ophthalmology, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Anieno Elkanem
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Ekene B Muoebenam
- Institute of Viral Haemorrhagic Fever and Emergent Pathogens, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Kingsley C Ojide
- Department of Medical Microbiology, Alex Ekwemen Federal Teaching Hospital, Abakaliki, Nigeria
| | - Elisa Pallasch
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Borstel-Lübeck-Riems, Germany
| | - Jonas Müller
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Julia Hinzmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Borstel-Lübeck-Riems, Germany
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Borstel-Lübeck-Riems, Germany
| | - Meike Pahlmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Anke Thielebein
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sophie Duraffour
- Department of Medical Microbiology, Alex Ekwemen Federal Teaching Hospital, Abakaliki, Nigeria
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Borstel-Lübeck-Riems, Germany
| | - Ralf Krumkamp
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Borstel-Lübeck-Riems, Germany
| |
Collapse
|
3
|
Moliner-Cubel S, Bahamontes-Rosa N, Rodriguez-Alejandre A, Nassau PM, Argyrou A, Bhardwaja A, Buxton RC, Calvo-Vicente D, Mouzon B, McDowell W, Mendoza-Losana A, Gomez-Lorenzo MG. Plasmodium RNA triphosphatase validation as antimalarial target. Int J Parasitol Drugs Drug Resist 2024; 25:100537. [PMID: 38810336 PMCID: PMC11157219 DOI: 10.1016/j.ijpddr.2024.100537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/23/2024] [Accepted: 04/04/2024] [Indexed: 05/31/2024]
Abstract
Target-based approaches have traditionally been used in the search for new anti-infective molecules. Target selection process, a critical step in Drug Discovery, identifies targets that are essential to establish or maintain the infection, tractable to be susceptible for inhibition, selective towards their human ortholog and amenable for large scale purification and high throughput screening. The work presented herein validates the Plasmodium falciparum mRNA 5' triphosphatase (PfPRT1), the first enzymatic step to cap parasite nuclear mRNAs, as a candidate target for the development of new antimalarial compounds. mRNA capping is essential to maintain the integrity and stability of the messengers, allowing their translation. PfPRT1 has been identified as a member of the tunnel, metal dependent mRNA 5' triphosphatase family which differs structurally and mechanistically from human metal independent mRNA 5' triphosphatase. In the present study the essentiality of PfPRT1 was confirmed and molecular biology tools and methods for target purification, enzymatic assessment and target engagement were developed, with the goal of running a future high throughput screening to discover PfPRT1 inhibitors.
Collapse
Affiliation(s)
- Sonia Moliner-Cubel
- Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Noemi Bahamontes-Rosa
- Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Ane Rodriguez-Alejandre
- Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Pamela M Nassau
- Department of Biological Sciences, GlaxoSmithKline, Stevenage, SG2 7NY, UK
| | - Argyrides Argyrou
- Department of Biological Sciences, GlaxoSmithKline, Stevenage, SG2 7NY, UK
| | - Anshu Bhardwaja
- Department of Biological Sciences, GlaxoSmithKline, Stevenage, SG2 7NY, UK
| | - Rachel C Buxton
- Department of Biological Sciences, GlaxoSmithKline, Stevenage, SG2 7NY, UK
| | - David Calvo-Vicente
- Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Bernadette Mouzon
- Department of Biological Sciences, GlaxoSmithKline, Stevenage, SG2 7NY, UK
| | - William McDowell
- Department of Biological Sciences, GlaxoSmithKline, Stevenage, SG2 7NY, UK
| | - Alfonso Mendoza-Losana
- Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain.
| |
Collapse
|
4
|
Welch SR, Garrison AR, Bente DA, Burt F, D'Addiego J, Devignot S, Dowall S, Fischer K, Hawman DW, Hewson R, Mirazimi A, Oestereich L, Vatansever Z, Spengler JR, Papa A. Third International Conference on Crimean-Congo Hemorrhagic Fever in Thessaloniki, Greece, September 19-21, 2023. Antiviral Res 2024; 225:105844. [PMID: 38428749 DOI: 10.1016/j.antiviral.2024.105844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/19/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
The Third International Conference on Crimean-Congo Hemorrhagic Fever (CCHF) was held in Thessaloniki, Greece, September 19-21, 2023, bringing together a diverse group of international partners, including public health professionals, clinicians, ecologists, epidemiologists, immunologists, and virologists. The conference was attended by 118 participants representing 24 countries and the World Health Organization (WHO). Meeting sessions covered the epidemiology of CCHF in humans; Crimean-Congo hemorrhagic fever virus (CCHFV) in ticks; wild and domestic animal hosts; molecular virology; pathogenesis and animal models; immune response related to therapeutics; and CCHF prevention in humans. The concluding session focused on recent WHO recommendations regarding disease prevention, control strategies, and innovations against CCHFV outbreaks. This meeting report summarizes lectures by the invited speakers and highlights advances in the field.
Collapse
Affiliation(s)
- Stephen R Welch
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Aura R Garrison
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Dennis A Bente
- Galveston National Laboratory, Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Felicity Burt
- Division of Virology, National Health Laboratory Service and Division of Virology, University of the Free State, Bloemfontein, South Africa
| | - Jake D'Addiego
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Stephanie Devignot
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Stuart Dowall
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Kerstin Fischer
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald-Insel Riems, Germany
| | - David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Roger Hewson
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Ali Mirazimi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine and German Center for Infectious Research, Partner Sites Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Zati Vatansever
- Kafkas University, Faculty of Veterinary Medicine, Dept. of Parasitology, Kars, Turkey
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anna Papa
- Department of Microbiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
5
|
Kamzeeva PN, Aralov AV, Alferova VA, Korshun VA. Recent Advances in Molecular Mechanisms of Nucleoside Antivirals. Curr Issues Mol Biol 2023; 45:6851-6879. [PMID: 37623252 PMCID: PMC10453654 DOI: 10.3390/cimb45080433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
The search for new drugs has been greatly accelerated by the emergence of new viruses and drug-resistant strains of known pathogens. Nucleoside analogues (NAs) are a prospective class of antivirals due to known safety profiles, which are important for rapid repurposing in the fight against emerging pathogens. Recent improvements in research methods have revealed new unexpected details in the mechanisms of action of NAs that can pave the way for new approaches for the further development of effective drugs. This review accounts advanced techniques in viral polymerase targeting, new viral and host enzyme targeting approaches, and prodrug-based strategies for the development of antiviral NAs.
Collapse
Affiliation(s)
| | | | | | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (P.N.K.); (A.V.A.); (V.A.A.)
| |
Collapse
|
6
|
Ohno H, Akamine S, Mochizuki M, Hayashi K, Akichika S, Suzuki T, Saito H. Versatile strategy using vaccinia virus-capping enzyme to synthesize functional 5' cap-modified mRNAs. Nucleic Acids Res 2023; 51:e34. [PMID: 36731515 PMCID: PMC10085709 DOI: 10.1093/nar/gkad019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/23/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
The potential of synthetic mRNA as a genetic carrier has increased its application in scientific fields. Because the 5' cap regulates the stability and translational activity of mRNAs, there are concerted efforts to search for and synthesize chemically-modified 5' caps that improve the functionality of mRNA. Here, we report an easy and efficient method to synthesize functional mRNAs by modifying multiple 5' cap analogs using a vaccinia virus-capping enzyme. We show that this enzyme can introduce a variety of GTP analogs to the 5' end of RNA to generate 5' cap-modified mRNAs that exhibit different translation levels. Notably, some of these modified mRNAs improve translation efficiency and can be conjugated to chemical structures, further increasing their functionality. Our versatile method to generate 5' cap-modified mRNAs will provide useful tools for RNA therapeutics and biological research.
Collapse
Affiliation(s)
- Hirohisa Ohno
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Sae Akamine
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Megumi Mochizuki
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Karin Hayashi
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinichiro Akichika
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tsutomu Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hirohide Saito
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
7
|
D'Addiego J, Elaldi N, Wand N, Osman K, Bagci BK, Kennedy E, Pektas AN, Hart E, Slack G, Hewson R. Investigating the effect of ribavirin treatment on genetic mutations in Crimean-Congo haemorrhagic fever virus (CCHFV) through next-generation sequencing. J Med Virol 2023; 95:e28548. [PMID: 36734067 DOI: 10.1002/jmv.28548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Crimean-Congo haemorrhagic fever (CCHF) is the most widespread tick-borne viral haemorrhagic fever affecting humans, and yet a licensed drug against the virus (CCHFV) is still not available. While several studies have suggested the efficacy of ribavirin against CCHFV, current literature remains inconclusive. In this study, we have utilised next-generation sequencing to investigate the mutagenic effect of ribavirin on the CCHFV genome during clinical disease. Samples collected from CCHF patients receiving ribavirin treatment or supportive care only at Sivas Cumhuriyet University Hospital, Turkey, were analysed. By comparing the frequency of mutations in each group, we found little evidence of an overall mutagenic effect. This suggests that ribavirin, administered at the acute stages of CCHFV infection (at the World Health Organization-recommended dose) is unable to induce lethal mutagenesis that would cause an extinction event in the CCHFV population and reduce viremia.
Collapse
Affiliation(s)
- Jake D'Addiego
- UK Health Security Agency, Salisbury, UK
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Nazif Elaldi
- Department of Infectious Diseases and Clinical Microbiology, Sivas Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| | | | | | - Binnur Koksal Bagci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | | | - Ayse Nur Pektas
- Cumhuriyet University Advanced Technology Application and Research Center (CUTAM), Sivas Cumhuriyet University, Sivas, Turkey
| | | | | | - Roger Hewson
- UK Health Security Agency, Salisbury, UK
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
8
|
Shannon A, Canard B. Kill or corrupt: Mechanisms of action and drug-resistance of nucleotide analogues against SARS-CoV-2. Antiviral Res 2023; 210:105501. [PMID: 36567022 PMCID: PMC9773703 DOI: 10.1016/j.antiviral.2022.105501] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Nucleoside/tide analogues (NAs) have long been used in the fight against viral diseases, and now present a promising option for the treatment of COVID-19. Once activated to the 5'-triphosphate state, NAs act by targeting the viral RNA-dependent RNA-polymerase for incorporation into the viral RNA genome. Incorporated analogues can either 'kill' (terminate) synthesis, or 'corrupt' (genetically or chemically) the RNA. Against coronaviruses, the use of NAs has been further complicated by the presence of a virally encoded exonuclease domain (nsp14) with proofreading and repair capacities. Here, we describe the mechanism of action of four promising anti-COVID-19 NAs; remdesivir, molnupiravir, favipiravir and bemnifosbuvir. Their distinct mechanisms of action best exemplify the concept of 'killers' and 'corruptors'. We review available data regarding their ability to be incorporated and excised, and discuss the specific structural features that dictate their overall potency, toxicity, and mutagenic potential. This should guide the synthesis of novel analogues, lend insight into the potential for resistance mutations, and provide a rational basis for upcoming combinations therapies.
