51
|
Lee YCJ, Shirkey JD, Park J, Bisht K, Cowan AJ. An Overview of Antiviral Peptides and Rational Biodesign Considerations. BIODESIGN RESEARCH 2022; 2022:9898241. [PMID: 37850133 PMCID: PMC10521750 DOI: 10.34133/2022/9898241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 10/19/2023] Open
Abstract
Viral diseases have contributed significantly to worldwide morbidity and mortality throughout history. Despite the existence of therapeutic treatments for many viral infections, antiviral resistance and the threat posed by novel viruses highlight the need for an increased number of effective therapeutics. In addition to small molecule drugs and biologics, antimicrobial peptides (AMPs) represent an emerging class of potential antiviral therapeutics. While AMPs have traditionally been regarded in the context of their antibacterial activities, many AMPs are now known to be antiviral. These antiviral peptides (AVPs) have been shown to target and perturb viral membrane envelopes and inhibit various stages of the viral life cycle, from preattachment inhibition through viral release from infected host cells. Rational design of AMPs has also proven effective in identifying highly active and specific peptides and can aid in the discovery of lead peptides with high therapeutic selectivity. In this review, we highlight AVPs with strong antiviral activity largely curated from a publicly available AMP database. We then compile the sequences present in our AVP database to generate structural predictions of generic AVP motifs. Finally, we cover the rational design approaches available for AVPs taking into account approaches currently used for the rational design of AMPs.
Collapse
Affiliation(s)
- Ying-Chiang J. Lee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jaden D. Shirkey
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jongbeom Park
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Karishma Bisht
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Alexis J. Cowan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
52
|
Olasunkanmi OI, Mageto J, Avala Ntsigouaye J, Yi M, Fei Y, Chen Y, Chen S, Xu W, Lin L, Zhao W, Wang Y, Zhong ZH. Novel Antiviral Activity of Ethyl 3-Hydroxyhexanoate Against Coxsackievirus B Infection. Front Microbiol 2022; 13:875485. [PMID: 35495645 PMCID: PMC9048257 DOI: 10.3389/fmicb.2022.875485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 01/26/2023] Open
Abstract
Coxsackievirus group B (CVB) is a member of the genus Enterovirus in the family Picornaviridae. CVB infection has been implicated as a major etiologic agent of viral myocarditis, dilated cardiomyopathy, meningitis, and pancreatitis among children and young adults. Until date, no antiviral agent has been licensed for the treatment of Coxsackievirus infection. In an effort to identify antiviral agents against diseases caused by the CVB, we found that ethyl 3-hydroxyhexanoate (EHX), a volatile compound present in fruits and food additives, is a potent antiviral compound. In this study, we demonstrated that EHX treatment significantly inhibits CVB replication both in vivo and in vitro. Furthermore, EHX possesses antiviral activity at 50% effective concentration (EC50) of 1.2 μM and 50% cytotoxicity (CC50) of 25.6 μM, yielding a selective index (SI) value as high as 20.8. Insights into the mechanism of antiviral activity of EHX showed that it acts at the step of viral RNA replication. Since EHX has received approval as food additives, treatment of CVB-related infections with EHX might be a safe therapeutic option and may be a promising strategy for the development of semi-synthetic antiviral drugs for viral diseases.
Collapse
Affiliation(s)
| | - James Mageto
- Department of Microbiology, Harbin Medical University, Harbin, China
| | | | - Ming Yi
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Yanru Fei
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Yang Chen
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Sijia Chen
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Weizhen Xu
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Lexun Lin
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Wenran Zhao
- Department of Cell Biology, Harbin Medical University, Harbin, China
| | - Yan Wang
- Department of Cell Biology, Harbin Medical University, Harbin, China
| | - Zhao-Hua Zhong
- Department of Microbiology, Harbin Medical University, Harbin, China
| |
Collapse
|
53
|
Khan F, Tabassum N, Bamunuarachchi NI, Kim YM. Phloroglucinol and Its Derivatives: Antimicrobial Properties toward Microbial Pathogens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:4817-4838. [PMID: 35418233 DOI: 10.1021/acs.jafc.2c00532] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Phloroglucinol (PG) is a natural product isolated from plants, algae, and microorganisms. Aside from that, the number of PG derivatives has expanded due to the discovery of their potential biological roles. Aside from its diverse biological activities, PG and its derivatives have been widely utilized to treat microbial infections caused by bacteria, fungus, and viruses. The rapid emergence of antimicrobial-resistant microbial infections necessitates the chemical synthesis of numerous PG derivatives in order to meet the growing demand for drugs. This review focuses on the use of PG and its derivatives to control microbial infection and the underlying mechanism of action. Furthermore, as future perspectives, some of the various alternative strategies, such as the use of PG and its derivatives in conjugation, nanoformulation, antibiotic combination, and encapsulation, have been thoroughly discussed. This review will enable the researcher to investigate the possible antibacterial properties of PG and its derivatives, either free or in the form of various formulations.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Nazia Tabassum
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | | | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
54
|
Keum H, Yoo D, Jon S. Photomedicine based on heme-derived compounds. Adv Drug Deliv Rev 2022; 182:114134. [PMID: 35122881 DOI: 10.1016/j.addr.2022.114134] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/16/2021] [Accepted: 01/30/2022] [Indexed: 02/08/2023]
Abstract
Photoimaging and phototherapy have become major platforms for the diagnosis and treatment of various health complications. These applications require a photosensitizer (PS) that is capable of absorbing light from a source and converting it into other energy forms for detection and therapy. While synthetic inorganic materials such as quantum dots and gold nanorods have been widely explored for their medical diagnosis and photodynamic (PDT) and photothermal (PTT) therapy capabilities, translation of these technologies has lagged, primarily owing to potential cytotoxicity and immunogenicity issues. Of the various photoreactive molecules, the naturally occurring endogenous compound heme, a constituent of red blood cells, and its derivatives, porphyrin, biliverdin and bilirubin, have shown immense potential as noteworthy candidates for clinically translatable photoreactive agents, as evidenced by previous reports. While porphyrin-based photomedicines have attracted significant attention and are well documented, research on photomedicines based on two other heme-derived compounds, biliverdin and bilirubin, has been relatively lacking. In this review, we summarize the unique photoproperties of heme-derived compounds and outline recent efforts to use them in biomedical imaging and phototherapy applications.
Collapse
|
55
|
Claus-Desbonnet H, Nikly E, Nalbantova V, Karcheva-Bahchevanska D, Ivanova S, Pierre G, Benbassat N, Katsarov P, Michaud P, Lukova P, Delattre C. Polysaccharides and Their Derivatives as Potential Antiviral Molecules. Viruses 2022; 14:426. [PMID: 35216019 PMCID: PMC8879384 DOI: 10.3390/v14020426] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023] Open
Abstract
In the current context of the COVID-19 pandemic, it appears that our scientific resources and the medical community are not sufficiently developed to combat rapid viral spread all over the world. A number of viruses causing epidemics have already disseminated across the world in the last few years, such as the dengue or chinkungunya virus, the Ebola virus, and other coronavirus families such as Middle East respiratory syndrome (MERS-CoV) and severe acute respiratory syndrome (SARS-CoV). The outbreaks of these infectious diseases have demonstrated the difficulty of treating an epidemic before the creation of vaccine. Different antiviral drugs already exist. However, several of them cause side effects or have lost their efficiency because of virus mutations. It is essential to develop new antiviral strategies, but ones that rely on more natural compounds to decrease the secondary effects. Polysaccharides, which have come to be known in recent years for their medicinal properties, including antiviral activities, are an excellent alternative. They are essential for the metabolism of plants, microorganisms, and animals, and are directly extractible. Polysaccharides have attracted more and more attention due to their therapeutic properties, low toxicity, and availability, and seem to be attractive candidates as antiviral drugs of tomorrow.
Collapse
Affiliation(s)
- Hadrien Claus-Desbonnet
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, F-63000 Clermont-Ferrand, France; (H.C.-D.); (E.N.); (G.P.); (P.M.)
| | - Elsa Nikly
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, F-63000 Clermont-Ferrand, France; (H.C.-D.); (E.N.); (G.P.); (P.M.)
| | - Vanya Nalbantova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (V.N.); (D.K.-B.); (N.B.); (P.L.)
| | - Diana Karcheva-Bahchevanska
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (V.N.); (D.K.-B.); (N.B.); (P.L.)
| | - Stanislava Ivanova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (V.N.); (D.K.-B.); (N.B.); (P.L.)
| | - Guillaume Pierre
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, F-63000 Clermont-Ferrand, France; (H.C.-D.); (E.N.); (G.P.); (P.M.)
| | - Niko Benbassat
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (V.N.); (D.K.-B.); (N.B.); (P.L.)
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Medical University Sofia, 1000 Sofia, Bulgaria
| | - Plamen Katsarov
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria;
- Research Institute, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Philippe Michaud
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, F-63000 Clermont-Ferrand, France; (H.C.-D.); (E.N.); (G.P.); (P.M.)
| | - Paolina Lukova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (V.N.); (D.K.-B.); (N.B.); (P.L.)
| | - Cédric Delattre
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, F-63000 Clermont-Ferrand, France; (H.C.-D.); (E.N.); (G.P.); (P.M.)
- Institut Universitaire de France (IUF), 1 Rue Descartes, 75005 Paris, France
| |
Collapse
|
56
|
Delcanale P, Abbruzzetti S, Viappiani C. Photodynamic treatment of pathogens. LA RIVISTA DEL NUOVO CIMENTO 2022; 45:407-459. [PMCID: PMC8921710 DOI: 10.1007/s40766-022-00031-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/10/2022] [Indexed: 06/01/2023]
Abstract
The current viral pandemic has highlighted the compelling need for effective and versatile treatments, that can be quickly tuned to tackle new threats, and are robust against mutations. Development of such treatments is made even more urgent in view of the decreasing effectiveness of current antibiotics, that makes microbial infections the next emerging global threat. Photodynamic effect is one such method. It relies on physical processes proceeding from excited states of particular organic molecules, called photosensitizers, generated upon absorption of visible or near infrared light. The excited states of these molecules, tailored to undergo efficient intersystem crossing, interact with molecular oxygen and generate short lived reactive oxygen species (ROS), mostly singlet oxygen. These species are highly cytotoxic through non-specific oxidation reactions and constitute the basis of the treatment. In spite of the apparent simplicity of the principle, the method still has to face important challenges. For instance, the short lifetime of ROS means that the photosensitizer must reach the target within a few tens nanometers, which requires proper molecular engineering at the nanoscale level. Photoactive nanostructures thus engineered should ideally comprise a functionality that turns the system into a theranostic means, for instance, through introduction of fluorophores suitable for nanoscopy. We discuss the principles of the method and the current molecular strategies that have been and still are being explored in antimicrobial and antiviral photodynamic treatment.
