1
|
Lei R, Kim W, Lv H, Mou Z, Scherm MJ, Schmitz AJ, Turner JS, Tan TJC, Wang Y, Ouyang WO, Liang W, Rivera-Cardona J, Teo C, Graham CS, Brooke CB, Presti RM, Mok CKP, Krammer F, Dai X, Ellebedy AH, Wu NC. Leveraging vaccination-induced protective antibodies to define conserved epitopes on influenza N2 neuraminidase. Immunity 2023; 56:2621-2634.e6. [PMID: 37967533 PMCID: PMC10655865 DOI: 10.1016/j.immuni.2023.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 07/19/2023] [Accepted: 10/10/2023] [Indexed: 11/17/2023]
Abstract
There is growing appreciation for neuraminidase (NA) as an influenza vaccine target; however, its antigenicity remains poorly characterized. In this study, we isolated three broadly reactive N2 antibodies from the plasmablasts of a single vaccinee, including one that cross-reacts with NAs from seasonal H3N2 strains spanning five decades. Although these three antibodies have diverse germline usages, they recognize similar epitopes that are distant from the NA active site and instead involve the highly conserved underside of NA head domain. We also showed that all three antibodies confer prophylactic and therapeutic protection in vivo, due to both Fc effector functions and NA inhibition through steric hindrance. Additionally, the contribution of Fc effector functions to protection in vivo inversely correlates with viral growth inhibition activity in vitro. Overall, our findings advance the understanding of NA antibody response and provide important insights into the development of a broadly protective influenza vaccine.
Collapse
Affiliation(s)
- Ruipeng Lei
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea
| | - Huibin Lv
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zongjun Mou
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Michael J Scherm
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aaron J Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Timothy J C Tan
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yiquan Wang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wenhao O Ouyang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Weiwen Liang
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Joel Rivera-Cardona
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Chuyun Teo
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Claire S Graham
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Christopher B Brooke
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rachel M Presti
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Chris K P Mok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; S.H. Ho Research Centre for Infectious Diseases, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Xinghong Dai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Nicholas C Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
2
|
Gattani A, Agrawal A, Khan MH, Gupta R, Singh P. Evaluation of catalytic activity of human and animal origin viral neuraminidase: Current prospect. Anal Biochem 2023; 671:115157. [PMID: 37061113 DOI: 10.1016/j.ab.2023.115157] [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: 10/13/2022] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
With the exception of plants, almost all living organisms synthesize neuraminidase/sialidase. It is a one among the crucial proteins that controls how virulent a microorganism is. An essential enzyme in orthomyxoviruses and paramyxoviruses that destroys receptors is neuraminidase. It plays a number of roles throughout the viral life cycle in addition to one that involves the release of progeny virus particles. This protein is an important target for therapeutic interventions and diagnostic assays. Neuraminidase inhibitors effectively prevent the spread of disease and viral infection. Sensitive, quick, and inexpensive high throughput assays are needed to screen for specific neuraminidase inhibitory chemicals. To characterize the neuraminidase catalytic activity, however, the traditional assays are still the most common in laboratories. This review gives a brief overview of these neuraminidase assays and recent, innovative developments, particularly those involving biosensors.
Collapse
Affiliation(s)
- Anil Gattani
- Department of Veterinary Biochemistry, College of Veterinary Science & Animal Husbandry, Jabalpur, M.P, India.