Collapse
Affiliation(s)
- Ashleigh Shannon
- AFMB, CNRS, Aix-Marseille University, UMR 7257, Case 925, 163 Avenue de Luminy, 13288, Marseille, Cedex 09, France
| | - Bruno Canard
- AFMB, CNRS, Aix-Marseille University, UMR 7257, Case 925, 163 Avenue de Luminy, 13288, Marseille, Cedex 09, France.
| |
Collapse
|
9
|
Burman B, Drutman SB, Fury MG, Wong RJ, Katabi N, Ho AL, Pfister DG. Pharmacodynamic and therapeutic pilot studies of single-agent ribavirin in patients with human papillomavirus-related malignancies. Oral Oncol 2022; 128:105806. [PMID: 35339025 PMCID: PMC9788648 DOI: 10.1016/j.oraloncology.2022.105806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/08/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Ribavirin inhibits eukaryotic translation initiation factor 4E (eIF4E), thereby decreasing cap-dependent translation. In this two-part study, we assessed the pharmacodynamic effects and therapeutic potential of ribavirin in human papillomavirus (HPV)-related malignancies. METHODS In the pharmacodynamic study, ribavirin (400 mg BID for 14 days) was evaluated in 8 patients with HPV-positive localized oropharyngeal carcinoma with phosphorylated-eIF4E (p-eIF4E) ≥ 30%. In the therapeutic study, ribavirin (1400 mg BID in 28-day cycles, continuously dosed) was evaluated in 12 patients with recurrent and/or metastatic HPV-related cancer. Dose interruptions or reductions were allowed according to prespecified criteria. Toxicities were assessed in accordance with National Cancer Institute Common Terminology Criteria for Adverse Events version 4; response was assessed using Response Evaluation Criteria in Solid Tumors version 1.1. Patients remained on study until disease progression or unacceptable toxicity. RESULTS Six patients were evaluable in the pharmacodynamic study: 4 had decreased p-eIF4E after 14 days of ribavirin. In the therapeutic study, 12 patients were evaluable for toxicity, and 9 were evaluable for response. Among these, median follow-up was 3.5 months, and best overall response was stable disease in 5 patients and progression of disease in 4 patients. Median progression-free survival was 1.8 months. The most common treatment-related adverse events (grade > 2) were anemia, dyspnea, and hyperbilirubinemia. All patients had anemia (grades 1-3), with 33% having at least 1 dose reduction. CONCLUSION Oral ribavirin decreases p-eIF4E levels and is well-tolerated. However, a clear signal of efficacy in patients with recurrent and/or metastatic HPV-related cancers was not observed. (NCT02308241, NCT01268579).
Collapse
Affiliation(s)
- Bharat Burman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Scott B. Drutman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Matthew G. Fury
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY,Weill Cornell Medical College, New York, NY
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY,Weill Cornell Medical College, New York, NY
| | - Nora Katabi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alan L. Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY,Weill Cornell Medical College, New York, NY
| | - David G. Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY,Weill Cornell Medical College, New York, NY
| |
Collapse
|
10
|
Salam AP, Duvignaud A, Jaspard M, Malvy D, Carroll M, Tarning J, Olliaro PL, Horby PW. Ribavirin for treating Lassa fever: A systematic review of pre-clinical studies and implications for human dosing. PLoS Negl Trop Dis 2022; 16:e0010289. [PMID: 35353804 PMCID: PMC9000057 DOI: 10.1371/journal.pntd.0010289] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/11/2022] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
Ribavirin is currently the standard of care for treating Lassa fever. However, the human clinical trial data supporting its use suffer from several serious flaws that render the results and conclusions unreliable. We performed a systematic review of available pre-clinical data and human pharmacokinetic data on ribavirin in Lassa. In in-vitro studies, the EC50 of ribavirin ranged from 0.6 μg/ml to 21.72 μg/ml and the EC90 ranged from 1.5 μg/ml to 29 μg/ml. The mean EC50 was 7 μg/ml and the mean EC90 was 15 μg/ml. Human PK data in patients with Lassa fever was sparse and did not allow for estimation of concentration profiles or pharmacokinetic parameters. Pharmacokinetic modelling based on healthy human data suggests that the concentration profiles of current ribavirin regimes only exceed the mean EC50 for less than 20% of the time and the mean EC90 for less than 10% of the time, raising the possibility that the current ribavirin regimens in clinical use are unlikely to reliably achieve serum concentrations required to inhibit Lassa virus replication. The results of this review highlight serious issues with the evidence, which, by today standards, would be unlikely to support the transition of ribavirin from pre-clinical studies to human clinical trials. Additional pre-clinical studies are needed before embarking on expensive and challenging clinical trials of ribavirin in Lassa fever.
Collapse
Affiliation(s)
- Alex P. Salam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- United Kingdom Public Health Rapid Support Team, London, United Kingdom
| | - Alexandre Duvignaud
- Department of Infectious Diseases and Tropical Medicine, Division of Tropical Medicine and Clinical International Health, CHU de Bordeaux, Bordeaux, France
- UMR1219, INSERM, French National Research Institute for Sustainable Development (IRD), and University of Bordeaux, Bordeaux, France
- Programme PAC-CI/ANRS Research Center, CHU de Treichville, Abidjan, Côte d’Ivoire
| | - Marie Jaspard
- UMR1219, INSERM, French National Research Institute for Sustainable Development (IRD), and University of Bordeaux, Bordeaux, France
- Programme PAC-CI/ANRS Research Center, CHU de Treichville, Abidjan, Côte d’Ivoire
- Alliance for International Medical Action, Dakar, Senegal
| | - Denis Malvy
- Department of Infectious Diseases and Tropical Medicine, Division of Tropical Medicine and Clinical International Health, CHU de Bordeaux, Bordeaux, France
- UMR1219, INSERM, French National Research Institute for Sustainable Development (IRD), and University of Bordeaux, Bordeaux, France
- Programme PAC-CI/ANRS Research Center, CHU de Treichville, Abidjan, Côte d’Ivoire
| | - Miles Carroll
- Wellcome Center for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Joel Tarning
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Piero L. Olliaro
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Peter W. Horby
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Abstract
The COVID-19 pandemic has highlighted the need to identify additional antiviral small molecules to complement existing therapies. Although increasing evidence suggests that metabolites produced by the human microbiome have diverse biological activities, their antiviral properties remain poorly explored. Using a cell-based SARS-CoV-2 infection assay, we screened culture broth extracts from a collection of phylogenetically diverse human-associated bacteria for the production of small molecules with antiviral activity. Bioassay-guided fractionation uncovered three bacterial metabolites capable of inhibiting SARS-CoV-2 infection. This included the nucleoside analogue N6-(Δ2-isopentenyl)adenosine, the 5-hydroxytryptamine receptor agonist tryptamine, and the pyrazine 2,5-bis(3-indolylmethyl)pyrazine. The most potent of these, N6-(Δ2-isopentenyl)adenosine, had a 50% inhibitory concentration (IC50) of 2 μM. These natural antiviral compounds exhibit structural and functional similarities to synthetic drugs that have been clinically examined for use against COVID-19. Our discovery of structurally diverse metabolites with anti-SARS-CoV-2 activity from screening a small fraction of the bacteria reported to be associated with the human microbiome suggests that continued exploration of phylogenetically diverse human-associated bacteria is likely to uncover additional small molecules that inhibit SARS-CoV-2 as well as other viral infections. IMPORTANCE The continued prevalence of COVID-19 and the emergence of new variants has once again put the spotlight on the need for the identification of SARS-CoV-2 antivirals. The human microbiome produces an array of small molecules with bioactivities (e.g., host receptor ligands), but its ability to produce antiviral small molecules is relatively underexplored. Here, using a cell-based screening platform, we describe the isolation of three microbiome-derived metabolites that are able to prevent SARS-CoV-2 infection in vitro. These molecules display structural similarities to synthetic drugs that have been explored for the treatment of COVID-19, and these results suggest that the microbiome may be a fruitful source of the discovery of small molecules with antiviral activities.
Collapse
|
12
|
Liatsos GD. Controversies’ clarification regarding ribavirin efficacy in measles and coronaviruses: Comprehensive therapeutic approach strictly tailored to COVID-19 disease stages. World J Clin Cases 2021; 9:5135-5178. [PMID: 34307564 PMCID: PMC8283580 DOI: 10.12998/wjcc.v9.i19.5135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/01/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ribavirin is a broad-spectrum nucleoside antiviral drug with multimodal mechanisms of action, which supports its longevity and quality as a clinical resource. It has been widely administered for measles and coronavirus infections. Despite the large amount of data concerning the use of ribavirin alone or in combination for measles, severe acute respiratory syndrome, Middle East respiratory syndrome, and coronavirus disease 2019 (COVID-19) outbreaks, the conclusions of these studies have been contradictory. Underlying reasons for these discrepancies include possible study design inaccuracies and failures and misinterpretations of data, and these potential confounds should be addressed.
AIM To determine the confounding factors of ribavirin treatment studies and propose a therapeutic scheme for COVID-19.
METHODS PubMed database was searched over a period of five decades utilizing the terms “ribavirin” alone or combined with other compounds in measles, severe acute respiratory syndrome, Middle East respiratory syndrome, and COVID-19 infections. The literature search was performed and described according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Articles were considered eligible when they reported on ribavirin dose regimens and/or specified outcomes concerning its efficacy and/or possible adverse-effects. In vitro and animal studies were also retrieved. A chapter on ribavirin’s pharmacology was included as well.
RESULTS In addition to the difficulties and pressures of an emerging pandemic, there is the burden of designing and conducting well-organized, double-blind, randomized controlled trials. Many studies have succumbed to specific pitfalls, one of which was identified in naturally ribavirin-resistant Vero cell lines in in vitro studies. Other pitfalls include study design inconsistent with the well-established clinical course of disease; inappropriate pharmacology of applied treatments; and the misinterpretation of study results with misconceived generalizations. A comprehensive treatment for COVID-19 is proposed, documented by thorough, long-term investigation of ribavirin regimens in coronavirus infections.