Collapse
Affiliation(s)
- Pietro Delcanale
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università degli Studi di Parma, Parco Area delle Scienze 7A, 43124 Parma, Italy
| | - Stefania Abbruzzetti
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università degli Studi di Parma, Parco Area delle Scienze 7A, 43124 Parma, Italy
| | - Cristiano Viappiani
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università degli Studi di Parma, Parco Area delle Scienze 7A, 43124 Parma, Italy
| |
Collapse
|
57
|
Zenchenko AA, Drenichev MS, Il’icheva IA, Mikhailov SN. Antiviral and Antimicrobial Nucleoside Derivatives: Structural Features and Mechanisms of Action. Mol Biol 2021; 55:786-812. [PMID: 34955556 PMCID: PMC8682041 DOI: 10.1134/s0026893321040105] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 11/23/2022]
Abstract
The emergence of new viruses and resistant strains of pathogenic microorganisms has become a powerful stimulus in the search for new drugs. Nucleosides are a promising class of natural compounds, and more than a hundred drugs have already been created based on them, including antiviral, antibacterial and antitumor agents. The review considers the structural and functional features and mechanisms of action of known nucleoside analogs with antiviral, antibacterial or antiprotozoal activity. Particular attention is paid to the mechanisms that determine the antiviral effect of nucleoside analogs containing hydrophobic fragments. Depending on the structure and position of the hydrophobic substituent, such nucleosides can either block the process of penetration of viruses into cells or inhibit the stage of genome replication. The mechanisms of inhibition of viral enzymes by compounds of nucleoside and non-nucleoside nature have been compared. The stages of creation of antiparasitic drugs, which are based on the peculiarities of metabolic transformations of nucleosides in humans body and parasites, have been considered. A new approach to the creation of drugs is described, based on the use of prodrugs of modified nucleosides, which, as a result of metabolic processes, are converted into an effective drug directly in the target organ or tissue. This strategy makes it possible to reduce the general toxicity of the drug to humans and to increase the effectiveness of its action on cells infected by the virus.
Collapse
Affiliation(s)
- A. A. Zenchenko
- Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia
| | - M. S. Drenichev
- Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia
| | - I. A. Il’icheva
- Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia
| | - S. N. Mikhailov
- Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia
| |
Collapse
|
58
|
Mathew C, Tamir S, Tripp RA, Ghildyal R. Reversible disruption of XPO1-mediated nuclear export inhibits respiratory syncytial virus (RSV) replication. Sci Rep 2021; 11:19223. [PMID: 34584169 PMCID: PMC8479129 DOI: 10.1038/s41598-021-98767-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/13/2021] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the primary cause of serious lower respiratory tract disease in infants, young children, the elderly and immunocompromised individuals. Therapy for RSV infections is limited to high risk infants and there are no safe and efficacious vaccines. Matrix (M) protein is a major RSV structural protein with a key role in virus assembly. Interestingly, M is localised to the nucleus early in infection and its export into the cytoplasm by the nuclear exporter, exportin-1 (XPO1) is essential for RSV assembly. We have shown previously that chemical inhibition of XPO1 function results in reduced RSV replication. In this study, we have investigated the anti-RSV efficacy of Selective Inhibitor of Nuclear Export (SINE) compounds, KPT-335 and KPT-185. Our data shows that therapeutic administration of the SINE compounds results in reduced RSV titre in human respiratory epithelial cell culture. Within 24 h of treatment, RSV replication and XPO1 expression was reduced, M protein was partially retained in the nucleus, and cell cycle progression was delayed. Notably, the effect of SINE compounds was reversible within 24 h after their removal. Our data show that reversible inhibition of XPO1 can disrupt RSV replication by affecting downstream pathways regulated by the nuclear exporter.
Collapse
Affiliation(s)
- Cynthia Mathew
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia
| | | | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Reena Ghildyal
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT 2617, Australia.
| |
Collapse
|
59
|
Bhutta ZA, Kanwal A, Ali M, Kulyar MFEA, Yao W, Shoaib M, Ashar A, Mahfooz A, Ijaz M, Ijaz N, Asif M, Nawaz S, Mahfooz MR, Kanwal T. Emerging nanotechnology role in the development of innovative solutions against COVID-19 pandemic. NANOTECHNOLOGY 2021; 32:482001. [PMID: 34320471 DOI: 10.1088/1361-6528/ac189e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/28/2021] [Indexed: 06/13/2023]
Abstract
The COVID-19 outbreak is creating severe impressions on all facets of the global community. Despite strong measures worldwide to try and re-achieve normalcy, the ability of SARS-CoV-2 to survive sturdy ecological settings may contribute to its rapid spread. Scientists from different aspects of life are working together to develop effective treatment strategies against SARS-CoV-2. Apart from using clinical devices for patient recovery, the key focus is on developing antiviral drugs and vaccines. Given the physical size of the SARS-CoV-2 pathogen and with the vaccine delivery platform currently undergoing clinical trials, the link between nanotechnology is clear, and previous antiviral research using nanomaterials confirms this link. Nanotechnology based products can effectively suppress various pathogens, including viruses, regardless of drug resistance, biological structure, or physiology. Thus, nanotechnology is opening up new dimensions for developing new strategies for diagnosing, preventing, treating COVID-19 and other viral ailments. This article describes the application of nanotechnology against the COVID-19 virus in terms of therapeutic purposes and vaccine development through the invention of nanomaterial based substances such as sanitizers (handwashing agents and surface disinfectants), masks and gowns, amongst other personal protective equipment, diagnostic tools, and nanocarrier systems, as well as the drawbacks and challenges of nanotechnology that need to be addressed.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, Scotland, United Kingdom
- Department of Clinical Medicine and Surgery, University of Agriculture Faisalabad, 38000, Pakistan
| | - Ayesha Kanwal
- Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Moazam Ali
- Department of Clinical Medicine and Surgery, University of Agriculture Faisalabad, 38000, Pakistan
| | | | - Wangyuan Yao
- College of Veterinary Medicine, Huazhong Agriculture University, Wuhan, 430070, People's Republic of China
| | - Muhammad Shoaib
- Institute of Microbiology, Faculty of Veterinary Science, University of Agriculture Faisalabad, 38000, Pakistan
| | - Ambreen Ashar
- Department of Chemistry, Government College Women University, Faisalabad, Pakistan
| | - Ashar Mahfooz
- Department of Clinical Medicine and Surgery, University of Agriculture Faisalabad, 38000, Pakistan
| | - Misbah Ijaz
- Department of Clinical Medicine and Surgery, University of Agriculture Faisalabad, 38000, Pakistan
| | - Nabeel Ijaz
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muhammad Asif
- Department of Surgery, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Shah Nawaz
- Department of Pathology, University of Agriculture Faisalabad, 38000, Pakistan
| | | | - Tahreem Kanwal
- Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, 63100, Pakistan
| |
Collapse
|
60
|
de Souza ML, de Albuquerque Wanderley Sales V, Alves L, Santos WM, Ferraz LR, Lima G, Mendes L, Rolim LA, Neto PJR. A systematic review of functionalized polymeric nanoparticles to improve intestinal permeability of drugs and biological products. Curr Pharm Des 2021; 28:410-426. [PMID: 34348618 DOI: 10.2174/1381612827666210804104205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The oral route is the most frequently used and the most convenient route of drug administration, since it has several advantages, such as ease of use, patient compliance and better cost-effectiveness. However, physicochemical and biopharmaceutical limitations of various active pharmaceutical ingredients (API) hinder suitability for this route, including degradation in the gastrointestinal tract, low intestinal permeability and low bioavailability. To overcome these problems, while maintaining therapeutic efficacy, polymeric nanoparticles have attracted considerable attention for their ability to increase drug solubility, promote controlled release, and improve stability. In addition, the functionalization of nanocarriers can increase uptake and accumulation at the target site of action, and intestinal absorption, making it possible to obtain more viable, safe and efficient treatments for oral administration. <P> Objective: This systematic review aimed to seek recent advances in the literature on the use of polymeric nanoparticles functionalization to increase intestinal permeability of APIs that are intended for oral administration. <P> Method: Two bibliographic databases were consulted (PubMed and ScienceDirect). The selected publications and the writing of this systematic review were based on the guidelines mentioned in the PRISMA statement. <P> Results: Out of a total of 3036 studies, 22 studies were included in this article based on our eligibility criteria. The results were consistent for the application of nanoparticle functionalization to increase intestinal permeability. <P> Conclusion: The functionalized polymeric nanoparticles can be considered as carrier systems that improve the intestinal permeability and bioavailability of APIs, with the potential to result, in the future, in the development of oral medicines.
Collapse
Affiliation(s)
- Myla Lôbo de Souza
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | | | - Larissa Alves
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Widson Michael Santos
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Leslie Raphael Ferraz
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Gustavo Lima
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Larissa Mendes
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| | - Larissa Araújo Rolim
- Central de Análise de Fármacos, Medicamentos e Alimentos. Federal University of Vale do São Francisco (UNIVASF), Petrolina-PE. Brazil
| | - Pedro José Rolim Neto
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Pernambuco. Brazil
| |
Collapse
|
61
|
Wang J, Huang Y, Lin Y, Wang Y. Exocellular polysaccharides extracted from mangrove fungus Paecilomyces Lilacinuson present anti-HSV-1 activity in mice. J Virol Methods 2021; 297:114246. [PMID: 34329630 DOI: 10.1016/j.jviromet.2021.114246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
This study examined the anti-HSV-1 activity of EPS extracts isolated from mangrove fungus Paecilomyces Lilacinuson after intraperitoneal administration in mice. Mice were experimentally infected with HSV-1 intracranially and treated intraperitoneally with three different doses of EPS extract (6 g/Kg, 8 g/Kg, and 10 g/Kg) for 7 days. One group of 15 mice was infected with HSV-1 but did not receive any treatment, while another group of 15 mice was mock-infected to remain a control group. Animals were observed twice a day for 14 days after virus infection, searching for clinical signs of weight loss, piloerection, isolation, or retardation movement. Compared with the mock-infected group, mortality was significantly increased (p < 0.05) in the virus-infected group and the groups that received 6 g/Kg and 8 g/Kg EPS extract. Interestingly, no significant differences in mortality were found between the 10 g/Kg EPS extract and the mock-infected group. Mortality in the 10 g/Kg EPS extract group was substantially improved compared with virus-infected(p < 0.05). Additionally, EPS extracts inhibited HSV-1 replication in the mice brain in a dose-dependent manner. Furthermore, the extracts decreased NF-κB protein and mRNA expression and the production of TNF-α in HSV-1-infected mice brain tissue. These effects were also dose-dependent. Our findings suggest that the EPS extract may be a potential candidate for developing an antiviral drug against HSV-1.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Key Laboratory of Tropical Translational Medicine of Hainan Medical University, Ministry of Education, Haikou, Hainan, 571199, China
| | - Yanni Huang
- Key Laboratory of Tropical Translational Medicine of Hainan Medical University, Ministry of Education, Haikou, Hainan, 571199, China
| | - Yingzi Lin
- Key Laboratory of Tropical Translational Medicine of Hainan Medical University, Ministry of Education, Haikou, Hainan, 571199, China
| | - Yongxia Wang
- Key Laboratory of Tropical Translational Medicine of Hainan Medical University, Ministry of Education, Haikou, Hainan, 571199, China.
| |
Collapse
|
62
|
Fotsing MCD, Njamen D, Tanee Fomum Z, Ndinteh DT. Synthesis of biologically active heterocyclic compounds from allenic and acetylenic nitriles and related compounds. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2020-0210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Cyclic and polycyclic compounds containing moieties such as imidazole, pyrazole, isoxazole, thiazoline, oxazine, indole, benzothiazole and benzoxazole benzimidazole are prized molecules because of the various pharmaceutical properties that they display. This led Prof. Landor and co-workers to engage in the synthesis of several of them such as alkylimidazolenes, oxazolines, thiazolines, pyrimidopyrimidines, pyridylpyrazoles, benzoxazines, quinolines, pyrimidobenzimidazoles and pyrimidobenzothiazolones. This review covers the synthesis of biologically active heterocyclic compounds by the Michael addition and the double Michael addition of various amines and diamines on allenic nitriles, acetylenic nitriles, hydroxyacetylenic nitriles, acetylenic acids and acetylenic aldehydes. The heterocycles were obtained in one step reaction and in most cases, did not give side products. A brief discussion on the biological activities of some heterocycles is also provided.