| | - Aditya Agrawal
- Department of Veterinary Biochemistry, College of Veterinary Science & Animal Husbandry, Rewa, M.P, India
| | - M Hira Khan
- Department of Veterinary Biochemistry, College of Veterinary Science & Animal Husbandry, Jabalpur, M.P, India
| | - Rohini Gupta
- Department of Medicine, College of Veterinary Science & Animal Husbandry, Jabalpur, M.P, India
| | - Praveen Singh
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute Izatnagar, 243122, Bareilly, UP, India; Biophysics Section, ICAR-Indian Veterinary Research Institute Izatnagar, 243122, Bareilly, UP, India
| |
Collapse
|
3
|
Kolosova NP, Ilyicheva TN, Unguryan VV, Danilenko AV, Svyatchenko SV, Onhonova GS, Goncharova NI, Kosenko MN, Gudymo AS, Marchenko VY, Shvalov AN, Susloparov IM, Tregubchak TV, Gavrilova EV, Maksyutov RA, Ryzhikov AB. Re-Emergence of Circulation of Seasonal Influenza during COVID-19 Pandemic in Russia and Receptor Specificity of New and Dominant Clade 3C.2a1b.2a.2 A(H3N2) Viruses in 2021-2022. Pathogens 2022; 11:1388. [PMID: 36422639 PMCID: PMC9698969 DOI: 10.3390/pathogens11111388] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2023] Open
Abstract
The circulation of seasonal influenza in 2020-2021 around the world was drastically reduced after the start of the COVID-19 pandemic and the implementation of mitigation strategies. The influenza virus circulation reemerged in 2021-2022 with the global spread of the new genetic clade 3C.2a1b.2a.2 of A(H3N2) viruses. The purpose of this study was to characterize influenza viruses in the 2021-2022 season in Russia and to analyze the receptor specificity properties of the 3C.2a1b.2a.2 A(H3N2) viruses. Clinical influenza samples were collected at the local Sanitary-and-Epidemiological Centers of Rospotrebnadzor. Whole genome sequencing was performed using NGS. The receptor specificity of hemagglutinin was evaluated using molecular modeling and bio-layer interferometry. Clinical samples from 854 cases of influenza A and B were studied; A(H3N2) viruses were in the majority of the samples. All genetically studied A(H3N2) viruses belonged to the new genetic clade 3C.2a1b.2a.2. Molecular modeling analysis suggested a higher affinity of hemagglutinin of 3C.2a1b.2a.2. A(H3N2) viruses to the α2,6 human receptor. In vitro analysis using a trisaccharide 6'-Sialyl-N-acetyllactosamine receptor analog did not resolve the differences in the receptor specificity of 3C.2a1b.2a.2 clade viruses from viruses belonging to the 3C.2a1b.2a.1 clade. Further investigation of the A(H3N2) viruses is required for the evaluation of their possible adaptive advantages. Constant monitoring and characterization of influenza are critical for epidemiological analysis.
Collapse
Affiliation(s)
- Natalia P. Kolosova
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Tatiana N. Ilyicheva
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Vasily V. Unguryan
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
- Department of Physics, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Alexey V. Danilenko
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Svetlana V. Svyatchenko
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Galina S. Onhonova
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Natalia I. Goncharova
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Maksim N. Kosenko
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Andrey S. Gudymo
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Vasiliy Y. Marchenko
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Alexander N. Shvalov
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Ivan M. Susloparov
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Tatiana V. Tregubchak
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Elena V. Gavrilova
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Rinat A. Maksyutov
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Alexander B. Ryzhikov
- State Research Centre of Virology and Biotechnology “Vector”, Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| |
Collapse
|
4
|
Stannard HL, Mifsud EJ, Wildum S, Brown SK, Koszalka P, Shishido T, Kojima S, Omoto S, Baba K, Kuhlbusch K, Hurt AC, Barr IG. Assessing the fitness of a dual-antiviral drug resistant human influenza virus in the ferret model. Commun Biol 2022; 5:1026. [PMID: 36171475 PMCID: PMC9517990 DOI: 10.1038/s42003-022-04005-4] [Citation(s) in RCA: 1] [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: 05/05/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022] Open
Abstract
Influenza antivirals are important tools in our fight against annual influenza epidemics and future influenza pandemics. Combinations of antivirals may reduce the likelihood of drug resistance and improve clinical outcomes. Previously, two hospitalised immunocompromised influenza patients, who received a combination of a neuraminidase inhibitor and baloxavir marboxil, shed influenza viruses resistant to both drugs. Here-in, the replicative fitness of one of these A(H1N1)pdm09 virus isolates with dual resistance mutations (NA-H275Y and PA-I38T) was similar to wild type virus (WT) in vitro, but reduced in the upper respiratory tracts of challenged ferrets. The dual-mutant virus transmitted well between ferrets in an airborne transmission model, but was outcompeted by the WT when the two viruses were co-administered. These results indicate the dual-mutant virus had a moderate loss of viral fitness compared to the WT virus, suggesting that while person-to-person transmission of the dual-resistant virus may be possible, widespread community transmission is unlikely.