CONCLUSION A comprehensive treatment strictly tailored to distinct disease stages was proposed based upon studies on ribavirin and coronavirus infections.
Collapse
Affiliation(s)
- George D Liatsos
- Department of Internal Medicine, "Hippokration" General Hospital, Athens 11527, Attiki, Greece
| |
Collapse
|
13
|
Sorouri F, Emamgholipour Z, Keykhaee M, Najafi A, Firoozpour L, Sabzevari O, Sharifzadeh M, Foroumadi A, Khoobi M. The situation of small molecules targeting key proteins to combat SARS-CoV-2: Synthesis, metabolic pathway, mechanism of action, and potential therapeutic applications. Mini Rev Med Chem 2021; 22:273-311. [PMID: 33687881 DOI: 10.2174/1389557521666210308144302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022]
Abstract
Due to the global epidemic and high mortality of 2019 coronavirus disease (COVID-19), there is an immediate need to discover drugs that can help before a vaccine becomes available. Given that the process of producing new drugs is so long, the strategy of repurposing existing drugs is one of the promising options for the urgent treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19 disease. Although FDA has approved Remdesivir for the use in hospitalized adults and pediatric patients suffering from COVID-19, no fully effective and reliable drug has been yet identified worldwide to treat COVID-19 specifically. Thus, scientists are still trying to find antivirals specific to COVID-19. This work reviews the chemical structure, metabolic pathway, mechanism of action of existing drugs with potential therapeutic applications for COVID-19. Further, we summarized the molecular docking stimulation of the medications related to key protein targets. These already drugs could be developed for further clinical trials to supply suitable therapeutic options for patients suffering from COVID-19.
Collapse
Affiliation(s)
- Farzaneh Sorouri
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Science, Tehran. Iran
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran. Iran
| | - Maryam Keykhaee
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Science, Tehran. Iran
| | - Alireza Najafi
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran. Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran. Iran
| | - Omid Sabzevari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran. Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran. Iran
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran. Iran
| | - Mehdi Khoobi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran. Iran
| |
Collapse
|
14
|
Ruggieri A, Helm M, Chatel-Chaix L. An epigenetic 'extreme makeover': the methylation of flaviviral RNA (and beyond). RNA Biol 2021; 18:696-708. [PMID: 33356825 DOI: 10.1080/15476286.2020.1868150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Beyond their high clinical relevance worldwide, flaviviruses (comprising dengue and Zika viruses) are of particular interest to understand the spatiotemporal control of RNA metabolism. Indeed, their positive single-stranded viral RNA genome (vRNA) undergoes in the cytoplasm replication, translation and encapsidation, three steps of the flavivirus life cycle that are coordinated through a fine-tuned equilibrium. Over the last years, RNA methylation has emerged as a powerful mechanism to regulate messenger RNA metabolism at the posttranscriptional level. Not surprisingly, flaviviruses exploit RNA epigenetic strategies to control crucial steps of their replication cycle as well as to evade sensing by the innate immune system. This review summarizes the current knowledge about vRNA methylation events and their impacts on flavivirus replication and pathogenesis. We also address the important challenges that the field of epitranscriptomics faces in reliably and accurately identifying RNA methylation sites, which should be considered in future studies on viral RNA modifications.
Collapse
Affiliation(s)
- Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, Centre for Integrative Infectious Disease Research University of Heidelberg, Heidelberg, Germany
| | - Mark Helm
- Johannes Gutenberg-Universität Mainz, Institute of Pharmaceutical and Biomedical Sciences, Mainz, Germany
| | - Laurent Chatel-Chaix
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| |
Collapse
|
15
|
Huchting J. Targeting viral genome synthesis as broad-spectrum approach against RNA virus infections. Antivir Chem Chemother 2020; 28:2040206620976786. [PMID: 33297724 PMCID: PMC7734526 DOI: 10.1177/2040206620976786] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Zoonotic spillover, i.e. pathogen transmission from animal to human, has repeatedly introduced RNA viruses into the human population. In some cases, where these viruses were then efficiently transmitted between humans, they caused large disease outbreaks such as the 1918 flu pandemic or, more recently, outbreaks of Ebola and Coronavirus disease. These examples demonstrate that RNA viruses pose an immense burden on individual and public health with outbreaks threatening the economy and social cohesion within and across borders. And while emerging RNA viruses are introduced more frequently as human activities increasingly disrupt wild-life eco-systems, therapeutic or preventative medicines satisfying the “one drug-multiple bugs”-aim are unavailable. As one central aspect of preparedness efforts, this review digs into the development of broadly acting antivirals via targeting viral genome synthesis with host- or virus-directed drugs centering around nucleotides, the genomes’ universal building blocks. Following the first strategy, selected examples of host de novo nucleotide synthesis inhibitors are presented that ultimately interfere with viral nucleic acid synthesis, with ribavirin being the most prominent and widely used example. For directly targeting the viral polymerase, nucleoside and nucleotide analogues (NNAs) have long been at the core of antiviral drug development and this review illustrates different molecular strategies by which NNAs inhibit viral infection. Highlighting well-known as well as recent, clinically promising compounds, structural features and mechanistic details that may confer broad-spectrum activity are discussed. The final part addresses limitations of NNAs for clinical development such as low efficacy or mitochondrial toxicity and illustrates strategies to overcome these.
Collapse
Affiliation(s)
- Johanna Huchting
- Chemistry Department, Institute for Organic Chemistry, Faculty of Mathematics, Computer Science and Natural Sciences, University of Hamburg, Hamburg, Germany
| |
Collapse
|
16
|
Tate PM, Mastrodomenico V, Mounce BC. Ribavirin Induces Polyamine Depletion via Nucleotide Depletion to Limit Virus Replication. Cell Rep 2020; 28:2620-2633.e4. [PMID: 31484073 DOI: 10.1016/j.celrep.2019.07.099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/24/2019] [Accepted: 07/26/2019] [Indexed: 11/16/2022] Open
Abstract
Common antivirals include nucleoside or nucleotide analogs with base prodrugs. The antiviral ribavirin, a US Food and Drug Administration (FDA)-approved nucleoside antimetabolite, halts guanine production, mutagenizes viral genomes, and activates interferon signaling. Here, we find that ribavirin induces spermidine-spermine N1-acetyltransferase (SAT1), a polyamine catabolic enzyme. Polyamines are small, positively charged molecules involved in cellular functions such as transcription and translation. Previous work showed that SAT1 activation and polyamine depletion interfere with RNA virus replication. We show ribavirin depletes polyamines via SAT1, in conjunction with its known mechanisms. SAT1 transcripts, protein, and activity are induced in a dose-dependent manner, which depletes polyamine levels and reduces viral titers. Inhibition of SAT1 activity, pharmacologically or genetically, reduces ribavirin's effectiveness against three virus infection models. Additionally, ribavirin-mediated polyamine depletion results from nucleotide pool depletion. These data demonstrate another mechanism of ribavirin that inform its clinical effectiveness, which may provide insight for improved therapies.
Collapse
Affiliation(s)
- Patrick M Tate
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Vincent Mastrodomenico
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Bryan C Mounce
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153, USA.
| |
Collapse
|
17
|
Mutations on VEEV nsP1 relate RNA capping efficiency to ribavirin susceptibility. Antiviral Res 2020; 182:104883. [PMID: 32750467 DOI: 10.1016/j.antiviral.2020.104883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 11/22/2022]
Abstract
Alphaviruses are arthropod-borne viruses of public health concern. To date no efficient vaccine nor antivirals are available for safe human use. During viral replication the nonstructural protein 1 (nsP1) catalyzes capping of genomic and subgenomic RNAs. The capping reaction is unique to the Alphavirus genus. The whole three-step process follows a particular order: (i) transfer of a methyl group from S-adenosyl methionine (SAM) onto a GTP forming m7GTP; (ii) guanylylation of the enzyme to form a m7GMP-nsP1adduct; (iii) transfer of m7GMP onto 5'-diphosphate RNA to yield capped RNA. Specificities of these reactions designate nsP1 as a promising target for antiviral drug development. In the current study we performed a mutational analysis on two nsP1 positions associated with Sindbis virus (SINV) ribavirin resistance in the Venezuelan equine encephalitis virus (VEEV) context through reverse genetics correlated to enzyme assays using purified recombinant VEEV nsP1 proteins. The results demonstrate that the targeted positions are strongly associated to the regulation of the capping reaction by increasing the affinity between GTP and nsP1. Data also show that in VEEV the S21A substitution, naturally occurring in Chikungunya virus (CHIKV), is a hallmark of ribavirin susceptibility. These findings uncover the specific mechanistic contributions of these residues to nsp1-mediated methyl-transfer and guanylylation reactions.
Collapse
|
18
|
Prutkov AN, Chudinov MV, Matveev AV, Grebenkina LE, Akimov MG, Berezovskaya YV. 5-alkylvinyl-1,2,4-triazole nucleosides: Synthesis and biological evaluation. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2020; 39:943-963. [PMID: 32126895 DOI: 10.1080/15257770.2020.1723624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Some 5-substituted ribavirin analogues have a high antiviral and anticancer activity, but their mechanisms of action are obviously not the same as their parent compound. The SAR studies performed on 3 (5)-substituted 1,2,4-triazole nucleosides have shown a high dependency between the structure of the 3 (5)-substituent and the level of antiviral/anticancer activity. The most active substances of the row contain coplanar with the 1,2,4-triazole ring aromatic substituent which is connected by a rigid ethynyl bond. However, the compounds with the trans-vinyl linker also had antiviral activity. We decided to study the antitumor activity of ribavirin analogues with alkyl/aryl vinyl substituents in the 5th position of the 1,2,4-triazole ring. Protected nucleoside analogues with various 5-alkylvinyl substituents were obtained by Horner-Wadsworth-Emmons reaction from the common precursor and converted to the nucleosides. Arylvinyl nucleosides were synthesised according the reported procedures. All compounds did not show significant antiproliferative activity on several tumour cell lines. Coplanar aromatic motif in the 5-substituent for the anticancer activity manifestation was confirmed.