Collapse
Affiliation(s)
- Marthe Carine Djuidje Fotsing
- Department of Chemical Sciences , University of Johannesburg , Doornfontein Campus, P.O. BOX 17011 , Johannesburg , 2028 , South Africa
| | - Dieudonné Njamen
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology , University of Yaoundé I, Faculty of Sciences , P.O. Box , 812 Yaoundé , Yaoundé , Cameroon
| | - Zacharias Tanee Fomum
- Department of Organic Chemistry , University of Yaoundé I, Faculty of Sciences , P.O. Box , 812 Yaoundé , Yaoundé , Cameroon
| | - Derek Tantoh Ndinteh
- Department of Chemical Sciences , University of Johannesburg , Doornfontein Campus, P.O. BOX 17011 , Johannesburg , 2028 , South Africa
| |
Collapse
|
63
|
Al-Azzam S, Mhaidat NM, Banat HA, Alfaour M, Ahmad DS, Muller A, Al-Nuseirat A, Lattyak EA, Conway BR, Aldeyab MA. An Assessment of the Impact of Coronavirus Disease (COVID-19) Pandemic on National Antimicrobial Consumption in Jordan. Antibiotics (Basel) 2021; 10:690. [PMID: 34207567 PMCID: PMC8229725 DOI: 10.3390/antibiotics10060690] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 12/27/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has overlapping clinical characteristics with bacterial respiratory tract infection, leading to the prescription of potentially unnecessary antibiotics. This study aimed at measuring changes and patterns of national antimicrobial use for one year preceding and one year during the COVID-19 pandemic. Annual national antimicrobial consumption for 2019 and 2020 was obtained from the Jordan Food and Drug Administration (JFDA) following the WHO surveillance methods. The WHO Access, Watch, and Reserve (AWaRe) classification was used. Total antibiotic consumption in 2020 (26.8 DDD per 1000 inhabitants per day) decreased by 5.5% compared to 2019 (28.4 DDD per 1000 inhabitants per day). There was an increase in the use of several antibiotics during 2020 compared with 2019 (third generation cephalosporins (19%), carbapenems (52%), macrolides (57%), and lincosamides (106%)). In 2020, there was a marked reduction in amoxicillin use (-53%), while the use of azithromycin increased by 74%. National antimicrobial consumption of the Access group decreased by 18% from 2019 to 2020 (59.1% vs. 48.1% of total consumption). The use of the Watch group increased in 2020 by 26%. The study highlighted an increase in the use of certain antibiotics during the pandemic period that are known to be associated with increasing resistance. Efforts to enhance national antimicrobial stewardship are needed to ensure rational use of antimicrobials.
Collapse
Affiliation(s)
- Sayer Al-Azzam
- Clinical Pharmacy Department, Jordan University of Science and Technology, Irbid 22110, Jordan; (S.A.-A.); (N.M.M.)
| | - Nizar Mahmoud Mhaidat
- Clinical Pharmacy Department, Jordan University of Science and Technology, Irbid 22110, Jordan; (S.A.-A.); (N.M.M.)
| | - Hayaa A. Banat
- Jordan Food and Drug Administration (JFDA), Amman 11181, Jordan; (H.A.B.); (M.A.); (D.S.A.)
| | - Mohammad Alfaour
- Jordan Food and Drug Administration (JFDA), Amman 11181, Jordan; (H.A.B.); (M.A.); (D.S.A.)
| | - Dana Samih Ahmad
- Jordan Food and Drug Administration (JFDA), Amman 11181, Jordan; (H.A.B.); (M.A.); (D.S.A.)
| | - Arno Muller
- Antimicrobial Resistance Division, World Health Organization, Avenue Appia 20, 1211 Geneva, Switzerland;
| | - Adi Al-Nuseirat
- World Health Organization Regional Office for the Eastern Mediterranean, Cairo 11371, Egypt;
| | | | - Barbara R. Conway
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK;
- Institute of Skin Integrity and Infection Prevention, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Mamoon A. Aldeyab
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK;
| |
Collapse
|
64
|
Chihab H, Elmessaoudi-Idrissi M, Kitab B, Elfihry R, Jadid FZ, Zaidane I, Kettani A, Pineau P, Ezzikouri S, Benjelloun S. Molecular and computational analysis of natural drug resistance mutations among Moroccan chronic hepatitis B carriers. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
65
|
Abstract
The host immune system is highly compromised in case of viral infections and relapses are very common. The capacity of the virus to destroy the host cell by liberating its own DNA or RNA and replicating inside the host cell poses challenges in the development of antiviral therapeutics. In recent years, many new technologies have been explored for diagnosis, prevention, and treatment of viral infections. Nanotechnology has emerged as one of the most promising technologies on account of its ability to deal with viral diseases in an effective manner, addressing the limitations of traditional antiviral medicines. It has not only helped us to overcome problems related to solubility and toxicity of drugs, but also imparted unique properties to drugs, which in turn has increased their potency and selectivity toward viral cells against the host cells. The initial part of the paper focuses on some important proteins of influenza, Ebola, HIV, herpes, Zika, dengue, and corona virus and those of the host cells important for their entry and replication into the host cells. This is followed by different types of nanomaterials which have served as delivery vehicles for the antiviral drugs. It includes various lipid-based, polymer-based, lipid-polymer hybrid-based, carbon-based, inorganic metal-based, surface-modified, and stimuli-sensitive nanomaterials and their application in antiviral therapeutics. The authors also highlight newer promising treatment approaches like nanotraps, nanorobots, nanobubbles, nanofibers, nanodiamonds, nanovaccines, and mathematical modeling for the future. The paper has been updated with the recent developments in nanotechnology-based approaches in view of the ongoing pandemic of COVID-19.Graphical abstract.
Collapse
Affiliation(s)
- Malobika Chakravarty
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Amisha Vora
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
66
|
Bhat R, Begum NS. Synthesis, characterization and molecular docking studies of new indol(1 H-3-yl)pyrimidine derivatives: Insights into their role in DNA interaction. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2021; 40:619-634. [PMID: 33988075 DOI: 10.1080/15257770.2021.1922700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
This study reports the synthesis of new indol(1H-3-yl) pyrimidine derivatives 4(a-e) using various substituted indole-3-carbaldehydes, urea and malononitrile in the presence of ammonium chloride. The resulting compounds were characterized using analytical and spectroscopic methods. The molecular docking study exhibits that among the synthesized compounds, 4(c-e) have great binding ability toward B-DNA. The binding efficiencies of compounds 4(c-e) with CT-DNA were evaluated via UV-visible absorption spectral and viscosity studies. The findings establish that the compounds firmly bind through an intercalative mode to CT-DNA and provide a unique pattern of DNA binding. The photo-induced cleavage indicates that the compounds have UV-visible photo nuclease properties toward plasmid DNA as revealed by agarose gel electrophoresis approach.
Collapse
Affiliation(s)
- Radhika Bhat
- Department of Studies in Chemistry, Bangalore University, Bangalore, Karnataka, India
| | - Noor Shahina Begum
- Department of Studies in Chemistry, Bangalore University, Bangalore, Karnataka, India
| |
Collapse
|
67
|
Serafim MSM, Dos Santos Júnior VS, Gertrudes JC, Maltarollo VG, Honorio KM. Machine learning techniques applied to the drug design and discovery of new antivirals: a brief look over the past decade. Expert Opin Drug Discov 2021; 16:961-975. [PMID: 33957833 DOI: 10.1080/17460441.2021.1918098] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Drug design and discovery of new antivirals will always be extremely important in medicinal chemistry, taking into account known and new viral diseases that are yet to come. Although machine learning (ML) have shown to improve predictions on the biological potential of chemicals and accelerate the discovery of drugs over the past decade, new methods and their combinations have improved their performance and established promising perspectives regarding ML in the search for new antivirals.Areas covered: The authors consider some interesting areas that deal with different ML techniques applied to antivirals. Recent innovative studies on ML and antivirals were selected and analyzed in detail. Also, the authors provide a brief look at the past to the present to detect advances and bottlenecks in the area.Expert opinion: From classical ML techniques, it was possible to boost the searches for antivirals. However, from the emergence of new algorithms and the improvement in old approaches, promising results will be achieved every day, as we have observed in the case of SARS-CoV-2. Recent experience has shown that it is possible to use ML to discover new antiviral candidates from virtual screening and drug repurposing.
Collapse
Affiliation(s)
- Mateus Sá Magalhães Serafim
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Jadson Castro Gertrudes
- Departamento de Computação, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Kathia Maria Honorio
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (USP), São Paulo, Brazil.,Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Santo André, Brazil
| |
Collapse
|
68
|
Tarasova O, Poroikov V. Machine Learning in Discovery of New Antivirals and Optimization of Viral Infections Therapy. Curr Med Chem 2021; 28:7840-7861. [PMID: 33949929 DOI: 10.2174/0929867328666210504114351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 11/22/2022]
Abstract
Nowadays, computational approaches play an important role in the design of new drug-like compounds and optimization of pharmacotherapeutic treatment of diseases. The emerging growth of viral infections, including those caused by the Human Immunodeficiency Virus (HIV), Ebola virus, recently detected coronavirus, and some others, leads to many newly infected people with a high risk of death or severe complications. A huge amount of chemical, biological, clinical data is at the disposal of the researchers. Therefore, there are many opportunities to find the relationships between the particular features of chemical data and the antiviral activity of biologically active compounds based on machine learning approaches. Biological and clinical data can also be used for building models to predict relationships between viral genotype and drug resistance, which might help determine the clinical outcome of treatment. In the current study, we consider machine-learning approaches in the antiviral research carried out during the past decade. We overview in detail the application of machine-learning methods for the design of new potential antiviral agents and vaccines, drug resistance prediction, and analysis of virus-host interactions. Our review also covers the perspectives of using the machine-learning approaches for antiviral research, including Dengue, Ebola viruses, Influenza A, Human Immunodeficiency Virus, coronaviruses, and some others.
Collapse
Affiliation(s)
- Olga Tarasova
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow. Russian Federation
| | - Vladimir Poroikov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow. Russian Federation
| |
Collapse
|
69
|
Leidner F, Yilmaz NK, Schiffer CA. Deciphering Complex Mechanisms of Resistance and Loss of Potency through Coupled Molecular Dynamics and Machine Learning. J Chem Theory Comput 2021; 17:2054-2064. [PMID: 33783217 PMCID: PMC8164521 DOI: 10.1021/acs.jctc.0c01244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Drug resistance threatens many critical therapeutics through mutations in the drug target. The molecular mechanisms by which combinations of mutations, especially those remote from the active site, alter drug binding to confer resistance are poorly understood and thus difficult to counteract. A machine learning strategy was developed that coupled parallel molecular dynamics simulations with experimental potency to identify specific conserved mechanisms underlying resistance. Physical features were extracted from the simulations, analyzed, and integrated into one consistent and interpretable elastic network model. To rigorously test this strategy, HIV-1 protease variants with diverse mutations were used, with potencies ranging from picomolar to micromolar to the drug darunavir. Feature reduction resulted in a model with four specific features that predicts for both the training and test sets inhibitor binding free energy within 1 kcal/mol of the experimental value over this entire range of potency. These predictive features are physically interpretable, as they vary specifically with affinity and diagonally transverse across the protease homodimer. This physics-based strategy of parallel molecular dynamics and machine learning captures mechanisms by which complex combinations of mutations confer resistance and identify critical features that serve as bellwethers of affinity, which will be critical in future drug design.