Collapse
Affiliation(s)
- Harry L Stannard
- WHO Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Edin J Mifsud
- WHO Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | | | - Sook Kwan Brown
- WHO Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Paulina Koszalka
- WHO Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | | | | | | | | | | | | | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Influenza A (N1-N9) and Influenza B (B/Victoria and B/Yamagata) Neuraminidase Pseudotypes as Tools for Pandemic Preparedness and Improved Influenza Vaccine Design. Vaccines (Basel) 2022; 10:vaccines10091520. [PMID: 36146598 PMCID: PMC9571397 DOI: 10.3390/vaccines10091520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
To better understand how inhibition of the influenza neuraminidase (NA) protein contributes to protection against influenza, we produced lentiviral vectors pseudotyped with an avian H11 hemagglutinin (HA) and the NA of all influenza A (N1–N9) subtypes and influenza B (B/Victoria and B/Yamagata). These NA viral pseudotypes (PV) possess stable NA activity and can be utilized as target antigens in in vitro assays to assess vaccine immunogenicity. Employing these NA PV, we developed an enzyme-linked lectin assay (pELLA) for routine serology to measure neuraminidase inhibition (NI) titers of reference antisera, monoclonal antibodies and post-vaccination sera with various influenza antigens. We also show that the pELLA is more sensitive than the commercially available NA-Fluor™ in detecting NA inhibition in these samples. Our studies may lead to establishing the protective NA titer that contributes to NA-based immunity. This will aid in the design of superior, longer lasting and more broadly protective vaccines that can be employed together with HA-targeted vaccines in a pre-pandemic approach.
Collapse
|
6
|
Protein Disulfide Isomerase A3 Regulates Influenza Neuraminidase Activity and Influenza Burden in the Lung. Int J Mol Sci 2022; 23:ijms23031078. [PMID: 35162999 PMCID: PMC8834910 DOI: 10.3390/ijms23031078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/02/2022] Open
Abstract
Influenza (IAV) neuraminidase (NA) is a glycoprotein required for the viral exit from the cell. NA requires disulfide bonds for proper function. We have recently demonstrated that protein disulfide isomerase (PDI)A3 is required for oxidative folding of IAV hemagglutinin (HA), and viral propagation. However, it not known whether PDIs are required for NA maturation or if these interactions represent a putative target for the treatment of influenza infection. We sought to determine whether PDIA3 is required for disulfide bonds of NA, its activity, and propagation of the virus. Requirement of disulfides for NA oligomerization and activity were determined using biotin switch and redox assays in WT and PDIA3−/− in A549 cells. A PDI specific inhibitor (LOC14) was utilized to determine the requirement of PDIs in NA activity, IAV burden, and inflammatory response in A549 and primary mouse tracheal epithelial cells. Mice were treated with the inhibitor LOC14 and subsequently examined for IAV burden, NA activity, cytokine, and immune response. IAV-NA interacts with PDIA3 and this interaction is required for NA activity. PDIA3 ablation or inhibition decreased NA activity, viral burden, and inflammatory response in lung epithelial cells. LOC14 treatment significantly attenuated the influenza-induced inflammatory response in mice including the overall viral burden. These results provide evidence for PDIA3 inhibition suppressing NA activity, potentially providing a novel platform for host-targeted antiviral therapies.