Collapse
Affiliation(s)
- Alexander N Prutkov
- Biotechnology & Industrial Pharmacy Department, Lomonosov Institute of Fine Chemical Tehnologies, MIREA - Russian Technological University, Moscow, Russia
| | - Mikhail V Chudinov
- Biotechnology & Industrial Pharmacy Department, Lomonosov Institute of Fine Chemical Tehnologies, MIREA - Russian Technological University, Moscow, Russia
| | - Andrey V Matveev
- Biotechnology & Industrial Pharmacy Department, Lomonosov Institute of Fine Chemical Tehnologies, MIREA - Russian Technological University, Moscow, Russia
| | - Lyubov E Grebenkina
- Biotechnology & Industrial Pharmacy Department, Lomonosov Institute of Fine Chemical Tehnologies, MIREA - Russian Technological University, Moscow, Russia
| | - Mikhail G Akimov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yulia V Berezovskaya
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
19
|
Nyström K, Waldenström J, Tang KW, Lagging M. Ribavirin: pharmacology, multiple modes of action and possible future perspectives. Future Virol 2019. [DOI: 10.2217/fvl-2018-0166] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ribavirin is a unique guanosine analog with broad-spectrum activity against many RNA and DNA viruses. In addition to its mutational properties, ribavirin exerts extensive perturbation of cellular and viral gene expression. Furthermore, recent advances indicate that the impact of ribavirin on divergent cellular and viral pathways may be concentration dependent. This review aims at providing an overview of the pharmacology and multiple modes of action of ribavirin as well as pointing to possible novel future uses.
Collapse
Affiliation(s)
- Kristina Nyström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Västra Götaland Region, Sweden
| | - Jesper Waldenström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Västra Götaland Region, Sweden
| | - Ka-Wei Tang
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Västra Götaland Region, Sweden
| | - Martin Lagging
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Västra Götaland Region, Sweden
| |
Collapse
|
20
|
Muttach F, Muthmann N, Rentmeister A. Synthetic mRNA capping. Beilstein J Org Chem 2017; 13:2819-2832. [PMID: 30018667 PMCID: PMC5753152 DOI: 10.3762/bjoc.13.274] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/04/2017] [Indexed: 12/25/2022] Open
Abstract
Eukaryotic mRNA with its 5'-cap is of central importance for the cell. Many studies involving mRNA require reliable preparation and modification of 5'-capped RNAs. Depending on the length of the desired capped RNA, chemical or enzymatic preparation - or a combination of both - can be advantageous. We review state-of-the art methods and give directions for choosing the appropriate approach. We also discuss the preparation and properties of mRNAs with non-natural caps providing novel features such as improved stability or enhanced translational efficiency.
Collapse
Affiliation(s)
- Fabian Muttach
- University of Münster, Department of Chemistry, Institute of Biochemistry, Wilhelm-Klemm-Str. 2, 48149 Münster, Germany
| | - Nils Muthmann
- University of Münster, Department of Chemistry, Institute of Biochemistry, Wilhelm-Klemm-Str. 2, 48149 Münster, Germany
| | - Andrea Rentmeister
- University of Münster, Department of Chemistry, Institute of Biochemistry, Wilhelm-Klemm-Str. 2, 48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany
| |
Collapse
|
21
|
Xu S, Yamamoto N. mRNA-Seq reveals accumulation followed by reduction of small nuclear and nucleolar RNAs in yeast exposed to antiviral ribavirin. FEMS Yeast Res 2017; 17:4085638. [PMID: 28934414 DOI: 10.1093/femsyr/fox067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
Ribavirin is an antiviral drug that is used to treat a wide range of human viral infections. However, the side effects are reported, and the mechanisms on eukaryotic cells are still largely unknown. Here we report our observation of accumulation followed by reduction of small nuclear (sn)RNAs and small nucleolar (sno)RNAs in Saccharomyces cerevisiae exposed to ribavirin. The three strains reported to contain dsRNA virus-like particle(s) were exposed to 100 μM of ribavirin, and snRNAs and snoRNAs from a total of 31 snR genes were differentially detected between the samples exposed to ribavirin and the respective negative controls by mRNA-Seq. Our results suggest that polyadenylated snRNAs and snoRNAs accumulated at 1 h but reduced to the subbasal levels at 4 h of ribavirin exposure. The tendency was reproducible across the three tested strains. Our study showed ribavirin affected snRNAs and snoRNAs in yeast. There may be a need to scrutinize the relationships between the side effects and such non-coding RNAs in humans who are treated with ribavirin.
Collapse
Affiliation(s)
- Siyu Xu
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, South Korea
| | - Naomichi Yamamoto
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
22
|
Carrillo-Bustamante P, Nguyen THT, Oestereich L, Günther S, Guedj J, Graw F. Determining Ribavirin's mechanism of action against Lassa virus infection. Sci Rep 2017; 7:11693. [PMID: 28916737 PMCID: PMC5601963 DOI: 10.1038/s41598-017-10198-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Ribavirin is a broad spectrum antiviral which inhibits Lassa virus (LASV) replication in vitro but exhibits a minor effect on viremia in vivo. However, ribavirin significantly improves the disease outcome when administered in combination with sub-optimal doses of favipiravir, a strong antiviral drug. The mechanisms explaining these conflicting findings have not been determined, so far. Here, we used an interdisciplinary approach combining mathematical models and experimental data in LASV-infected mice that were treated with ribavirin alone or in combination with the drug favipiravir to explore different putative mechanisms of action for ribavirin. We test four different hypotheses that have been previously suggested for ribavirin’s mode of action: (i) acting as a mutagen, thereby limiting the infectivity of new virions; (ii) reducing viremia by impairing viral production; (iii) modulating cell damage, i.e., by reducing inflammation, and (iv) enhancing antiviral immunity. Our analysis indicates that enhancement of antiviral immunity, as well as effects on viral production or transmission are unlikely to be ribavirin’s main mechanism mediating its antiviral effectiveness against LASV infection. Instead, the modeled viral kinetics suggest that the main mode of action of ribavirin is to protect infected cells from dying, possibly reducing the inflammatory response.
Collapse
Affiliation(s)
- Paola Carrillo-Bustamante
- Center for Modeling and Simulation in the Biosciences (BIOMS), BioQuant-Center, Heidelberg University, Heidelberg, Germany.
| | - Thi Huyen Tram Nguyen
- INSERM, IAME, UMR, 1137, Paris, France.,Université Paris Diderot, IAME, UMR, 1137, Sorbonne Paris Cité, France
| | - Lisa Oestereich
- Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Germany
| | - Stephan Günther
- Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Germany
| | - Jeremie Guedj
- INSERM, IAME, UMR, 1137, Paris, France.,Université Paris Diderot, IAME, UMR, 1137, Sorbonne Paris Cité, France
| | - Frederik Graw
- Center for Modeling and Simulation in the Biosciences (BIOMS), BioQuant-Center, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
23
|
Smee DF, Prichard MN. Comparison of three dimensional synergistic analyses of percentage versus logarithmic data in antiviral studies. Antiviral Res 2017; 145:1-5. [PMID: 28676302 DOI: 10.1016/j.antiviral.2017.06.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/28/2017] [Accepted: 06/30/2017] [Indexed: 01/26/2023]
Abstract
Cell culture antiviral experiments were conducted in order to understand the relationship between percentage data generated by plaque reduction (PR) and logarithmic data derived by virus yield reduction (VYR) assays, using three-dimensional MacSynergy II software. The relationship between percentage and logarithmic data has not been investigated previously. Interpretation of drug-drug interactions is based on a Volume of Synergy (VS) calculation, which can be positive (synergy), negative (antagonistic), or neutral (no or minimal interaction). Interactions of two known inhibitors of vaccinia virus replication, cidofovir and 6-azauridine, used in combination by PR assay yielded a VS value of 265, indicative of strong synergy. By VYR, the VS value was only 37, or weak synergy using the same criterion, even though profound log10 reductions in virus titer occurred at multiple drug combinations. These results confirm that the differences in VS values is dependent of the measurement scale, and not that the degree of synergy differed between the assays. We propose that for logarithmic data, the calculated VS values will be lower for significant synergy and antagonism and that volumes of >10 μM2log10 PFU/ml (or other units such as μM2log10 genomic equivalents/ml or μM2log10 copies/ml) and <-10 μM2log10 PFU/ml are likely to be indicative of strong synergy and strong antagonism, respectively. Data presented here show that the interaction of cidofovir and 6-azauridine was strongly synergistic in vitro.
Collapse
Affiliation(s)
- Donald F Smee
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322-5600, USA.
| | - Mark N Prichard
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, 35233-1711, USA
| |
Collapse
|
24
|
Ogino M, Ogino T. 5'-Phospho-RNA Acceptor Specificity of GDP Polyribonucleotidyltransferase of Vesicular Stomatitis Virus in mRNA Capping. J Virol 2017; 91:e02322-16. [PMID: 28053102 PMCID: PMC5331801 DOI: 10.1128/jvi.02322-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 12/23/2016] [Indexed: 11/20/2022] Open
Abstract
The GDP polyribonucleotidyltransferase (PRNTase) domain of the multifunctional L protein of rhabdoviruses, such as vesicular stomatitis virus (VSV) and rabies virus, catalyzes the transfer of 5'-phospho-RNA (pRNA) from 5'-triphospho-RNA (pppRNA) to GDP via a covalent enzyme-pRNA intermediate to generate a 5'-cap structure (GpppA). Here, using an improved oligo-RNA capping assay with the VSV L protein, we showed that the Michaelis constants for GDP and pppAACAG (VSV mRNA-start sequence) are 0.03 and 0.4 μM, respectively. A competition assay between GDP and GDP analogues in the GpppA formation and pRNA transfer assay using GDP analogues as pRNA acceptors indicated that the PRNTase domain recognizes the C-2-amino group, but not the C-6-oxo group, N-1-hydrogen, or N-7-nitrogen, of GDP for the cap formation. 2,6-Diaminopurine-riboside (DAP), 7-deazaguanosine (7-deaza-G), and 7-methylguanosine (m7G) diphosphates efficiently accepted pRNA, resulting in the formation of DAPpppA, 7-deaza-GpppA, and m7GpppA (cap 0), respectively. Furthermore, either the 2'- or 3'-hydroxyl group of GDP was found to be required for efficient pRNA transfer. A 5'-diphosphate form of antiviral ribavirin weakly inhibited the GpppA formation but did not act as a pRNA acceptor. These results indicate that the PRNTase domain has a unique guanosine-binding mode different from that of eukaryotic mRNA capping enzyme, guanylyltransferase. IMPORTANCE mRNAs of nonsegmented negative-strand (NNS) RNA viruses, such as VSV, possess a fully methylated cap structure, which is required for mRNA stability, efficient translation, and evasion of antiviral innate immunity in host cells. GDP polyribonucleotidyltransferase (PRNTase) is an unconventional mRNA capping enzyme of NNS RNA viruses that is distinct from the eukaryotic mRNA capping enzyme, guanylyltransferase. In this study, we studied the pRNA acceptor specificity of VSV PRNTase using various GDP analogues and identified chemical groups of GDP as essential for the substrate activity. The findings presented here are useful not only for understanding the mechanism of the substrate recognition with PRNTase but also for designing antiviral agents targeting this enzyme.