Collapse
Affiliation(s)
- Florian Leidner
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
70
|
Fu Y, Jaarsma AH, Kuipers OP. Antiviral activities and applications of ribosomally synthesized and post-translationally modified peptides (RiPPs). Cell Mol Life Sci 2021; 78:3921-3940. [PMID: 33532865 PMCID: PMC7853169 DOI: 10.1007/s00018-021-03759-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/15/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022]
Abstract
The emergence and re-emergence of viral epidemics and the risks of antiviral drug resistance are a serious threat to global public health. New options to supplement or replace currently used drugs for antiviral therapy are urgently needed. The research in the field of ribosomally synthesized and post-translationally modified peptides (RiPPs) has been booming in the last few decades, in particular in view of their strong antimicrobial activities and high stability. The RiPPs with antiviral activity, especially those against enveloped viruses, are now also gaining more interest. RiPPs have a number of advantages over small molecule drugs in terms of specificity and affinity for targets, and over protein-based drugs in terms of cellular penetrability, stability and size. Moreover, the great engineering potential of RiPPs provides an efficient way to optimize them as potent antiviral drugs candidates. These intrinsic advantages underscore the good therapeutic prospects of RiPPs in viral treatment. With the aim to highlight the underrated antiviral potential of RiPPs and explore their development as antiviral drugs, we review the current literature describing the antiviral activities and mechanisms of action of RiPPs, discussing the ongoing efforts to improve their antiviral potential and demonstrate their suitability as antiviral therapeutics. We propose that antiviral RiPPs may overcome the limits of peptide-based antiviral therapy, providing an innovative option for the treatment of viral disease.
Collapse
Affiliation(s)
- Yuxin Fu
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Ate H Jaarsma
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands
- Department of Environmental Science, Aarhus University, 4000, Roskilde, Denmark
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
71
|
Tummanapalli SS, Willcox MD. Antimicrobial resistance of ocular microbes and the role of antimicrobial peptides. Clin Exp Optom 2021; 104:295-307. [PMID: 32924208 DOI: 10.1111/cxo.13125] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Isolation of antimicrobial-resistant microbes from ocular infections may be becoming more frequent. Infections caused by these microbes can be difficult to treat and lead to poor outcomes. However, new therapies are being developed which may help improve clinical outcomes. This review examines recent reports on the isolation of antibiotic-resistant microbes from ocular infections. In addition, an overview of the development of some new antibiotic therapies is given. The recent literature regarding antibiotic use and resistance, isolation of antibiotic-resistant microbes from ocular infections and the development of potential new antibiotics that can be used to treat these infections was reviewed. Ocular microbial infections are a global public health issue as they can result in vision loss which compromises quality of life. Approximately 70 per cent of ocular infections are caused by bacteria including Chlamydia trachomatis, Staphylococcus aureus, and Pseudomonas aeruginosa and fungi such as Candida albicans, Aspergillus spp. and Fusarium spp. Resistance to first-line antibiotics such as fluoroquinolones and azoles has increased, with resistance of S. aureus isolates from the USA to fluoroquinolones reaching 32 per cent of isolates and 35 per cent being methicillin-resistant (MRSA). Lower levels of MRSA (seven per cent) were isolated by an Australian study. Antimicrobial peptides, which are broad-spectrum alternatives to antibiotics, have been tested as possible new drugs. Several have shown promise in animal models of keratitis, especially treating P. aeruginosa, S. aureus or C. albicans infections. Reports of increasing resistance of ocular isolates to mainstay antibiotics are a concern, and there is evidence that for ocular surface disease this resistance translates into worse clinical outcomes. New antibiotics are being developed, but not by large pharmaceutical companies and mostly in university research laboratories and smaller biotech companies. Antimicrobial peptides show promise in treating keratitis.
Collapse
Affiliation(s)
| | - Mark Dp Willcox
- School of Optometry and Vision Science, The University of New South Wales, Sydney, Australia
| |
Collapse
|
72
|
Khan F, Bamunuarachchi NI, Tabassum N, Kim YM. Caffeic Acid and Its Derivatives: Antimicrobial Drugs toward Microbial Pathogens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:2979-3004. [PMID: 33656341 DOI: 10.1021/acs.jafc.0c07579] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Caffeic acid is a plant-derived compound that is classified as hydroxycinnamic acid which contains both phenolic and acrylic functional groups. Caffeic acid has been greatly employed as an alternative strategy to combat microbial pathogenesis and chronic infection induced by microbes such as bacteria, fungi, and viruses. Similarly, several derivatives of caffeic acid such as sugar esters, organic esters, glycosides, and amides have been chemically synthesized or naturally isolated as potential antimicrobial agents. To overcome the issue of water insolubility and poor stability, caffeic acid and its derivative have been utilized either in conjugation with other bioactive molecules or in nanoformulation. Besides, caffeic acid and its derivatives have also been applied in combination with antibiotics or photoirradiation to achieve a synergistic mode of action. The present review describes the antimicrobial roles of caffeic acid and its derivatives exploited either in free form or in combination or in nanoformulation to kill a diverse range of microbial pathogens along with their mode of action. The chemistry employed for the synthesis of the caffeic acid derivatives has been discussed in detail as well.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Institute of Food Science, Pukyong National University, Busan 48513, South Korea
| | - Nilushi Indika Bamunuarachchi
- Department of Food Science and Technology, Pukyong National University, Busan 48513, South Korea
- Department of Fisheries and Marine Sciences, Ocean University of Sri Lanka, Tangalle 82200, Sri Lanka
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan 48513, South Korea
| | - Young-Mog Kim
- Institute of Food Science, Pukyong National University, Busan 48513, South Korea
- Department of Food Science and Technology, Pukyong National University, Busan 48513, South Korea
| |
Collapse
|
73
|
Pouyan P, Nie C, Bhatia S, Wedepohl S, Achazi K, Osterrieder N, Haag R. Inhibition of Herpes Simplex Virus Type 1 Attachment and Infection by Sulfated Polyglycerols with Different Architectures. Biomacromolecules 2021; 22:1545-1554. [PMID: 33706509 DOI: 10.1021/acs.biomac.0c01789] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibition of herpes simplex virus type 1 (HSV-1) binding to the host cell surface by highly sulfated architectures is among the promising strategies to prevent virus entry and infection. However, the structural flexibility of multivalent inhibitors plays a major role in effective blockage and inhibition of virus receptors. In this study, we demonstrate the inhibitory effect of a polymer scaffold on the HSV-1 infection by using highly sulfated polyglycerols with different architectures (linear, dendronized, and hyperbranched). IC50 values for all synthesized sulfated polyglycerols and the natural sulfated polymer heparin were determined using plaque reduction infection assays. Interestingly, an increase in the IC50 value from 0.03 to 374 nM from highly flexible linear polyglycerol sulfate (LPGS) to less flexible scaffolds, namely, dendronized polyglycerol sulfate and hyperbranched polyglycerol sulfate was observed. The most potent LPGS inhibits HSV-1 infection 295 times more efficiently than heparin, and we show that LPGS has a much reduced anticoagulant capacity when compared to heparin as evidenced by measuring the activated partial thromboplastin time. Furthermore, prevention of infection by LPGS and the commercially available drug acyclovir were compared. All tested sulfated polymers do not show any cytotoxicity at concentrations of up to 1 mg/mL in different cell lines. We conclude from our results that more flexible polyglycerol sulfates are superior to less flexible sulfated polymers with respect to inhibition of HSV-1 infection and may constitute an alternative to the current antiviral treatments of this ubiquitous pathogen.
Collapse
Affiliation(s)
- Paria Pouyan
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Chuanxiong Nie
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany.,Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Street 7-13, Berlin 14163, Germany
| | - Sumati Bhatia
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Stefanie Wedepohl
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee. 22, Berlin 14195, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee. 22, Berlin 14195, Germany
| | - Nikolaus Osterrieder
- Institut für Virologie, Robert von Ostertag-Haus, Zentrum für Infektionsmedizin, Freie Universität Berlin, Robert-von-Ostertag-Street 7-13, Berlin 14163, Germany.,Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon Tong TYB-1B-507, Hong Kong
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| |
Collapse
|
74
|
Looking for Solutions to the Pitfalls of Developing Novel Antibacterials in an Economically Challenging System. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12010013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The increase in antibacterial resistance (ABR) currently equates in the minds of many with the distant fear that certain antibiotics will not work in 30 years on certain bacteria found in places the majority of us never go to. However, in reality, rising ABR already seriously threatens the effectiveness of compounds with which we treat common bacterial infections, which means that ABR is currently and will continue to undermine the foundations of modern medicine, including surgery and cancer treatment in hospitals, cities and countries across the world. That is why ABR is widely considered a global threat and one of the biggest problems of our current civilization. Conversely, antibiotic developments to market are few. Therefore, in this paper, we have illustrated the barriers to antimicrobial R&D the following questions and provided solutions to effective antimicrobial R&D.
Collapse
|
75
|
Besednova NN, Andryukov BG, Zaporozhets TS, Kryzhanovsky SP, Fedyanina LN, Kuznetsova TA, Zvyagintseva TN, Shchelkanov MY. Antiviral Effects of Polyphenols from Marine Algae. Biomedicines 2021; 9:200. [PMID: 33671278 PMCID: PMC7921925 DOI: 10.3390/biomedicines9020200] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023] Open
Abstract
The disease-preventive and medicinal properties of plant polyphenolic compounds have long been known. As active ingredients, they are used to prevent and treat many noncommunicable diseases. In recent decades, marine macroalgae have attracted the attention of biotechnologists and pharmacologists as a promising and almost inexhaustible source of polyphenols. This heterogeneous group of compounds contains many biopolymers with unique structure and biological properties that exhibit high anti-infective activity. In the present review, the authors focus on the antiviral potential of polyphenolic compounds (phlorotannins) from marine algae and consider the mechanisms of their action as well as other biological properties of these compounds that have effects on the progress and outcome of viral infections. Effective nutraceuticals, to be potentially developed on the basis of algal polyphenols, can also be used in the complex therapy of viral diseases. It is necessary to extend in vivo studies on laboratory animals, which subsequently will allow proceeding to clinical tests. Polyphenolic compounds have a great potential as active ingredients to be used for the creation of new antiviral pharmaceutical substances.