Collapse
|
7
|
MARCH8 Restricts Influenza A Virus Infectivity but Does Not Downregulate Viral Glycoprotein Expression at the Surface of Infected Cells. mBio 2021; 12:e0148421. [PMID: 34517760 PMCID: PMC8546552 DOI: 10.1128/mbio.01484-21] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Membrane-associated RING-CH8 (MARCH8) impairs the cell surface expression of envelope glycoproteins from different viruses, reducing their incorporation into virions. Using stable cell lines with inducible MARCH8 expression, we show that MARCH8 did not alter susceptibility to influenza A virus (IAV) infection, but virions released from infected cells were markedly less infectious. Knockdown of endogenous MARCH8 confirmed its effect on IAV infectivity. The expression of MARCH8 impaired the infectivity of both H3N2 and H1N1 strains and was dependent on its E3 ligase activity. Although virions released in the presence of MARCH8 expressed smaller amounts of viral hemagglutinin (HA) and neuraminidase (NA) proteins, there was no impact on levels of the viral HA, NA, or matrix 2 (M2) proteins detected on the surface of infected cells. Moreover, mutation of lysine residues in the cytoplasmic tails of HA, NA, and/or M2, or in the viral M1 protein, did not abrogate MARCH8-mediated restriction. While MARCH1 and -8 target similar immunological ligands and both restrict HIV-1, only MARCH8 inhibited IAV infectivity. Deletion of the N-terminal cytoplasmic (N-CT) domain of MARCH8 confirmed it to be a critical determinant of IAV inhibition. Of interest, deletion of the MARCH1 N-CT or its replacement with the MARCH8 N-CT resulted in acquisition of IAV restriction. Together, these data demonstrate that MARCH8 restricts a late stage in IAV replication by a mechanism distinct to its reported activity against other viruses. Moreover, we show that the N-CT of MARCH8 is essential for anti-IAV activity, whereas the MARCH1 N-CT inhibits its ability to restrict IAV.
Collapse
|
8
|
Hussain S, Daniels RS, Wharton SA, Howell S, Halai C, Kunzelmann S, Whittaker L, McCauley JW. Reduced sialidase activity of influenza A(H3N2) neuraminidase associated with positively charged amino acid substitutions. J Gen Virol 2021; 102. [PMID: 34596510 DOI: 10.1099/jgv.0.001648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuraminidase (NA) inhibitors (NAI), oseltamivir and zanamivir, are the main antiviral medications for influenza and monitoring of susceptibility to these antivirals is routinely done by determining 50 % inhibitory concentrations (IC50) with MUNANA substrate. During 2010-2019, levels of A(H3N2) viruses presenting reduced NAI inhibition (RI) were low (~0.75 %) but varied year-on-year. The highest proportions of viruses showing RI were observed during the 2013-2014, 2016-2017 and 2017-2018 Northern Hemisphere seasons. The majority of RI viruses were found to contain positively charged NA amino acid substitutions of N329K, K/S329R, S331R or S334R, being notably higher during the 2016-2017 season. Sialidase activity kinetics were determined for viruses of RI phenotype and contemporary wild-type (WT) viruses showing close genetic relatedness and displaying normal inhibition (NI). RI phenotypes resulted from reduced sialidase activity compared to relevant WT viruses. Those containing S329R or N329K or S331R showed markedly higher Km for the substrate and Ki values for NAIs, while those with S334R showed smaller effects. Substitutions at N329 and S331 disrupt a glycosylation sequon (NDS), confirmed to be utilised by mass spectrometry. However, gain of positive charge at all three positions was the major factor influencing the kinetic effects, not loss of glycosylation. Because of the altered enzyme characteristics NAs carrying these substitutions cannot be assessed reliably for susceptibility to NAIs using standard MUNANA-based assays due to reductions in the affinity of the enzyme for its substrate and the concentration of the substrate usually used.
Collapse
Affiliation(s)
- Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| | - Rodney S Daniels
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| | - Stephen A Wharton
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| | - Steven Howell
- Protein Analysis and Proteomics Science Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Chandrika Halai
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| | - Simone Kunzelmann
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Lynne Whittaker
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| | - John W McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| |
Collapse
|
9
|
Campbell AC, Tanner JJ, Krause KL. Optimisation of Neuraminidase Expression for Use in Drug Discovery by Using HEK293-6E Cells. Viruses 2021; 13:v13101893. [PMID: 34696326 PMCID: PMC8538103 DOI: 10.3390/v13101893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 12/02/2022] Open
Abstract
Influenza virus is a highly contagious virus that causes significant human mortality and morbidity annually. The most effective drugs for treating influenza are the neuraminidase inhibitors, but resistance to these inhibitors has emerged, and additional drug discovery research on neuraminidase and other targets is needed. Traditional methods of neuraminidase production from embryonated eggs are cumbersome, while insect cell derived protein is less reflective of neuraminidase produced during human infection. Herein we describe a method for producing neuraminidase from a human cell line, HEK293-6E, and demonstrate the method by producing the neuraminidase from the 1918 H1N1 pandemic influenza strain. This method produced high levels of soluble neuraminidase expression (>3000 EU/mL), was enhanced by including a secretion signal from a viral chemokine binding protein, and does not require co-expression of additional proteins. The neuraminidase produced was of sufficient quantity and purity to support high resolution crystal structure determination. The structure solved using this protein conformed to the previously reported structure. Notably the glycosylation at three asparagine residues was superior in quality to that from insect cell derived neuraminidase. This method of production of neuraminidase should prove useful in further studies, such as the characterisation of inhibitor binding.