Collapse
Affiliation(s)
- Minako Ogino
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Tomoaki Ogino
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
25
|
Todt D, Walter S, Brown RJP, Steinmann E. Mutagenic Effects of Ribavirin on Hepatitis E Virus-Viral Extinction versus Selection of Fitness-Enhancing Mutations. Viruses 2016; 8:E283. [PMID: 27754363 PMCID: PMC5086615 DOI: 10.3390/v8100283] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/30/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022] Open
Abstract
Hepatitis E virus (HEV), an important agent of viral hepatitis worldwide, can cause severe courses of infection in pregnant women and immunosuppressed patients. To date, HEV infections can only be treated with ribavirin (RBV). Major drawbacks of this therapy are that RBV is not approved for administration to pregnant women and that the virus can acquire mutations, which render the intra-host population less sensitive or even resistant to RBV. One of the proposed modes of action of RBV is a direct mutagenic effect on viral genomes, inducing mismatches and subsequent nucleotide substitutions. These transition events can drive the already error-prone viral replication beyond an error threshold, causing viral population extinction. In contrast, the expanded heterogeneous viral population can facilitate selection of mutant viruses with enhanced replication fitness. Emergence of these mutant viruses can lead to therapeutic failure. Consequently, the onset of RBV treatment in chronically HEV-infected individuals can result in two divergent outcomes: viral extinction versus selection of fitness-enhanced viruses. Following an overview of RNA viruses treated with RBV in clinics and a summary of the different antiviral modes of action of this drug, we focus on the mutagenic effect of RBV on HEV intrahost populations, and how HEV is able to overcome lethal mutagenesis.
Collapse
Affiliation(s)
- Daniel Todt
- Institute of Experimental Virology, Twincore-Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany.
| | - Stephanie Walter
- Institute of Experimental Virology, Twincore-Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany.
| | - Richard J P Brown
- Institute of Experimental Virology, Twincore-Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany.
| | - Eike Steinmann
- Institute of Experimental Virology, Twincore-Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany.
| |
Collapse
|
26
|
A Sensitive and Robust High-Throughput Screening Assay for Inhibitors of the Chikungunya Virus nsP1 Capping Enzyme. PLoS One 2016; 11:e0158923. [PMID: 27427769 PMCID: PMC4948833 DOI: 10.1371/journal.pone.0158923] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/23/2016] [Indexed: 12/26/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne Alphavirus that causes severe and debilitating disease symptoms. Alarmingly, transmission rates of CHIKV have increased dramatically over the last decade resulting in 1.7 million suspected cases in the Western hemisphere alone. There are currently no antivirals for treatment of CHIKV infection and novel anti-alphaviral compounds are badly needed. nsP1 is the alphavirus protein responsible for the methyltransferase and guanylyltransferase activities necessary for formation of the 5’ type 0 cap structure added to newly formed viral RNA. Formation of this cap depends on nsP1 binding GTP and transferring a methylated GMP to nascent viral RNA. We have developed a fluorescence polarization-based assay that monitors displacement of a fluorescently-labeled GTP analog in real time. Determining the relative affinities of 15 GTP analogs for nsP1 GTP revealed important structural aspects of GTP that will inform identification of inhibitors able to outcompete GTP for the nsP1 binding site. Validation of the assay for HTS was completed and a secondary orthogonal assay that measures guanylation activity was developed in order to evaluate hits from future drug screens. This platform provides an avenue for identification of potent nsP1 inhibitors, which would potentially provide compounds capable of treating disease caused by CHIKV infection.
Collapse
|
27
|
Structural insights into the reaction mechanism of S-adenosyl-L-homocysteine hydrolase. Sci Rep 2015; 5:16641. [PMID: 26573329 PMCID: PMC4647836 DOI: 10.1038/srep16641] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022] Open
Abstract
S-adenosyl-L-homocysteine hydrolase (SAH hydrolase or SAHH) is a highly conserved enzyme that catalyses the reversible hydrolysis of SAH to L-homocysteine (HCY) and adenosine (ADO). High-resolution crystal structures have been reported for bacterial and plant SAHHs, but not mammalian SAHHs. Here, we report the first high-resolution crystal structure of mammalian SAHH (mouse SAHH) in complex with a reaction product (ADO) and with two reaction intermediate analogues—3’-keto-aristeromycin (3KA) and noraristeromycin (NRN)—at resolutions of 1.55, 1.55, and 1.65 Å. Each of the three structures constitutes a structural snapshot of one of the last three steps of the five-step process of SAH hydrolysis by SAHH. In the NRN complex, a water molecule, which is an essential substrate for ADO formation, is structurally identified for the first time as the candidate donor in a Michael addition by SAHH to the 3’-keto-4’,5’-didehydroadenosine reaction intermediate. The presence of the water molecule is consistent with the reaction mechanism proposed by Palmer & Abeles in 1979. These results provide insights into the reaction mechanism of the SAHH enzyme.
Collapse
|
28
|
Rivas-Aravena A, Guajardo S, Valenzuela B, Cartagena J, Imarai M, Spencer E, Sandino A. Ribavirin stimulates the immune response of Atlantic salmon. Vet Immunol Immunopathol 2015; 164:93-100. [DOI: 10.1016/j.vetimm.2015.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/29/2014] [Accepted: 01/05/2015] [Indexed: 01/15/2023]
|
29
|
Resveratrol inhibits enterovirus 71 replication and pro-inflammatory cytokine secretion in rhabdosarcoma cells through blocking IKKs/NF-κB signaling pathway. PLoS One 2015; 10:e0116879. [PMID: 25692777 PMCID: PMC4333343 DOI: 10.1371/journal.pone.0116879] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/16/2014] [Indexed: 11/19/2022] Open
Abstract
Polydatin and resveratrol, as major active components in Polygonum cuspidatum, have anti-inflammatory, antioxidant and antitumor functions. However, the effect and mechanism of polydatin and resveratrol on enterovirus 71 (EV71) have not been reported. In this study, resveratrol revealed strong antiviral activity on EV71, while polydatin had weak effect. Neither polydatin nor resveratrol exhibited influence on viral attachment. Resveratrol could effectively inhibit the synthesis of EV71/VP1 and the phosphorylation of IKKα, IKKβ, IKKγ, IKBα, NF-κB p50 and NF-κB p65, respectively. Meanwhile, the remarkably increased secretion of IL-6 and TNF-α in EV71-infected rhabdosarcoma (RD) cells could be blocked by resveratrol. These results demonstrated that resveratrol inhibited EV71 replication and cytokine secretion in EV71-infected RD cells through blocking IKKs/NF-κB signaling pathway. Thus, resveratrol may have potent antiviral effect on EV71 infection.
Collapse
|
30
|
Abstract
UNLABELLED Lethal mutagenesis is a broad-spectrum antiviral strategy that exploits the high mutation rate and low mutational tolerance of many RNA viruses. This approach uses mutagenic drugs to increase viral mutation rates and burden viral populations with mutations that reduce the number of infectious progeny. We investigated the effectiveness of lethal mutagenesis as a strategy against influenza virus using three nucleoside analogs, ribavirin, 5-azacytidine, and 5-fluorouracil. All three drugs were active against a panel of seasonal H3N2 and laboratory-adapted H1N1 strains. We found that each drug increased the frequency of mutations in influenza virus populations and decreased the virus' specific infectivity, indicating a mutagenic mode of action. We were able to drive viral populations to extinction by passaging influenza virus in the presence of each drug, indicating that complete lethal mutagenesis of influenza virus populations can be achieved when a sufficient mutational burden is applied. Population-wide resistance to these mutagenic agents did not arise after serial passage of influenza virus populations in sublethal concentrations of drug. Sequencing of these drug-passaged viral populations revealed genome-wide accumulation of mutations at low frequency. The replicative capacity of drug-passaged populations was reduced at higher multiplicities of infection, suggesting the presence of defective interfering particles and a possible barrier to the evolution of resistance. Together, our data suggest that lethal mutagenesis may be a particularly effective therapeutic approach with a high genetic barrier to resistance for influenza virus. IMPORTANCE Influenza virus is an RNA virus that causes significant morbidity and mortality during annual epidemics. Novel therapies for RNA viruses are needed due to the ease with which these viruses evolve resistance to existing therapeutics. Lethal mutagenesis is a broad-spectrum strategy that exploits the high mutation rate and the low mutational tolerance of most RNA viruses. It is thought to possess a higher barrier to resistance than conventional antiviral strategies. We investigated the effectiveness of lethal mutagenesis against influenza virus using three different drugs. We showed that influenza virus was sensitive to lethal mutagenesis by demonstrating that all three drugs induced mutations and led to an increase in the generation of defective viral particles. We also found that it may be difficult for resistance to these drugs to arise at a population-wide level. Our data suggest that lethal mutagenesis may be an attractive anti-influenza strategy that warrants further investigation.