Collapse
Affiliation(s)
- Natalya N. Besednova
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
| | - Boris G. Andryukov
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia;
| | - Tatyana S. Zaporozhets
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
| | - Sergey P. Kryzhanovsky
- Medical Association of the Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia;
| | - Ludmila N. Fedyanina
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia;
| | - Tatyana A. Kuznetsova
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
| | | | - Mikhail Yu. Shchelkanov
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia;
- Federal Scientific Center of the Eastern Asia Terrestrial Biodiversity, Far Eastern Branch of Russian Academy of Sciences, 690091 Vladivostok, Russia
- National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, 690091 Vladivostok, Russia
| |
Collapse
|
76
|
Hanafin PO, Jermain B, Hickey AJ, Kabanov AV, Kashuba ADM, Sheahan TP, Rao GG. A mechanism-based pharmacokinetic model of remdesivir leveraging interspecies scaling to simulate COVID-19 treatment in humans. CPT Pharmacometrics Syst Pharmacol 2021; 10:89-99. [PMID: 33296558 PMCID: PMC7894405 DOI: 10.1002/psp4.12584] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak initiated the global coronavirus disease 2019 (COVID-19) pandemic resulting in 42.9 million confirmed infections and > 1.1 million deaths worldwide as of October 26, 2020. Remdesivir is a broad-spectrum nucleotide prodrug shown to be effective against enzootic coronaviruses. The pharmacokinetics (PKs) of remdesivir in plasma have recently been described. However, the distribution of its active metabolite nucleoside triphosphate (NTP) to the site of pulmonary infection is unknown in humans. Our objective was to use existing in vivo mouse PK data for remdesivir and its metabolites to develop a mechanism-based model to allometrically scale and simulate the human PK of remdesivir in plasma and NTP in lung homogenate. Remdesivir and GS-441524 concentrations in plasma and total phosphorylated nucleoside concentrations in lung homogenate from Ces1c-/- mice administered 25 or 50 mg/kg of remdesivir subcutaneously were simultaneously fit to estimate PK parameters. The mouse PK model was allometrically scaled to predict human PK parameters to simulate the clinically recommended 200 mg loading dose followed by 100 mg daily maintenance doses administered as 30-minute intravenous infusions. Simulations of unbound remdesivir concentrations in human plasma were below 2.48 μM, the 90% maximal inhibitory concentration for SARS-CoV-2 inhibition in vitro. Simulations of NTP in the lungs were below high efficacy in vitro thresholds. We have identified a need for alternative dosing strategies to achieve more efficacious concentrations of NTP in human lungs, perhaps by reformulating remdesivir for direct pulmonary delivery.
Collapse
Affiliation(s)
- Patrick O. Hanafin
- Division of Pharmacotherapy and Experimental TherapeuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Brian Jermain
- Division of Pharmacotherapy and Experimental TherapeuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Anthony J. Hickey
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNCUSA
- RTI InternationalResearch Triangle ParkNCUSA
| | - Alexander V. Kabanov
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Angela DM. Kashuba
- Division of Pharmacotherapy and Experimental TherapeuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Timothy P. Sheahan
- Department of EpidemiologyGillings School of Global Public HealthUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Gauri G. Rao
- Division of Pharmacotherapy and Experimental TherapeuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| |
Collapse
|
77
|
Pradhan D, Biswasroy P, Goyal A, Ghosh G, Rath G. Recent Advancement in Nanotechnology-Based Drug Delivery System Against Viral Infections. AAPS PharmSciTech 2021; 22:47. [PMID: 33447909 PMCID: PMC7808403 DOI: 10.1208/s12249-020-01908-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
In the last few decades, the exponential rise in the incidence of viral infections sets a global health emergency across the world. The biomimetic architecture, the ability to hijack host immune responses, continuous antigen shifting, and drafting are the major critical factors that are responsible for the unavailability of a concrete therapeutic regimen against viral infections. Further, inappropriate pharmacodynamic physicochemical and biological parameters such as low aqueous solubility, poor permeability, high affinity for plasm proteins, short biological half-lives, and fast elimination from the systemic circulation are the major critical factors that govern the suboptimal drug concentration at the target site that leads to the development of drug resistance. To address this issue, nanotechnology-based drug delivery approach is emerged as an altering method to attain the optimal drug concentration at the target site for a prolonged period by integrating the nanoengineering tools in the synthesis of nanoparticles. Nanodimensional configuration with enhanced permeability and retention effect, increased surface-area-to-volume ratio, provision for surface functionalization, etc., are the privileged aspects that make it an effective drug delivery system for dispensing the antiviral therapeutics. However, size, shape, charge, and surface topology of nanoparticles are the greater influential factors that determine target-specific drug delivery, optimum cellular uptake, degree of opsonization by the host immune cells, drug retention time, transcytosis, the extension of biological half-life, in vivo stability, and cytotoxicity. The review will enlighten the elaborative role of nanotechnology-based drug delivery and the major challenging aspect of clinical safety and efficacy.
Collapse
|
78
|
Saravanan M, Belete MA, Niguse S, Tsegay E, Araya T, Hadush B, Nigussie K, Prakash P. Antimicrobial Resistance and Antimicrobial Nanomaterials. ADVANCES IN MEDICAL TECHNOLOGIES AND CLINICAL PRACTICE 2021:1-28. [DOI: http:/doi:10.4018/978-1-7998-5049-6.ch001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2023]
Abstract
Back in the mid-nineties, the discovery of antimicrobials denoted a profound and remarkable achievement in medicine which was capable of saving lives. However, recently, antimicrobial resistance became a major global issue facing modern medicine and significantly increased among bacteria, fungi, and viruses which results in reduced efficacy of many clinically important and lifesaving antimicrobials. The growing rise of antimicrobial resistance inflicts a remarkable economic and social burden on the health care system globally. The replacement of conventional antimicrobials by new technology to counteract and lessen antimicrobial resistance is currently ongoing. Nanotechnology is an advanced approach to overcome challenges of such resisted conventional drug delivery systems mainly based on the development and fabrication of nanoparticulate structures. Numerous forms of nanoparticulate systems have been discovered and tried as prospective drug delivery systems, comprising organic and inorganic nanoparticles.
Collapse
Affiliation(s)
- Muthupandian Saravanan
- Mekelle University, Ethiopia & Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), India
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Saravanan M, Belete MA, Niguse S, Tsegay E, Araya T, Hadush B, Nigussie K, Prakash P. Antimicrobial Resistance and Antimicrobial Nanomaterials. HANDBOOK OF RESEARCH ON NANO-STRATEGIES FOR COMBATTING ANTIMICROBIAL RESISTANCE AND CANCER 2021. [DOI: 10.4018/978-1-7998-5049-6.ch001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Back in the mid-nineties, the discovery of antimicrobials denoted a profound and remarkable achievement in medicine which was capable of saving lives. However, recently, antimicrobial resistance became a major global issue facing modern medicine and significantly increased among bacteria, fungi, and viruses which results in reduced efficacy of many clinically important and lifesaving antimicrobials. The growing rise of antimicrobial resistance inflicts a remarkable economic and social burden on the health care system globally. The replacement of conventional antimicrobials by new technology to counteract and lessen antimicrobial resistance is currently ongoing. Nanotechnology is an advanced approach to overcome challenges of such resisted conventional drug delivery systems mainly based on the development and fabrication of nanoparticulate structures. Numerous forms of nanoparticulate systems have been discovered and tried as prospective drug delivery systems, comprising organic and inorganic nanoparticles.
Collapse
Affiliation(s)
- Muthupandian Saravanan
- Mekelle University, Ethiopia & Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), India
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Viegas DJ, da Silva VD, Buarque CD, Bloom DC, Abreu PA. Antiviral activity of 1,4-disubstituted-1,2,3-triazoles against HSV-1 in vitro. Antivir Ther 2021; 25:399-410. [PMID: 33705354 DOI: 10.3851/imp3387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2020] [Indexed: 10/21/2022]
Abstract
BACKGROUND Herpes simplex virus 1 (HSV-1) affects a large part of the adult population. Anti-HSV-1 drugs, such as acyclovir, target thymidine kinase and viral DNA polymerase. However, the emerging of resistance of HSV-1 alerts for the urgency in developing new antivirals with other therapeutic targets. Thus, this study evaluated a series of 1,4-disubstituted-1,2,3-triazole derivatives against HSV-1 acute infection and provided deeper insights into the possible mechanisms of action. METHODS Human fibroblast cells (HFL-1) were infected with HSV-1 17syn+ and treated with the triazole compounds at 50 μM for 24 h. The 50% effective drug concentration (EC50) was determined for the active compounds. Their cytotoxicity was also evaluated in HFL-1 with the 50% cytotoxic concentration (CC50) determined using CellTiter-Glo® solution. The most promising compounds were evaluated by virucidal activity and influence on virus egress, DNA replication and transcription, and effect on an acyclovir-resistant HSV-1 strain. RESULTS Compounds 3 ((E)-4-methyl-N'-(2-(4-(phenoxymethyl)-1H-1,2,3-triazol1yl)benzylidene)benzenesulfonohydrazide) and 4 (2,2'-(4,4'-((1,3-phenylenebis(oxy))bis(methylene))bis(1H-1,2,3-triazole-4,1 diyl)) dibenzaldehyde) were the most promising, with an EC50 of 16 and 21 μM and CC50 of 285 and 2,593 μM, respectively. Compound 3 was able to inhibit acyclovir-resistant strain replication and to interfere with virus egress. Both compounds did not affect viral DNA replication, but inhibited significantly the expression of ICP0, ICP4 and gC. Compound 4 also affected the transcription of UL30 and ICP34.5. CONCLUSIONS Our findings demonstrated that these compounds are promising antiviral candidates with different mechanisms of action from acyclovir and further studies are merited.
Collapse
Affiliation(s)
- Daiane J Viegas
- LAMCIFAR, Laboratório de Modelagem Molecular e Pesquisa em Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
| | - Verônica D da Silva
- Laboratório de Síntese Orgânica, Pontifícia Universidade Católica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camilla D Buarque
- Laboratório de Síntese Orgânica, Pontifícia Universidade Católica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - David C Bloom
- College of Medicine, Department of Microbiology and Molecular Genetics, University of Florida, Gainesville, FL, USA
| | - Paula A Abreu
- LAMCIFAR, Laboratório de Modelagem Molecular e Pesquisa em Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
| |
Collapse
|
81
|
Tunstall T, Portelli S, Phelan J, Clark TG, Ascher DB, Furnham N. Combining structure and genomics to understand antimicrobial resistance. Comput Struct Biotechnol J 2020; 18:3377-3394. [PMID: 33294134 PMCID: PMC7683289 DOI: 10.1016/j.csbj.2020.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023] Open
Abstract
Antimicrobials against bacterial, viral and parasitic pathogens have transformed human and animal health. Nevertheless, their widespread use (and misuse) has led to the emergence of antimicrobial resistance (AMR) which poses a potentially catastrophic threat to public health and animal husbandry. There are several routes, both intrinsic and acquired, by which AMR can develop. One major route is through non-synonymous single nucleotide polymorphisms (nsSNPs) in coding regions. Large scale genomic studies using high-throughput sequencing data have provided powerful new ways to rapidly detect and respond to such genetic mutations linked to AMR. However, these studies are limited in their mechanistic insight. Computational tools can rapidly and inexpensively evaluate the effect of mutations on protein function and evolution. Subsequent insights can then inform experimental studies, and direct existing or new computational methods. Here we review a range of sequence and structure-based computational tools, focussing on tools successfully used to investigate mutational effect on drug targets in clinically important pathogens, particularly Mycobacterium tuberculosis. Combining genomic results with the biophysical effects of mutations can help reveal the molecular basis and consequences of resistance development. Furthermore, we summarise how the application of such a mechanistic understanding of drug resistance can be applied to limit the impact of AMR.