Collapse
Affiliation(s)
- Ashley C. Campbell
- Department of Biochemistry, University of Otago, 710 Cumberland St., Dunedin 9016, New Zealand;
| | - John J. Tanner
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA;
- Department of Chemistry, University of Missouri, Columbia, MO 65211, USA
| | - Kurt L. Krause
- Department of Biochemistry, University of Otago, 710 Cumberland St., Dunedin 9016, New Zealand;
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1010, New Zealand
- Correspondence:
| |
Collapse
|
10
|
Nishioka Y, Nagano K, Koga Y, Okada Y, Mori I, Hayase A, Mori T, Manabe K. Lactic acid as a major contributor to hand surface infection barrier and its association with morbidity to infectious disease. Sci Rep 2021; 11:18608. [PMID: 34545150 PMCID: PMC8452697 DOI: 10.1038/s41598-021-98042-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
Although the surface of the human hands contains high antimicrobial activity, studies investigating the precise components involved and the relationship between natural antimicrobial activity and morbidity in infectious diseases are limited. In this study, we developed a method to quantitatively measure the antimicrobial activity of hand surface components. Using a clinical survey, we validated the feasibility of our method and identified antimicrobial factors on the surface of the human hand. In a retrospective observational study, we compared the medical histories of the participants to assess infectious diseases. We found that the antimicrobial activity on the surface of the hands was significantly lower in the high morbidity group (N = 55) than in the low morbidity group (N = 54), indicating a positive association with the history of infection in individuals. A comprehensive analysis of the hand surface components indicated that organic acids, especially lactic acid and antimicrobial peptides, are highly correlated with antimicrobial activity. Moreover, the application of lactic acid using the amount present on the surface of the hand significantly improved the antimicrobial activity. These findings suggest that hand hygiene must be improved to enhance natural antimicrobial activity on the surface of the hands.
Collapse
Affiliation(s)
- Yuki Nishioka
- Personal Health Care Products Research, Kao Corporation, 2-1-3, Bunka, Sumida-ku, Tokyo, 131-8501, Japan
| | - Kenichi Nagano
- Analytical Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai, Haga, Tochigi, 321-3497, Japan
| | - Yoshitaka Koga
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai, Haga, Tochigi, 321-3497, Japan
| | - Yasuhiro Okada
- Personal Health Care Products Research, Kao Corporation, 2-1-3, Bunka, Sumida-ku, Tokyo, 131-8501, Japan
| | - Ichiro Mori
- Personal Health Care Products Research, Kao Corporation, 2-1-3, Bunka, Sumida-ku, Tokyo, 131-8501, Japan
| | - Atsuko Hayase
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai, Haga, Tochigi, 321-3497, Japan
| | - Takuya Mori
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai, Haga, Tochigi, 321-3497, Japan
| | - Kenji Manabe
- Personal Health Care Products Research, Kao Corporation, 2-1-3, Bunka, Sumida-ku, Tokyo, 131-8501, Japan.