Collapse
|
31
|
|
32
|
Krajczyk A, Kulinska K, Kulinski T, Hurst BL, Day CW, Smee DF, Ostrowski T, Januszczyk P, Zeidler J. Antivirally active ribavirin analogues--4,5-disubstituted 1,2,3-triazole nucleosides: biological evaluation against certain respiratory viruses and computational modelling. Antivir Chem Chemother 2014; 23:161-71. [PMID: 23538746 DOI: 10.3851/imp2564] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Ribavirin is a broad-spectrum antiviral agent that derives some of its activity from inhibition of cellular inosine monophosphate dehydrogenase (IMPDH), resulting in lower guanosine triphosphate (GTP) levels. Here we report the biological activities of three ribavirin analogues. METHODS Antiviral activities of test compounds were performed by in vitro cytopathic effect inhibition assays against influenza A (H1N1, H3N2 and H5N1), influenza B, measles, parainfluenza type 3 (PIV-3) and respiratory syncytial viruses. Compounds were modelled into the ribavirin 5'-monophosphate binding site of the crystallographic structure of the human type II IMPDH (hIMPDH2) ternary complex. Effects of compounds on intracellular GTP levels were performed by strong anion exchange HPLC analysis. RESULTS Of the three compounds evaluated, the 5-ethynyl nucleoside (ETCAR) exhibited virus-inhibitory activities (at 1.2-20 μM, depending upon the virus) against most of the viruses, except for weak activity against PIV-3 (62 μM). Antiviral activity of ETCAR was similar to ribavirin; however, cytotoxicity of ETCAR was greater than ribavirin. Replacing the 5-ethynyl group with a 5-propynyl or bromo substituent (BrCAR) considerably reduced antiviral activity. Computational studies of ternary complexes of hIMPDH2 enzyme with 5'-monophosphates of the compounds helped rationalize the observed differences in biological activity. All compounds suppressed GTP levels in cells; additionally, BrCAR suppressed adenosine triphosphate and elevated uridine triphosphate levels. CONCLUSIONS Three compounds related to ribavirin inhibited IMPDH and had weak to moderate antiviral activity. Cytotoxicity adversely affected the antiviral selectivity of ETCAR. As with ribavirin, reduction in intracellular GTP may play a role in virus inhibition.
Collapse
Affiliation(s)
- Anna Krajczyk
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li X, Liu Y, Hou X, Peng H, Zhang L, Jiang Q, Shi M, Ji Y, Wang Y, Shi W. Chlorogenic acid inhibits the replication and viability of enterovirus 71 in vitro. PLoS One 2013; 8:e76007. [PMID: 24098754 PMCID: PMC3786884 DOI: 10.1371/journal.pone.0076007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/16/2013] [Indexed: 12/17/2022] Open
Abstract
Enterovirus 71 (EV71) is an etiology for a number of diseases in humans. Traditional Chinese herbs have been reported to be effective for treating EV71 infection. However, there is no report about the antiviral effects of CHA against EV71. In this study, plaque reduction assay demonstrated that the inhibitory concentration 50% (IC50) of CHA on EV71 replication is 6.3 µg/ml. When both CHA (20 µg/ml) and EV71 were added, or added post-infection at different time points, CHA was able to effectively inhibit EV71 replication between 0 and 10 h. In addition, CHA inhibited EV71 2A transcription and translation in EV71-infected RD cells, but did not affect VP1, 3C, and 3D expression. Furthermore, CHA inhibited secretions of IL-6, TNF-α, IFN-γ and MCP-1 in EV71-infected RD cells. Altogether, these results revealed that CHA may have antiviral properties for treating EV71 infection.
Collapse
Affiliation(s)
- Xiang Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Yuanyuan Liu
- Department of Endocrinology, The the Huai-an First Affliated Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Xueling Hou
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Hongjun Peng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Li Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Qingbo Jiang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Mei Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Yun Ji
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Yuyue Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Weifeng Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- * E-mail:
| |
Collapse
|
34
|
Issur M, Bougie I, Despins S, Bisaillon M. Enzymatic synthesis of RNAs capped with nucleotide analogues reveals the molecular basis for substrate selectivity of RNA capping enzyme: impacts on RNA metabolism. PLoS One 2013; 8:e75310. [PMID: 24086504 PMCID: PMC3783409 DOI: 10.1371/journal.pone.0075310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/13/2013] [Indexed: 12/16/2022] Open
Abstract
RNA cap binding proteins have evolved to specifically bind to the N7-methyl guanosine cap structure found at the 5’ ends of eukaryotic mRNAs. The specificity of RNA capping enzymes towards GTP for the synthesis of this structure is therefore crucial for mRNA metabolism. The fact that ribavirin triphosphate was described as a substrate of a viral RNA capping enzyme, raised the possibility that RNAs capped with nucleotide analogues could be generated in cellulo. Owing to the fact that this prospect potentially has wide pharmacological implications, we decided to investigate whether the active site of the model Parameciumbursaria Chlorella virus-1 RNA capping enzyme was flexible enough to accommodate various purine analogues. Using this approach, we identified several key structural determinants at each step of the RNA capping reaction and generated RNAs harboring various different cap analogues. Moreover, we monitored the binding affinity of these novel capped RNAs to the eIF4E protein and evaluated their translational properties in cellulo. Overall, this study establishes a molecular rationale for the specific selection of GTP over other NTPs by RNA capping enzyme It also demonstrates that RNAs can be enzymatically capped with certain purine nucleotide analogs, and it also describes the impacts of modified RNA caps on specific steps involved in mRNA metabolism. For instance, our results indicate that the N7-methyl group of the classical N7-methyl guanosine cap is not always indispensable for binding to eIF4E and subsequently for translation when compensatory modifications are present on the capped residue. Overall, these findings have important implications for our understanding of the molecular determinants involved in both RNA capping and RNA metabolism.
Collapse
Affiliation(s)
- Moheshwarnath Issur
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Isabelle Bougie
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Simon Despins
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Martin Bisaillon
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
35
|
Picard-Jean F, Bougie I, Shuto S, Bisaillon M. The immunosuppressive agent mizoribine monophosphate is an inhibitor of the human RNA capping enzyme. PLoS One 2013; 8:e54621. [PMID: 23349942 PMCID: PMC3547949 DOI: 10.1371/journal.pone.0054621] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 12/13/2012] [Indexed: 11/18/2022] Open
Abstract
Mizoribine monophosphate (MZP) is a specific inhibitor of the cellular inosine-5′-monophosphate dehydrogenase (IMPDH), the enzyme catalyzing the rate-limiting step of de novo guanine nucleotide biosynthesis. MZP is a highly potent antagonistic inhibitor of IMPDH that blocks the proliferation of T and B lymphocytes that use the de novo pathway of guanine nucleotide synthesis almost exclusively. In the present study, we investigated the ability of MZP to directly inhibit the human RNA capping enzyme (HCE), a protein harboring both RNA 5′-triphosphatase and RNA guanylyltransferase activities. HCE is involved in the synthesis of the cap structure found at the 5′ end of eukaryotic mRNAs, which is critical for the splicing of the cap-proximal intron, the transport of mRNAs from the nucleus to the cytoplasm, and for both the stability and translation of mRNAs. Our biochemical studies provide the first insight that MZP can inhibit the formation of the RNA cap structure catalyzed by HCE. In the presence of MZP, the RNA 5′-triphosphatase activity appears to be relatively unaffected while the RNA guanylyltransferase activity is inhibited, indicating that the RNA guanylyltransferase activity is the main target of MZP inhibition. Kinetic studies reveal that MZP is a non-competitive inhibitor that likely targets an allosteric site on HCE. Mizoribine also impairs mRNA capping in living cells, which could account for the global mechanism of action of this therapeutic agent. Together, our study clearly demonstrates that mizoribine monophosphate inhibits the human RNA guanylyltransferase in vitro and impair mRNA capping in cellulo.
Collapse
Affiliation(s)
- Frédéric Picard-Jean
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Isabelle Bougie
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Satoshi Shuto
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Martin Bisaillon
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- * E-mail:
| |
Collapse
|
36
|
Goyal R, Kumar A, Panda SK, Paul SB, Acharya SK. Ribavirin therapy for hepatitis E virus-induced acute on chronic liver failure: a preliminary report. Antivir Ther 2012; 17:1091-6. [PMID: 22910532 DOI: 10.3851/imp2317] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2011] [Indexed: 12/14/2022]
Abstract
Hepatitis E virus (HEV) is a common cause of acute on chronic liver failure (ACLF) in HEV hyperendemic regions with high mortality. Treatment for HEV-induced ACLF is currently not available. Recently, efficacy of ribavirin in genotype 3 chronic hepatitis E patients has been reported; however, whether ribavirin is effective in genotype 1 HEV infection is not yet known. The present study includes four patients with HEV-induced ACLF treated with ribavirin in a genotype 1 HEV hyperendemic region. Diagnosis of ACLF was made by conventional criteria and HEV as the cause of ACLF was confirmed by detection of HEV RNA by reverse transcriptase PCR. Ribavirin dose ranged from 200 to 600 mg/day and was used for a median duration of 12 (range 3-24) weeks. All patients had undetectable HEV in 3-8 weeks, survived and none had serious adverse effects. This preliminary observation from a single centre indicates that ribavirin may be an effective therapeutic agent for HEV-induced ACLF and a randomized control trial is needed to establish its efficacy.
Collapse
Affiliation(s)
- Rohit Goyal
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | |
Collapse
|
37
|
Ferron F, Decroly E, Selisko B, Canard B. The viral RNA capping machinery as a target for antiviral drugs. Antiviral Res 2012; 96:21-31. [PMID: 22841701 PMCID: PMC7114304 DOI: 10.1016/j.antiviral.2012.07.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 07/09/2012] [Accepted: 07/13/2012] [Indexed: 12/18/2022]
Abstract
Most viruses modify their genomic and mRNA 5′-ends with the addition of an RNA cap, allowing efficient mRNA translation, limiting degradation by cellular 5′–3′ exonucleases, and avoiding its recognition as foreign RNA by the host cell. Viral RNA caps can be synthesized or acquired through the use of a capping machinery which exhibits a significant diversity in organization, structure and mechanism relative to that of their cellular host. Therefore, viral RNA capping has emerged as an interesting field for antiviral drug design. Here, we review the different pathways and mechanisms used to produce viral mRNA 5′-caps, and present current structures, mechanisms, and inhibitors known to act on viral RNA capping.