Collapse
Affiliation(s)
- Tanushree Tunstall
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Stephanie Portelli
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Australia
- Structural Biology and Bioinformatics, Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Australia
| | - Jody Phelan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Taane G. Clark
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - David B. Ascher
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Australia
- Structural Biology and Bioinformatics, Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Australia
| | - Nicholas Furnham
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
82
|
Zitterl-Eglseer K, Marschik T. Antiviral Medicinal Plants of Veterinary Importance: A Literature Review. PLANTA MEDICA 2020; 86:1058-1072. [PMID: 32777833 DOI: 10.1055/a-1224-6115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Viruses have a high mutation rate, and, thus, there is a continual emergence of new antiviral-resistant strains. Therefore, it becomes imperative to explore and develop new antiviral compounds continually. The search for pharmacological substances of plant origin that are effective against animal viruses, which have a high mortality rate or cause large economic losses, has garnered interest in the last few decades. This systematic review compiles 130 plant species that exhibit antiviral activity on 37 different virus species causing serious diseases in animals. The kind of extract, fraction, or compound exhibiting the antiviral activity and the design of the trial were particularly considered for review. The literature revealed details regarding plant species exhibiting antiviral activities against pathogenic animal virus species of the following families-Herpesviridae, Orthomyxoviridae, Paramyxoviridae, Parvoviridae, Poxviridae, Nimaviridae, Coronaviridae, Reoviridae, and Rhabdoviridae-that cause infections, among others, in poultry, cattle, pigs, horses, shrimps, and fish. Overall, 30 plant species exhibited activity against various influenza viruses, most of them causing avian influenza. Furthermore, 30 plant species were noted to be active against Newcastle disease virus. In addition, regarding the pathogens most frequently investigated, this review provides a compilation of 20 plant species active against bovine herpesvirus, 16 against fowlpox virus, 12 against white spot syndrome virus in marine shrimps, and 10 against suide herpesvirus. Nevertheless, some plant extracts, particularly their compounds, are promising candidates for the development of new antiviral remedies, which are urgently required.
Collapse
Affiliation(s)
- Karin Zitterl-Eglseer
- Institute of Animal Nutrition and Functional Plant Compounds, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Tatiana Marschik
- Unit of Veterinary Public Health and Epidemiology, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
83
|
Campos EVR, Pereira AES, de Oliveira JL, Carvalho LB, Guilger-Casagrande M, de Lima R, Fraceto LF. How can nanotechnology help to combat COVID-19? Opportunities and urgent need. J Nanobiotechnology 2020; 18:125. [PMID: 32891146 PMCID: PMC7474329 DOI: 10.1186/s12951-020-00685-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
Incidents of viral outbreaks have increased at an alarming rate over the past decades. The most recent human coronavirus known as COVID-19 (SARS-CoV-2) has already spread around the world and shown R0 values from 2.2 to 2.68. However, the ratio between mortality and number of infections seems to be lower in this case in comparison to other human coronaviruses (such as severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV)). These outbreaks have tested the limits of healthcare systems and have posed serious questions about management using conventional therapies and diagnostic tools. In this regard, the use of nanotechnology offers new opportunities for the development of novel strategies in terms of prevention, diagnosis and treatment of COVID-19 and other viral infections. In this review, we discuss the use of nanotechnology for COVID-19 virus management by the development of nano-based materials, such as disinfectants, personal protective equipment, diagnostic systems and nanocarrier systems, for treatments and vaccine development, as well as the challenges and drawbacks that need addressing.
Collapse
Affiliation(s)
- Estefânia V R Campos
- Human and Natural Sciences Center, Federal University of ABC. Av. dos Estados, 5001. Bl. A, T3 Lab. 503-3. Bangú, Santo André, SP, Brazil
| | - Anderson E S Pereira
- São Paulo State University-UNESP, Institute of Science and Technology, Sorocaba, SP, Brazil
| | | | | | | | - Renata de Lima
- Universidade de Sorocaba, Rodovia Raposo Tavares km 92,5, Sorocaba, São Paulo, Brazil.
| | | |
Collapse
|
84
|
Trompet E, Topalis D, Gillemot S, Snoeck R, Andrei G. Viral fitness of MHV-68 viruses harboring drug resistance mutations in the protein kinase or thymidine kinase. Antiviral Res 2020; 182:104901. [PMID: 32763314 DOI: 10.1016/j.antiviral.2020.104901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022]
Abstract
Murine γ-herpesvirus-68 (MHV-68), genetically and biologically related to human γ-herpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, can be easily propagated in vitro allowing drug resistance studies. Previously, we described specific changes in MHV-68 protein kinase (PK) or thymidine kinase (TK) associated with resistance to various purine or pyrimidine nucleoside analogues, respectively. To investigate how specific TK and PK mutations affect viral replication capacity, we performed dual infection competition assays in which wild-type and drug-resistant virus compete in absence or presence of antivirals in Vero cells. The composition of the mixed viral population was analyzed using next-generation sequencing and relative fitness of seven MHV-68 PK or TK mutants was calculated based on the frequency of viral variants at the time of infection and after 5-days growth. A MHV-68 mutant losing the PK function due to a 2-nucleotide deletion was less fit than the wild-type virus in absence of antivirals, consistent with the essential role of viral PKs during lytic replication, but overgrew the wild-type virus under pressure of purine nucleosides. TK mutant viruses, with frameshift or missense mutations, grew equal to wild-type virus in absence of antivirals, in accordance with the viral TK function only being essential in non-replicating or in TK-deficient cells, but were more fit when treated with pyrimidine nucleosides. Moreover, TK missense mutant viruses also increased fitness under pressure of antivirals other than pyrimidine nucleosides, indicating that MHV-68 TK mutations might influence viral fitness by acting on cellular and/or viral functions that are unrelated to nucleoside activation.
Collapse
Affiliation(s)
- Erika Trompet
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Sarah Gillemot
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Robert Snoeck
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Graciela Andrei
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
85
|
Makino T, Ishida J, Yamanaka T, Ohki H, Uchida M, Sawada M, Barrett D. Discovery of a novel 9-position modified second-generation anti-HCV candidate via bioconversion and semi-synthesis of FR901459. Bioorg Med Chem Lett 2020; 30:127423. [PMID: 32731088 DOI: 10.1016/j.bmcl.2020.127423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 11/28/2022]
Abstract
Evidence that hepatitis C virus (HCV) utilizes cellular cyclophilin proteins in the virus replication cycle has increased attention on cyclophilin inhibitors as attractive therapeutic targets in the treatment of HCV. Previous reports have described a number of non-immunosuppressive cyclophilin inhibitors, most of which require many synthetic steps for their preparation. Sasamura et al. have previously reported the isolation of bioconversion derivative 4. This analog is a convenient starting point for optimization due to the presence of the readily modifiable primary hydroxyl group and because it shows moderate anti-HCV activity and decreased immunosuppressive activity. We have also established an efficient C-alkylation reaction at the 3-position. Through a detailed structure-activity relationship study, we discovered a new type of clinical candidate 14 which requires a short synthetic process and has potent anti-HCV activity and reduced immunosuppressive activity, as well as improved aqueous solubility and pharmacokinetics.
Collapse
Affiliation(s)
- Takuya Makino
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan.
| | - Junya Ishida
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Toshio Yamanaka
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Hidenori Ohki
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Masao Uchida
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Masae Sawada
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - David Barrett
- Drug Discovery Research, Astellas Pharma Inc.; 21, Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| |
Collapse
|
86
|
Papanicolaou GA, Silveira FP, Langston AA, Pereira MR, Avery RK, Uknis M, Wijatyk A, Wu J, Boeckh M, Marty FM, Villano S. Maribavir for Refractory or Resistant Cytomegalovirus Infections in Hematopoietic-cell or Solid-organ Transplant Recipients: A Randomized, Dose-ranging, Double-blind, Phase 2 Study. Clin Infect Dis 2020; 68:1255-1264. [PMID: 30329038 PMCID: PMC6451997 DOI: 10.1093/cid/ciy706] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/03/2018] [Indexed: 12/27/2022] Open
Abstract
Background Cytomegalovirus (CMV) infections that are refractory or resistant (RR) to available antivirals ([val]ganciclovir, foscarnet, cidofovir) are associated with higher mortality in transplant patients. Maribavir is active against RR CMV strains. Methods Hematopoietic-cell or solid-organ transplant recipients ≥12 years old with RR CMV infections and plasma CMV deoxyribonucleic acid (DNA) ≥1000 copies/mL were randomized (1:1:1) to twice-daily dose-blinded maribavir 400, 800, or 1200 mg for up to 24 weeks. The primary efficacy endpoint was the proportion of patients with confirmed undetectable plasma CMV DNA within 6 weeks of treatment. Safety analyses included the frequency and severity of treatment-emergent adverse events (TEAEs). Results From July 2012 to December 2014, 120 patients were randomized and treated (40 per dose group): 80/120 (67%) patients achieved undetectable CMV DNA within 6 weeks of treatment (95% confidence interval, 57–75%), with rates of 70%, 63%, and 68%, respectively, for maribavir 400, 800, and 1200 mg twice daily. Recurrent on-treatment CMV infections occurred in 25 patients; 13 developed mutations conferring maribavir resistance. Maribavir was discontinued due to adverse events in 41/120 (34%) patients, and 17/41 discontinued due to CMV infections. During the study, 32 (27%) patients died, 4 due to CMV disease. Dysgeusia was the most common TEAE (78/120; 65%) and led to maribavir discontinuation in 1 patient. Absolute neutrophil counts <1000/µL were noted in 12/106 (11%) evaluable patients, with rates similar across doses. Conclusions Maribavir ≥400 mg twice daily was active against RR CMV infections in transplant recipients; no new safety signals were identified. Clinical Trials Registration NCT01611974.
Collapse
Affiliation(s)
| | - Fernanda P Silveira
- The Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Marcus R Pereira
- Department of Medicine, Columbia University Medical Center, New York, New York
| | | | - Marc Uknis
- Shire Pharmaceuticals, Wayne, Pennsylvania
| | | | - Jingyang Wu
- Shire Pharmaceuticals, Lexington, Massachusetts
| | - Michael Boeckh
- The Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | |
Collapse
|
87
|
Luo W, Zhang JW, Zhang W, Lin YL, Wang Q. Circulating levels of IL-2, IL-4, TNF-α, IFN-γ, and C-reactive protein are not associated with severity of COVID-19 symptoms. J Med Virol 2020; 93:89-91. [PMID: 32519779 PMCID: PMC7300996 DOI: 10.1002/jmv.26156] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/07/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Wen Luo
- Department of Respiratory and Critical Care, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jia-Wen Zhang
- Department of Respiratory and Critical Care, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Zhang
- Department of Respiratory and Critical Care, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuan-Long Lin
- Department of Infection, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qi Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| |
Collapse
|
88
|
Yu J, Kimble B, Norris JM, Govendir M. Pharmacokinetic Profile of Oral Administration of Mefloquine to Clinically Normal Cats: A Preliminary In-Vivo Study of a Potential Treatment for Feline Infectious Peritonitis (FIP). Animals (Basel) 2020; 10:ani10061000. [PMID: 32521771 PMCID: PMC7341284 DOI: 10.3390/ani10061000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary In searching for antiviral agents against feline coronaviruses and feline caliciviruses, mefloquine, a human anti-malarial drug, has been demonstrated to reduce viral load of feline coronaviruses and feline calicivirus in infected cells. In this study, mefloquine was administered orally to seven clinically healthy cats twice weekly for four doses and mefloquine concentrations in blood were measured to investigate the pharmacokinetic profile—the movement of drug in the body. The maximum blood concentration of mefloquine was 2.71 ug/mL and was reached 15 h after a single oral dose was administered. Mefloquine side effects included vomiting following administration without food in some cats, and mild increases in symmetric dimethylarginine (SDMA), an early kidney biomarker. This study provides valuable information on mefloquine’s profile in cats as an introductory step towards investigating it as a potential treatment for feline coronavirus and feline calicivirus infection in cats. Abstract The pharmacokinetic profile of mefloquine was investigated as a preliminary study towards a potential treatment for feline coronavirus infections (such as feline infectious peritonitis) or feline calicivirus infections. Mefloquine was administered at 62.5 mg orally to seven clinically healthy cats twice weekly for four doses and mefloquine plasma concentrations over 336 h were measured using high pressure liquid chromatography (HPLC). The peak plasma concentration (Cmax) after a single oral dose of mefloquine was 2.71 ug/mL and time to reach Cmax (Tmax) was 15 h. The elimination half-life was 224 h. The plasma concentration reached a higher level at 4.06 ug/mL when mefloquine was administered with food. Adverse effects of dosing included vomiting following administration without food in some cats. Mild increases in serum symmetric dimethylarginine (SDMA), but not creatinine, concentrations were observed. Mefloquine may provide a safe effective treatment for feline coronavirus and feline calicivirus infections in cats.