| |
Collapse
|
11
|
Lin X, Lin F, Liang T, Ducatez MF, Zanin M, Wong SS. Antibody Responsiveness to Influenza: What Drives It? Viruses 2021; 13:v13071400. [PMID: 34372607 PMCID: PMC8310379 DOI: 10.3390/v13071400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023] Open
Abstract
The induction of a specific antibody response has long been accepted as a serological hallmark of recent infection or antigen exposure. Much of our understanding of the influenza antibody response has been derived from studying antibodies that target the hemagglutinin (HA) protein. However, growing evidence points to limitations associated with this approach. In this review, we aim to highlight the issue of antibody non-responsiveness after influenza virus infection and vaccination. We will then provide an overview of the major factors known to influence antibody responsiveness to influenza after infection and vaccination. We discuss the biological factors such as age, sex, influence of prior immunity, genetics, and some chronic infections that may affect the induction of influenza antibody responses. We also discuss the technical factors, such as assay choices, strain variations, and viral properties that may influence the sensitivity of the assays used to measure influenza antibodies. Understanding these factors will hopefully provide a more comprehensive picture of what influenza immunogenicity and protection means, which will be important in our effort to improve influenza vaccines.
Collapse
Affiliation(s)
- Xia Lin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | - Fangmei Lin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | - Tingting Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | | | - Mark Zanin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Sook-San Wong
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
- School of Public Health, The University of Hong Kong, Hong Kong, China
- Correspondence: ; Tel.: +86-178-2584-6078
| |
Collapse
|
12
|
Petrou A, Fesatidou M, Geronikaki A. Thiazole Ring-A Biologically Active Scaffold. Molecules 2021; 26:3166. [PMID: 34070661 PMCID: PMC8198555 DOI: 10.3390/molecules26113166] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/15/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Thiazole is a good pharmacophore nucleus due to its various pharmaceutical applications. Its derivatives have a wide range of biological activities such as antioxidant, analgesic, and antimicrobial including antibacterial, antifungal, antimalarial, anticancer, antiallergic, antihypertensive, anti-inflammatory, and antipsychotic. Indeed, the thiazole scaffold is contained in more than 18 FDA-approved drugs as well as in numerous experimental drugs. OBJECTIVE To summarize recent literature on the biological activities of thiazole ring-containing compounds Methods: A literature survey regarding the topics from the year 2015 up to now was carried out. Older publications were not included, since they were previously analyzed in available peer reviews. RESULTS Nearly 124 research articles were found, critically analyzed, and arranged regarding the synthesis and biological activities of thiazoles derivatives in the last 5 years.
Collapse
Affiliation(s)
| | | | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.P.); (M.F.)
| |
Collapse
|
13
|
Boonnak K, Mansanguan C, Schuerch D, Boonyuen U, Lerdsamran H, Jiamsomboon K, Sae Wang F, Huntrup A, Prasertsopon J, Kosoltanapiwat N, Puthavathana P. Molecular Characterization of Seasonal Influenza A and B from Hospitalized Patients in Thailand in 2018-2019. Viruses 2021; 13:977. [PMID: 34070388 PMCID: PMC8228477 DOI: 10.3390/v13060977] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/11/2022] Open
Abstract
Influenza viruses continue to be a major public health threat due to the possible emergence of more virulent influenza virus strains resulting from dynamic changes in virus adaptability, consequent of functional mutations and antigenic drift in surface proteins, especially hemagglutinin (HA) and neuraminidase (NA). In this study, we describe the genetic and evolutionary characteristics of H1N1, H3N2, and influenza B strains detected in severe cases of seasonal influenza in Thailand from 2018 to 2019. We genetically characterized seven A/H1N1 isolates, seven A/H3N2 isolates, and six influenza B isolates. Five of the seven A/H1N1 viruses were found to belong to clade 6B.1 and were antigenically similar to A/Switzerland/3330/2017 (H1N1), whereas two isolates belonged to clade 6B.1A1 and clustered with A/Brisbane/02/2018 (H1N1). Interestingly, we observed additional mutations at antigenic sites (S91R, S181T, T202I) as well as a unique mutation at a receptor binding site (S200P). Three-dimensional (3D) protein structure analysis of hemagglutinin protein reveals that this unique mutation may lead to the altered binding of the HA protein to a sialic acid receptor. A/H3N2 isolates were found to belong to clade 3C.2a2 and 3C.2a1b, clustering with A/Switzerland/8060/2017 (H3N2) and A/South Australia/34/2019 (H3N2), respectively. Amino acid sequence analysis revealed 10 mutations at antigenic sites including T144A/I, T151K, Q213R, S214P, T176K, D69N, Q277R, N137K, N187K, and E78K/G. All influenza B isolates in this study belong to the Victoria lineage. Five out of six isolates belong to clade 1A3-DEL, which relate closely to B/Washington/02/2009, with one isolate lacking the three amino acid deletion on the HA segment at position K162, N163, and D164. In comparison to the B/Colorado/06/2017, which is the representative of influenza B Victoria lineage vaccine strain, these substitutions include G129D, G133R, K136E, and V180R for HA protein. Importantly, the susceptibility to oseltamivir of influenza B isolates, but not A/H1N1 and A/H3N2 isolates, were reduced as assessed by the phenotypic assay. This study demonstrates the importance of monitoring genetic variation in influenza viruses regarding how acquired mutations could be associated with an improved adaptability for efficient transmission.