Collapse
Affiliation(s)
- François Ferron
- Centre National de la Recherche Scientifique and Aix-Marseille Université, UMR 7257, Architecture et Fonction des Macromolécules Biologiques, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | | | | | | |
Collapse
|
38
|
Jia Y, Polunovsky V, Bitterman PB, Wagner CR. Cap-dependent translation initiation factor eIF4E: an emerging anticancer drug target. Med Res Rev 2012; 32:786-814. [PMID: 22495651 PMCID: PMC7168506 DOI: 10.1002/med.21260] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer cells tend to be more highly dependent on cap‐dependent translation than normal tissues. Thus, proteins involved in the initiation of cap‐dependent translation have emerged as potential anti‐cancer drug targets. Cap‐dependent translation is initiated by the binding of the factor eIF4E to the cap domain of mRNA. Detailed x‐ray crystal and NMR structures are available for eIF4E in association with cap‐analogs, as well as domains of other initiation factors. This review will summarize efforts to design potential antagonist of eIF4E that could be used as new pharmacological tools and anti‐cancer agents and. Insights drawn from these studies should aid in the design of future inhibitors of eIF4E dependent translation initiation. © 2012 Wiley Periodicals, Inc. Med Res Rev., 32, No. 4, 786‐814, 2012
Collapse
Affiliation(s)
- Yan Jia
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
39
|
Tremblay-Létourneau M, Despins S, Bougie I, Bisaillon M. Virtual high-throughput screening identifies mycophenolic acid as a novel RNA capping inhibitor. PLoS One 2011; 6:e24806. [PMID: 21935470 PMCID: PMC3174198 DOI: 10.1371/journal.pone.0024806] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/18/2011] [Indexed: 12/25/2022] Open
Abstract
The RNA guanylyltransferase (GTase) is involved in the synthesis of the m7Gppp-RNA cap structure found at the 5′ end of eukaryotic mRNAs. GTases are members of the covalent nucleotidyl transferase superfamily, which also includes DNA and RNA ligases. GTases catalyze a two-step reaction in which they initially utilize GTP as a substrate to form a covalent enzyme-GMP intermediate. The GMP moiety is then transferred to the diphosphate end of the RNA transcript in the second step of the reaction to form the Gppp-RNA structure. In the current study, we used a combination of virtual database screening, homology modeling, and biochemical assays to search for novel GTase inhibitors. Using this approach, we demonstrate that mycophenolic acid (MPA) can inhibit the GTase reaction by preventing the catalytic transfer of the GMP moiety onto an acceptor RNA. As such, MPA represents a novel type of inhibitor against RNA guanylyltransferases that inhibits the second step of the catalytic reaction. Moreover, we show that the addition of MPA to S. cerevisiae cells leads to a reduction of capped mRNAs. Finally, biochemical assays also demonstrate that MPA can inhibit DNA ligases through inhibition of the second step of the reaction. The biological implications of these findings for the MPA-mediated inhibition of members of the covalent nucleotidyl superfamily are discussed.
Collapse
Affiliation(s)
- Maude Tremblay-Létourneau
- RNA Group, Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Simon Despins
- RNA Group, Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Isabelle Bougie
- RNA Group, Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Bisaillon
- RNA Group, Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- * E-mail:
| |
Collapse
|
40
|
Inhibitory effect of a nucleotide analog on infectious salmon anemia virus infection. J Virol 2011; 85:8037-45. [PMID: 21653663 DOI: 10.1128/jvi.00533-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The infectious salmon anemia virus (ISAV), which belongs to the Orthomyxoviridae family, has been responsible for major losses in the salmon industry, with mortalities close to 100% in areas where Atlantic salmon (Salmo salar) is grown. This work studied the effect of ribavirin (1-β-d-ribofuranosyl-1,2,3-triazole-3-carbaxaide), a broad-spectrum antiviral compound with proven ability to inhibit the replicative cycle of the DNA and RNA viruses. The results show that ribavirin was able to inhibit the infectivity of ISAV in in vitro assays. In these assays, a significant inhibition of the replicative viral cycle was observed with a 50% inhibitory concentration (IC₅₀) of 0.02 μg/ml and an IC₉₀ of 0.4 μg/ml of ribavirin. After ribavirin treatment, viral proteins were not detectable and a reduction of viral mRNA association with ribosomes was observed. Ribavirin does not affect the levels of EF1a, nor its association with polysomes, suggesting that the inhibition of RNA synthesis occurs specifically for the virus mRNAs and not for cellular mRNAs. Moreover, ribavirin caused a significant reduction in genomic and viral RNA messenger levels. The study of the inhibitory mechanism showed that it was not reversed by the addition of guanosine. Furthermore, in vivo assays showed a reduction in the mortality of Salmo salar by more than 90% in fish infected with ISAV and treated with ribavirin without adverse effects. In fact, these results show that ribavirin is an antiviral that could be used to prevent ISAV replication either in vitro or in vivo.
Collapse
|
41
|
Brillanti S, Mazzella G, Roda E. Ribavirin for chronic hepatitis C: and the mystery goes on. Dig Liver Dis 2011; 43:425-30. [PMID: 21093391 DOI: 10.1016/j.dld.2010.10.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 10/12/2010] [Indexed: 12/11/2022]
Abstract
Twenty years ago, ribavirin was first used in the treatment for chronic hepatitis C. After few years, ribavirin, in combination with interferon-alpha, showed a dramatic synergistic efficacy against hepatitis C virus infection, leading to viral clearance in about 50% of patients. Recent discovery of potent inhibitors of hepatitis C virus proteases did not replace ribavirin as the mainstay of combination therapy for chronic hepatitis C. Despite this fundamental role of ribavirin, many aspects of the mechanism of action and of the optimal dose and duration of therapy remain to be discovered or settled. In the present review, the authors recall the milestones in the history of ribavirin and try to shed light on the more relevant features of ribavirin action and utilization, and on the clinical problems encountered in managing and optimizing treatment for chronic hepatitis C. Finally, some potential off-label use of this drug in most difficult-to-treat subjects is pointed out. In conclusion, even if a sort of mystery surrounds ribavirin, its efficacy against hepatitis C virus infection fortunately remains lasting and stable.
Collapse
Affiliation(s)
- Stefano Brillanti
- Divisione di Gastroenterologia, Policlinico S. Orsola - Malpighi, Bologna, Italy.
| | | | | |
Collapse
|
42
|
Issur M, Picard‐Jean F, Bisaillon M. The RNA capping machinery as an anti-infective target. WILEY INTERDISCIPLINARY REVIEWS. RNA 2011; 2:184-92. [PMID: 21957005 PMCID: PMC7169851 DOI: 10.1002/wrna.43] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A number of different human pathogens code for their own enzymes involved in the synthesis of the RNA cap structure. Although the RNA cap structures originating from human and microbial enzymes are often identical, the subunit composition, structure and catalytic mechanisms of the microbial-encoded enzymes involved in the synthesis of the RNA cap structure are often significantly different from those of host cells. As a consequence, these pathogenic cap-forming enzymes are potential targets for antimicrobial drugs. During the past few years, experimental studies have started to demonstrate that inhibition of the RNA capping activity is a reasonable approach for the development of antimicrobial agents. The combination of structural, biochemical, and molecular modeling studies are starting to reveal novel molecules that can serve as starting blocks for the design of more potent and specific antimicrobial agents. Here, we examine various strategies that have been developed to inhibit microbial enzymes involved in the synthesis of the RNA cap structure, emphasizing the challenges remaining to design potent and selective drugs.
Collapse
Affiliation(s)
- Moheshwarnath Issur
- Département de Biochimie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| | - Frédéric Picard‐Jean
- Département de Biochimie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| | - Martin Bisaillon
- Département de Biochimie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| |
Collapse
|
43
|
Bollati M, Alvarez K, Assenberg R, Baronti C, Canard B, Cook S, Coutard B, Decroly E, de Lamballerie X, Gould EA, Grard G, Grimes JM, Hilgenfeld R, Jansson AM, Malet H, Mancini EJ, Mastrangelo E, Mattevi A, Milani M, Moureau G, Neyts J, Owens RJ, Ren J, Selisko B, Speroni S, Steuber H, Stuart DI, Unge T, Bolognesi M. Structure and functionality in flavivirus NS-proteins: perspectives for drug design. Antiviral Res 2010; 87:125-48. [PMID: 19945487 PMCID: PMC3918146 DOI: 10.1016/j.antiviral.2009.11.009] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 09/08/2009] [Accepted: 11/21/2009] [Indexed: 12/28/2022]
Abstract
Flaviviridae are small enveloped viruses hosting a positive-sense single-stranded RNA genome. Besides yellow fever virus, a landmark case in the history of virology, members of the Flavivirus genus, such as West Nile virus and dengue virus, are increasingly gaining attention due to their re-emergence and incidence in different areas of the world. Additional environmental and demographic considerations suggest that novel or known flaviviruses will continue to emerge in the future. Nevertheless, up to few years ago flaviviruses were considered low interest candidates for drug design. At the start of the European Union VIZIER Project, in 2004, just two crystal structures of protein domains from the flaviviral replication machinery were known. Such pioneering studies, however, indicated the flaviviral replication complex as a promising target for the development of antiviral compounds. Here we review structural and functional aspects emerging from the characterization of two main components (NS3 and NS5 proteins) of the flavivirus replication complex. Most of the reviewed results were achieved within the European Union VIZIER Project, and cover topics that span from viral genomics to structural biology and inhibition mechanisms. The ultimate aim of the reported approaches is to shed light on the design and development of antiviral drug leads.
Collapse
Affiliation(s)
- Michela Bollati
- Department of Biomolecular Sciences and Biotechnology, University of Milano, Via Celoria 26, 20133 Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Issur M, Geiss BJ, Bougie I, Picard-Jean F, Despins S, Mayette J, Hobdey SE, Bisaillon M. The flavivirus NS5 protein is a true RNA guanylyltransferase that catalyzes a two-step reaction to form the RNA cap structure. RNA (NEW YORK, N.Y.) 2009; 15:2340-2350. [PMID: 19850911 PMCID: PMC2779676 DOI: 10.1261/rna.1609709] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Accepted: 08/31/2009] [Indexed: 05/28/2023]
Abstract
The 5'-end of the flavivirus genome harbors a methylated (m7)GpppA(2'OMe) cap structure, which is generated by the virus-encoded RNA triphosphatase, RNA (guanine-N7) methyltransferase, nucleoside 2'-O-methyltransferase, and RNA guanylyltransferase. The presence of the flavivirus guanylyltransferase activity in NS5 has been suggested by several groups but has not been empirically proven. Here we provide evidence that the N-terminus of the flavivirus NS5 protein is a true RNA guanylyltransferase. We demonstrate that GTP can be used as a substrate by the enzyme to form a covalent GMP-enzyme intermediate via a phosphoamide bond. Mutational studies also confirm the importance of a specific lysine residue in the GTP binding site for the enzymatic activity. We show that the GMP moiety can be transferred to the diphosphate end of an RNA transcript harboring an adenosine as the initiating residue. We also demonstrate that the flavivirus RNA triphosphatase (NS3 protein) stimulates the RNA guanylyltransferase activity of the NS5 protein. Finally, we show that both enzymes are sufficient and necessary to catalyze the de novo formation of a methylated RNA cap structure in vitro using a triphosphorylated RNA transcript. Our study provides biochemical evidence that flaviviruses encode a complete RNA capping machinery.