Collapse
|
89
|
Bogožalec Košir A, Cvelbar T, Kammel M, Grunert HP, Zeichhardt H, Milavec M. Digital PCR method for detection and quantification of specific antimicrobial drug-resistance mutations in human cytomegalovirus. J Virol Methods 2020; 281:113864. [PMID: 32380093 DOI: 10.1016/j.jviromet.2020.113864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
Antimicrobial drug resistance is one of the biggest threats to human health worldwide. Timely detection and quantification of infectious agents and their susceptibility to antimicrobial drugs are crucial for efficient management of resistance to antiviral drugs. In clinical settings, viral drug resistance is most often associated with prolonged treatment of chronic infections, and assessed by genotyping methods; e.g., sequencing and PCR. These approaches have limitations: sequencing can be expensive and does not provide quantification; and qPCR quantification is hampered by a lack of reference materials for standard curves. In recent years, digital PCR has been introduced, which provides absolute quantification without the need for reference materials for standard curves. Using digital PCR, we have developed a rapid, sensitive and accurate method for genotyping and quantification of the most prevalent mutations that cause human cytomegalovirus resistance to ganciclovir.
Collapse
Affiliation(s)
- Alexandra Bogožalec Košir
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| | - Tašja Cvelbar
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Martin Kammel
- INSTAND, Gesellschaft Zur Förderung Der Qualitätssicherung in Medizinischen Laboratorien e.V. Ubierstr.20, 40223 Düsseldorf, Germany; IQVD GmbH, Institut Für Qualitätssicherung in Der Virusdiagnostik, Potsdamer Chaussee 80, 14129 Berlin, Germany
| | - Hans-Peter Grunert
- GBD Gesellschaft Für Biotechnologische Diagnostik mbH, Potsdamer Chaussee 80, 14129 Berlin, Germany
| | - Heinz Zeichhardt
- INSTAND, Gesellschaft Zur Förderung Der Qualitätssicherung in Medizinischen Laboratorien e.V. Ubierstr.20, 40223 Düsseldorf, Germany; IQVD GmbH, Institut Für Qualitätssicherung in Der Virusdiagnostik, Potsdamer Chaussee 80, 14129 Berlin, Germany; GBD Gesellschaft Für Biotechnologische Diagnostik mbH, Potsdamer Chaussee 80, 14129 Berlin, Germany
| | - Mojca Milavec
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia.
| |
Collapse
|
90
|
Alves NG, Mata AI, Luís JP, Brito RMM, Simões CJV. An Innovative Sequence-to-Structure-Based Approach to Drug Resistance Interpretation and Prediction: The Use of Molecular Interaction Fields to Detect HIV-1 Protease Binding-Site Dissimilarities. Front Chem 2020; 8:243. [PMID: 32411655 PMCID: PMC7202381 DOI: 10.3389/fchem.2020.00243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/13/2020] [Indexed: 12/15/2022] Open
Abstract
In silico methodologies have opened new avenues of research to understanding and predicting drug resistance, a pressing health issue that keeps rising at alarming pace. Sequence-based interpretation systems are routinely applied in clinical context in an attempt to predict mutation-based drug resistance and thus aid the choice of the most adequate antibiotic and antiviral therapy. An important limitation of approaches based on genotypic data exclusively is that mutations are not considered in the context of the three-dimensional (3D) structure of the target. Structure-based in silico methodologies are inherently more suitable to interpreting and predicting the impact of mutations on target-drug interactions, at the cost of higher computational and time demands when compared with sequence-based approaches. Herein, we present a fast, computationally inexpensive, sequence-to-structure-based approach to drug resistance prediction, which makes use of 3D protein structures encoded by input target sequences to draw binding-site comparisons with susceptible templates. Rather than performing atom-by-atom comparisons between input target and template structures, our workflow generates and compares Molecular Interaction Fields (MIFs) that map the areas of energetically favorable interactions between several chemical probe types and the target binding site. Quantitative, pairwise dissimilarity measurements between the target and the template binding sites are thus produced. The method is particularly suited to understanding changes to the 3D structure and the physicochemical environment introduced by mutations into the target binding site. Furthermore, the workflow relies exclusively on freeware, making it accessible to anyone. Using four datasets of known HIV-1 protease sequences as a case-study, we show that our approach is capable of correctly classifying resistant and susceptible sequences given as input. Guided by ROC curve analyses, we fined-tuned a dissimilarity threshold of classification that results in remarkable discriminatory performance (accuracy ≈ ROC AUC ≈ 0.99), illustrating the high potential of sequence-to-structure-, MIF-based approaches in the context of drug resistance prediction. We discuss the complementarity of the proposed methodology to existing prediction algorithms based on genotypic data. The present work represents a new step toward a more comprehensive and structurally-informed interpretation of the impact of genetic variability on the response to HIV-1 therapies.
Collapse
Affiliation(s)
- Nuno G Alves
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal
| | - Ana I Mata
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal
| | - João P Luís
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal
| | - Rui M M Brito
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal.,BSIM Therapeutics, Instituto Pedro Nunes, Coimbra, Portugal
| | - Carlos J V Simões
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal.,BSIM Therapeutics, Instituto Pedro Nunes, Coimbra, Portugal
| |
Collapse
|
91
|
Soria-Martinez L, Bauer S, Giesler M, Schelhaas S, Materlik J, Janus K, Pierzyna P, Becker M, Snyder NL, Hartmann L, Schelhaas M. Prophylactic Antiviral Activity of Sulfated Glycomimetic Oligomers and Polymers. J Am Chem Soc 2020; 142:5252-5265. [PMID: 32105452 DOI: 10.1021/jacs.9b13484] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this work, we investigate the potential of highly sulfated synthetic glycomimetics to act as inhibitors of viral binding/infection. Our results indicate that both long-chain glycopolymers and short-chain glycooligomers are capable of preventing viral infection. Notably, glycopolymers efficiently inhibit Human Papillomavirus (HPV16) infection in vitro and maintain their antiviral activity in vivo, while the glycooligomers exert their inhibitory function post attachment of viruses to cells. Moreover, when we tested the potential for broader activity against several other human pathogenic viruses, we observed broad-spectrum antiviral activity of these compounds beyond our initial assumptions. While the compounds tested displayed a range of antiviral efficacies, viruses with rather diverse glycan specificities such as Herpes Simplex Virus (HSV), Influenza A Virus (IAV), and Merkel Cell Polyomavirus (MCPyV) could be targeted. This opens new opportunities to develop broadly active glycomimetic inhibitors of viral entry and infection.
Collapse
Affiliation(s)
- Laura Soria-Martinez
- Institute of Cellular Virology, ZMBE, University of Münster, Münster 48149, Germany.,Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany
| | - Sebastian Bauer
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Markus Giesler
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Sonja Schelhaas
- European Institute for Molecular Imaging, University of Münster, Münster 48149, Germany.,Cells in Motion Interfaculty Centre CiMIC, University of Münster, Münster 48149, Germany
| | - Jennifer Materlik
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Kevin Janus
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Patrick Pierzyna
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Miriam Becker
- Institute of Cellular Virology, ZMBE, University of Münster, Münster 48149, Germany.,Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany
| | - Nicole L Snyder
- Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany.,Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany.,Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Mario Schelhaas
- Institute of Cellular Virology, ZMBE, University of Münster, Münster 48149, Germany.,Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany.,Cells in Motion Interfaculty Centre CiMIC, University of Münster, Münster 48149, Germany
| |
Collapse
|
92
|
Nanomaterials Designed for Antiviral Drug Delivery Transport across Biological Barriers. Pharmaceutics 2020; 12:pharmaceutics12020171. [PMID: 32085535 PMCID: PMC7076512 DOI: 10.3390/pharmaceutics12020171] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/10/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022] Open
Abstract
Viral infections are a major global health problem, representing a significant cause of mortality with an unfavorable continuously amplified socio-economic impact. The increased drug resistance and constant viral replication have been the trigger for important studies regarding the use of nanotechnology in antiviral therapies. Nanomaterials offer unique physico-chemical properties that have linked benefits for drug delivery as ideal tools for viral treatment. Currently, different types of nanomaterials namely nanoparticles, liposomes, nanospheres, nanogels, nanosuspensions and nanoemulsions were studied either in vitro or in vivo for drug delivery of antiviral agents with prospects to be translated in clinical practice. This review highlights the drug delivery nanosystems incorporating the major antiviral classes and their transport across specific barriers at cellular and intracellular level. Important reflections on nanomedicines currently approved or undergoing investigations for the treatment of viral infections are also discussed. Finally, the authors present an overview on the requirements for the design of antiviral nanotherapeutics.
Collapse
|
93
|
Herpes Virus Infections Other than Cytomegalovirus in the Recipients of Hematopoietic Stem Cell Transplantation. Infect Dis Clin North Am 2019; 33:467-484. [PMID: 31005137 DOI: 10.1016/j.idc.2019.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This review discusses the epidemiologic and clinical aspects of herpes viruses other than cytomegalovirus in patients who have undergone hematopoietic stem cell transplantation.
Collapse
|
94
|
Xu J, Cai Y, Jiang B, Li X, Jin H, Liu W, Kong Z, Hong J, Sealy JE, Iqbal M, Li Y. An optimized aptamer-binding viral tegument protein VP8 inhibits the production of Bovine Herpesvirus-1 through blocking nucleocytoplasmic shuttling. Int J Biol Macromol 2019; 140:1226-1238. [PMID: 31445153 DOI: 10.1016/j.ijbiomac.2019.08.165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 10/26/2022]
Abstract
Bovine herpesvirus 1 (BoHV-1) is a major pathogen of infectious bovine rhinotracheitis in bovine. Previously, we generated the aptamer IBRV A4 using systemic evolution of ligands by exponential enrichment. This aptamer inhibited infectivity of BoHV-1 by blocking viral particle absorption onto cell membranes. In this study, we found that the major tegument protein VP8 of BoHV-1 was involved in inhibition of infectious virus production by IBRV A4. We improved the affinity of IBRV A4 for VP8 by optimizing aptamer's structure and repeat conformation. An optimized aptamer, IBRV A4.7, was constructed with quadruple binding sites and a new stem-loop structure, which had a stronger binding affinity for VP8 or BoHV-1 than raw aptamer IBRV A4. IBRV A4.7 bound to VP8 with a dissociation constant (Kd) value of 0.2054 ± 0.03948 nM and bound to BoHV-1 with a Kd value of 0.3637 ± 0.05452 nM. Crucially, IBRV A4.7 had improved antiviral activity compared to IBRV A4, with a half-maximal inhibitory concentration of 1.16 ± 0.042 μM. Our results also revealed IBRV A4.7 inhibited BoHV-1 production in MDBK cells through blocking nucleocytoplasmic shuttling of viral VP8 in BoHV-1-infected MDBK cells. In conclusion, the aptamer IBRV A4.7 may have potency in preventing outbreaks in herds due to reactivation of latency.