Collapse
Affiliation(s)
- Moheshwarnath Issur
- Département de Biochimie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Pecou S, Moinard N, Walschaerts M, Pasquier C, Daudin M, Bujan L. Ribavirin and pegylated interferon treatment for hepatitis C was associated not only with semen alterations but also with sperm deoxyribonucleic acid fragmentation in humans. Fertil Steril 2008; 91:933.e17-22. [PMID: 18930227 DOI: 10.1016/j.fertnstert.2008.07.1755] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2008] [Revised: 07/17/2008] [Accepted: 07/23/2008] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To report the effects of ribavirin plus pegylated interferon on semen parameters and sperm DNA integrity in a man given treatment for hepatitis C. DESIGN Case report. SETTING University-affiliated teaching hospital. INTERVENTION(S) None. PATIENT(S) A 37-year-old man given treatment with ribavirin and pegylated interferon for hepatitis C. MAIN OUTCOME MEASURE(S) Semen parameters (sperm count, motility, round cells), sperm protamination state measured by sperm chromatin structure assay, and sperm DNA fragmentation measured by terminal uridine nick-end labeling assay. RESULT(S) The percentage of progressive spermatozoa and the number of motile sperm per ejaculate decreased during treatment. The round cell/spermatozoa ratio, which reflects spermatogenic abnormality, increased from 2.6% +/- 1.4% to 23.6% +/- 13.0% during treatment and returned to baseline value 4 months later. Moreover, the sperm DNA fragmentation index (as measured by sperm chromatin structure assay) increased very markedly during treatment (from 14.5% before to 69.2% at 7 months of treatment) and remained elevated 8 months later. CONCLUSION(S) This study reports for the first time not only quantitative but also qualitative alterations of spermatogenesis with DNA packaging abnormalities and emphasizes the need for prospective clinical studies. While the results of other studies are awaited, the alterations that persisted 8 months after treatment argue for a longer contraception period after treatment discontinuation in men.
Collapse
Affiliation(s)
- Stephane Pecou
- Human Fertility Research Group, Université Toulouse III Paul Sabatier (EA 3694), Toulouse, France
| | | | | | | | | | | |
Collapse
|
46
|
Lee BJ, Matsunaga H, Ikuta K, Tomonaga K. Ribavirin inhibits Borna disease virus proliferation and fatal neurological diseases in neonatally infected gerbils. Antiviral Res 2008; 80:380-4. [PMID: 18778737 DOI: 10.1016/j.antiviral.2008.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 08/01/2008] [Accepted: 08/07/2008] [Indexed: 11/30/2022]
Abstract
By using neonatal gerbils, we assessed the effect of ribavirin on the proliferation of Borna disease virus (BDV) in the brain. The intracranial inoculation of ribavirin reduced viral propagation in the acutely infected brain, resulting in protection from fatal neurological disorders. We found that the treatment with ribavirin markedly reduces the numbers of OX-42-positive microglial cells, but does not activate expression of Th1 cytokines, in BDV-infected gerbil brains. Our results suggested that ribavirin directly inhibits BDV replication and might be a potential tool for the treatment of BDV infection.
Collapse
Affiliation(s)
- Byeong-Jae Lee
- Department of Virology, Research Institute for Microbial Diseases (BIKEN), Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
47
|
Silverman JEY, Ciustea M, Shudofsky AMD, Bender F, Shoemaker RH, Ricciardi RP. Identification of polymerase and processivity inhibitors of vaccinia DNA synthesis using a stepwise screening approach. Antiviral Res 2008; 80:114-23. [PMID: 18621425 DOI: 10.1016/j.antiviral.2008.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 05/08/2008] [Accepted: 05/14/2008] [Indexed: 12/16/2022]
Abstract
Nearly all DNA polymerases require processivity factors to ensure continuous incorporation of nucleotides. Processivity factors are specific for their cognate DNA polymerases. For this reason, the vaccinia DNA polymerase (E9) and the proteins associated with processivity (A20 and D4) are excellent therapeutic targets. In this study, we show the utility of stepwise rapid plate assays that (i) screen for compounds that block vaccinia DNA synthesis, (ii) eliminate trivial inhibitors, e.g. DNA intercalators, and (iii) distinguish whether inhibitors are specific for blocking DNA polymerase activity or processivity. The sequential plate screening of 2222 compounds from the NCI Diversity Set library yielded a DNA polymerase inhibitor (NSC 55636) and a processivity inhibitor (NSC 123526) that were capable of reducing vaccinia viral plaques with minimal cellular cytotoxicity. These compounds are predicted to block cellular infection by the smallpox virus, variola, based on the very high sequence identity between A20, D4 and E9 of vaccinia and the corresponding proteins of variola.
Collapse
Affiliation(s)
- Janice Elaine Y Silverman
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
48
|
Leyssen P, De Clercq E, Neyts J. Molecular strategies to inhibit the replication of RNA viruses. Antiviral Res 2008; 78:9-25. [PMID: 18313769 PMCID: PMC7114363 DOI: 10.1016/j.antiviral.2008.01.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 01/04/2008] [Accepted: 01/08/2008] [Indexed: 12/24/2022]
Abstract
There are virtually no antiviral drugs available for the treatment of infections with RNA viruses. This is particularly worrisome since most of the highly pathogenic and emerging viruses are, and will likely continue to be, RNA viruses. These viruses can cause acute, severe illness, including severe respiratory disease, hemorrhagic fever and encephalitis, with a high case fatality rate. It is important to have potent and safe drugs at hand that can be used for the treatment or prophylaxis of such infections. Drugs approved for the treatment of RNA virus infections (other than HIV) are the influenza M2 channel inhibitors, amantadine and rimantadine; the influenza neuraminidase inhibitors, oseltamivir and zanamivir, and ribavirin for the treatment of infections with respiratory syncytial virus and hepatitis C virus. The molecular mechanism(s) by which ribavirin inhibits viral replication, such as depletion of intracellular GTP pools and induction of error catastrophe, may not readily allow the design of analogues that are more potent/selective than the parent drug. Highly pathogenic RNA viruses belong to a variety of virus families, each having a particular replication strategy, thus offering a wealth of potential targets to selectively inhibit viral replication. We here provide a non-exhaustive review of potential experimental strategies, using small molecules, to inhibit the replication of several RNA viruses. Other approaches, such as the use of interferon or other host-response modifiers, immune serum or neutralizing antibodies, are not addressed in this review.
Collapse
Affiliation(s)
| | | | - Johan Neyts
- Rega Institute for Medical Research, Catholic University Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| |
Collapse
|
49
|
Soulière MF, Perreault JP, Bisaillon M. Magnesium-binding studies reveal fundamental differences between closely related RNA triphosphatases. Nucleic Acids Res 2007; 36:451-61. [PMID: 18039706 PMCID: PMC2241848 DOI: 10.1093/nar/gkm1067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The Chlorella virus RNA triphosphatase (cvRTPase) is involved in the formation of the RNA cap structure found at the 5′-end of the viral mRNAs and requires magnesium ions to mediate its catalytic activity. To extend our studies on the role of metal ions in phosphohydrolysis, we have used a combination of fluorescence spectroscopy, circular dichroism, denaturation studies and thermodynamic analyses to monitor the binding of magnesium ions to the cvRTPase. Using these techniques, the thermodynamic forces responsible for the interaction of metal ions with an RNA triphosphatase were also evaluated for the first time. Our thermodynamic analyses indicate that the initial association of magnesium with the cvRTPase is dominated by a favorable entropic effect and is accompanied by the release of eight water molecules from the enzyme. Moreover, both fluorescence spectroscopy and circular dichroism assays indicated that minor conformational changes were occurring upon magnesium binding. Mutational studies were also performed and confirmed the importance of three specific glutamate residues located in the active site of the enzyme for the binding of magnesium ions. Finally, in contrast to the yeast RNA triphosphatase, we demonstrate that the binding of magnesium ions to the cvRTPase does not lead to the stabilization of the ground state binding of the RNA substrate. Based on the results of the present study, we hypothesize that the binding of magnesium ions induces local conformational perturbations in the active site residues that ultimately positions the lateral chains of critical amino acids involved in catalysis. Our results highlight fundamental differences in the role of magnesium ions in the phosphohydrolase reactions catalyzed by the cvRTPase and the closely related yeast RNA triphosphatase.
Collapse
Affiliation(s)
- Marie F Soulière
- Département de Biochimie, RNA Group, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | | | | |
Collapse
|
50
|
Elia G, Belloli C, Cirone F, Lucente MS, Caruso M, Martella V, Decaro N, Buonavoglia C, Ormas P. In vitro efficacy of ribavirin against canine distemper virus. Antiviral Res 2007; 77:108-13. [PMID: 17949825 DOI: 10.1016/j.antiviral.2007.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 09/12/2007] [Accepted: 09/12/2007] [Indexed: 01/21/2023]
Abstract
Despite vaccination, canine distemper virus (CDV) remains one of the important pathogen of dogs with worldwide distribution. Ribavirin (RIB) inhibits replication of measles virus (MV), a morbillivirus closely related to CDV, both in vitro and in vivo. In this report the antiviral activity of RIB against CDV in cell cultures was assessed. Quantitative real-time RT-PCR was used to measure viral RNA in VERO cells infected by CDV and to evaluate the inhibitory effects of RIB. RIB caused a dose- and time-dependent decrease in accumulation of CDV RNA when added after virus adsorption. RIB was highly effective in preventing CDV replication at low concentrations with 50% virus-inhibitory concentrations ranging from 0.02 to 0.05 mM. Such low values were comparable to values displayed by highly susceptible strains of MV. In addition, CDV was passaged sequentially in VERO cell monolayers in the presence of RIB to trigger viral extinction. The virus was no longer detected after three passages, suggesting that error catastrophe is one of the modes of action of RIB against CDV. These findings suggest RIB as a promising tool for the therapy of CD in dogs.
Collapse
Affiliation(s)
- Gabriella Elia
- Department of Animal Health and Well-being, Faculty of Veterinary Medicine of Bari, 70010 Valenzano, Bari, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|