Collapse
Affiliation(s)
- Jian Xu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China
| | - Yunhong Cai
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China
| | - Bo Jiang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China
| | - Xiaoyang Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China
| | - Huan Jin
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China
| | - Wenxiao Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China
| | - Zimeng Kong
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, PR China
| | - Jiabing Hong
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, PR China
| | - Joshua E Sealy
- Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China; The Pirbright Institute, Ash Rd, Pirbright, Woking GU24 0NF, United Kingdom
| | - Munir Iqbal
- Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China; The Pirbright Institute, Ash Rd, Pirbright, Woking GU24 0NF, United Kingdom
| | - Yongqing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, PR China; Sino-UK Joint Laboratory for Prevention & Control of Infectious Diseases in Livestock and Poultry, Beijing 100097, PR China.
| |
Collapse
|
95
|
Wiehe A, O'Brien JM, Senge MO. Trends and targets in antiviral phototherapy. Photochem Photobiol Sci 2019; 18:2565-2612. [PMID: 31397467 DOI: 10.1039/c9pp00211a] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Photodynamic therapy (PDT) is a well-established treatment option in the treatment of certain cancerous and pre-cancerous lesions. Though best-known for its application in tumor therapy, historically the photodynamic effect was first demonstrated against bacteria at the beginning of the 20th century. Today, in light of spreading antibiotic resistance and the rise of new infections, this photodynamic inactivation (PDI) of microbes, such as bacteria, fungi, and viruses, is gaining considerable attention. This review focuses on the PDI of viruses as an alternative treatment in antiviral therapy, but also as a means of viral decontamination, covering mainly the literature of the last decade. The PDI of viruses shares the general action mechanism of photodynamic applications: the irradiation of a dye with light and the subsequent generation of reactive oxygen species (ROS) which are the effective phototoxic agents damaging virus targets by reacting with viral nucleic acids, lipids and proteins. Interestingly, a light-independent antiviral activity has also been found for some of these dyes. This review covers the compound classes employed in the PDI of viruses and their various areas of use. In the medical area, currently two fields stand out in which the PDI of viruses has found broader application: the purification of blood products and the treatment of human papilloma virus manifestations. However, the PDI of viruses has also found interest in such diverse areas as water and surface decontamination, and biosafety.
Collapse
Affiliation(s)
- Arno Wiehe
- biolitec research GmbH, Otto-Schott-Str. 15, 07745 Jena, Germany. and Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Jessica M O'Brien
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland.
| | - Mathias O Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
96
|
Caesar LK, Cech NB. Synergy and antagonism in natural product extracts: when 1 + 1 does not equal 2. Nat Prod Rep 2019; 36:869-888. [PMID: 31187844 PMCID: PMC6820002 DOI: 10.1039/c9np00011a] [Citation(s) in RCA: 353] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Covering: 2000 to 2019 According to a 2012 survey from the Centers for Disease Control and Prevention, approximately 18% of the U.S. population uses natural products (including plant-based or botanical preparations) for treatment or prevention of disease. The use of plant-based medicines is even more prevalent in developing countries, where for many they constitute the primary health care modality. Proponents of the medicinal use of natural product mixtures often claim that they are more effective than purified compounds due to beneficial "synergistic" interactions. A less-discussed phenomenon, antagonism, in which effects of active constituents are masked by other compounds in a complex mixture, also occurs in natural product mixtures. Synergy and antagonism are notoriously difficult to study in a rigorous fashion, particularly given that natural products chemistry research methodology is typically devoted to reducing complexity and identifying single active constituents for drug development. This report represents a critical review with commentary about the current state of the scientific literature as it relates to studying combination effects (including both synergy and antagonism) in natural product extracts. We provide particular emphasis on analytical and Big Data approaches for identifying synergistic or antagonistic combinations and elucidating the mechanisms that underlie their interactions. Specific case studies of botanicals in which synergistic interactions have been documented are also discussed. The topic of synergy is important given that consumer use of botanical natural products and associated safety concerns continue to garner attention by the public and the media. Guidance by the natural products community is needed to provide strategies for effective evaluation of safety and toxicity of botanical mixtures and to drive discovery in botanical natural product research.
Collapse
Affiliation(s)
- Lindsay K Caesar
- Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA.
| | | |
Collapse
|
97
|
Case Report: Varicella-zoster Encephalitis with Acute Retinal Necrosis and Oculomotor Nerve Palsy. Optom Vis Sci 2019; 96:367-371. [DOI: 10.1097/opx.0000000000001370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
98
|
Koujah L, Shukla D, Naqvi AR. CRISPR-Cas based targeting of host and viral genes as an antiviral strategy. Semin Cell Dev Biol 2019; 96:53-64. [PMID: 30953741 DOI: 10.1016/j.semcdb.2019.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/23/2022]
Abstract
Viral infections in human are leading cause of mortality and morbidity across the globe. Several viruses (including HIV and Herpesvirus), have evolved ingenious strategies to evade host-immune system and persist life-long. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated (Cas) is an ancient antiviral system recently discovered in bacteria that has shown tremendous potential as a precise, invariant genome editing tool. Using CRISPR-Cas based system to activate host defenses or genetic modification of viral genome can provide novel, exciting and successful antiviral mechanisms and treatment modalities. In this review, we will provide progress on the CRISPR-Cas based antiviral approaches that facilitate clearance of virus-infected cells and/or prohibit virus infection or replication. We will discuss on the possibilities of CRIPSR-Cas as prophylaxis and therapy in viral infections and review the challenges of this potent gene editing technology.
Collapse
Affiliation(s)
- Lulia Koujah
- Department of Microbiology and Immunology, University of Illinois at Chicago, IL, 60612, USA; Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, IL, 60612, USA; Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA.
| | - Afsar R Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
99
|
Patra U, Mukhopadhyay U, Sarkar R, Mukherjee A, Chawla-Sarkar M. RA-839, a selective agonist of Nrf2/ARE pathway, exerts potent anti-rotaviral efficacy in vitro. Antiviral Res 2018; 161:53-62. [PMID: 30465784 DOI: 10.1016/j.antiviral.2018.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 12/21/2022]
Abstract
Acute watery diarrhea due to Rotavirus (RV) infection is associated with high infantile morbidity and mortality in countries with compromised socio-economic backgrounds. Although showing promising trends in developed countries, the efficacy of currently licensed RV vaccines is sub-optimal in socio-economically poor settings with high disease burden. Currently, there are no approved anti-rotaviral drugs adjunct to classical vaccination program. Interestingly, dissecting host-rotavirus interaction has yielded novel, non-mutable host determinants which can be subjected to interventions by selective small molecules. The present study was undertaken to evaluate the anti-RV potential of RA-839, a recently discovered small molecule with potent and highly selective agonistic activity towards cellular redox stress-sensitive Nuclear factor erytheroid-derived-2-like 2 (Nrf2)/Antioxidant Response Element (ARE) pathway. In vitro studies revealed that RA-839 inhibits RV RNA and protein expression, viroplasm formation, yield of virion progeny and virus-induced cytopathy independent of RV strains, RV-permissive cell lines and without bystander cytotoxicity. Anti-RV potency of RA-839 was subsequently identified to be independent of stochastic Interferon (IFN) stimulation but to be dependent on RA-839's ability to stimulate Nrf2/ARE signaling. Interestingly, anti-rotaviral effects of RA-839 were also mimicked by 2-Cyano-3, 12-dioxo-oleana-1, 9(11)-dien-28-oic acid methyl ester (CDDO-Me) and Hemin, two classical pharmacological activators of Nrf2/ARE pathway. Overall, this study highlights that RA-839 is a potent antagonist of RV propagation in vitro and can be developed as anti-rotaviral therapeutics.
Collapse
Affiliation(s)
- Upayan Patra
- Division of Virology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road Scheme- XM, Beliaghata, Kolkata 700010, India
| | - Urbi Mukhopadhyay
- Division of Virology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road Scheme- XM, Beliaghata, Kolkata 700010, India
| | - Rakesh Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road Scheme- XM, Beliaghata, Kolkata 700010, India
| | - Arpita Mukherjee
- Division of Virology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road Scheme- XM, Beliaghata, Kolkata 700010, India
| | - Mamta Chawla-Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road Scheme- XM, Beliaghata, Kolkata 700010, India.
| |
Collapse
|
100
|
Tajpara P, Mildner M, Schmidt R, Vierhapper M, Matiasek J, Popow-Kraupp T, Schuster C, Elbe-Bürger A. A Preclinical Model for Studying Herpes Simplex Virus Infection. J Invest Dermatol 2018; 139:673-682. [PMID: 30414908 PMCID: PMC7100788 DOI: 10.1016/j.jid.2018.08.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/03/2018] [Accepted: 08/12/2018] [Indexed: 01/29/2023]
Abstract
Herpes simplex virus (HSV) infections can cause considerable morbidity. Currently, nucleoside analogues such as acyclovir are widely used for treatment. However, HSV infections resistant to these drugs are a clinical problem among immunocompromised patients. To provide more efficient therapy and to counteract resistance, a different class of antiviral compounds has been developed. Pritelivir, a helicase primase inhibitor, represents a promising candidate for improved therapy. Here, we established an HSV-1 infection model on microneedle-pretreated human skin ex vivo. We identified HSV-1–specific histological changes (e.g., cytopathic effects, multinucleated giant cells), down-regulation of nectin-1, nuclear translocation of NF-κB (p65), interferon regulatory factor 3 (IRF3), and signaling of the IFN-inducible protein MxA. Accordingly, this model was used to test the potency of pritelivir compared with the standard drug acyclovir. We discovered that both drugs had a comparable efficacy for inhibiting HSV-1 replication, suggesting that pritelivir could be an alternative therapeutic agent for patients infected with acyclovir-resistant strains. To our knowledge, we present a previously unreported ex vivo HSV-1 infection model with abdominal human skin to test antiviral drugs, thus bridging the gap between in vitro and in vivo drug screening and providing a valuable preclinical platform for HSV research.
Collapse
Affiliation(s)
- Poojabahen Tajpara
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Ralf Schmidt
- Department of Laboratory Medicine, Division of Clinical Virology, Medical University of Vienna, Vienna, Austria
| | - Martin Vierhapper
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Johannes Matiasek
- Department of Plastic, Aesthetic and Reconstructive Surgery, St. Josef Hospital, Vienna, Austria
| | - Theresia Popow-Kraupp
- Department of Laboratory Medicine, Division of Clinical Virology, Medical University of Vienna, Vienna, Austria
| | - Christopher Schuster
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - Adelheid Elbe-Bürger
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|