Collapse
Affiliation(s)
- Kobporn Boonnak
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (K.B.); (D.S.); (K.J.); (F.S.W.); (N.K.)
| | - Chayasin Mansanguan
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Dennis Schuerch
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (K.B.); (D.S.); (K.J.); (F.S.W.); (N.K.)
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Hatairat Lerdsamran
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand; (H.L.); (J.P.)
| | - Kultida Jiamsomboon
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (K.B.); (D.S.); (K.J.); (F.S.W.); (N.K.)
| | - Fanny Sae Wang
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (K.B.); (D.S.); (K.J.); (F.S.W.); (N.K.)
| | - Arun Huntrup
- Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Jarunee Prasertsopon
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand; (H.L.); (J.P.)
| | - Nathamon Kosoltanapiwat
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (K.B.); (D.S.); (K.J.); (F.S.W.); (N.K.)
| | - Pilaipan Puthavathana
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand; (H.L.); (J.P.)
| |
Collapse
|
14
|
Jeong JH, Choi WS, Antigua KJC, Choi YK, Govorkova EA, Webby RJ, Baek YH, Song MS. In Vitro Profiling of Laninamivir-Resistant Substitutions in N3 to N9 Avian Influenza Virus Neuraminidase Subtypes and Their Association with In Vivo Susceptibility. J Virol 2020; 95:e01679-20. [PMID: 33055248 PMCID: PMC7737746 DOI: 10.1128/jvi.01679-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/05/2020] [Indexed: 11/20/2022] Open
Abstract
Laninamivir (LAN) is a long-acting neuraminidase (NA) inhibitor (NAI) with a similar binding profile in the influenza NA enzyme active site as those of other NAIs, oseltamivir (OS), zanamivir (ZAN), and peramivir, and may share common resistance markers with these NAIs. We screened viruses with NA substitutions previously found during OS and ZAN selection in avian influenza viruses (AIVs) of the N3 to N9 subtypes for LAN susceptibility. Of the 72 NA substitutions, 19 conferred resistance to LAN, which ranged from 11.2- to 549.8-fold-decreased inhibitory activity over that of their parental viruses. Ten NA substitutions reduced the susceptibility to all four NAIs, whereas the remaining 26 substitutions yielded susceptibility to one or more NAIs. To determine whether the in vitro susceptibility of multi-NAI-resistant AIVs is associated with in vivo susceptibility, we infected BALB/c mice with recombinant AIVs with R292K (ma81K-N3R292K) or Q136K (ma81K-N8Q136K) NA substitutions, which impart in vitro susceptibility only to LAN or OS, respectively. Both ma81K-N3R292K and ma81K-N8Q136K virus-infected mice exhibited reduced weight loss, mortality, and lung viral titers when treated with their susceptible NAIs, confirming the in vitro susceptibility of these substitutions. Together, LAN resistance profiling of AIVs of a range of NA subtypes improves the understanding of NAI resistance mechanisms. Furthermore, the association of in vitro and in vivo NAI susceptibility indicates that our models are useful tools for monitoring NAI susceptibility of AIVs.IMPORTANCE The chemical structures of neuraminidase inhibitors (NAIs) possess similarities, but slight differences can result in variable susceptibility of avian influenza viruses (AIVs) carrying resistance-associated NA substitutions. Therefore, comprehensive susceptibility profiling of these substitutions in AIVs is critical for understanding the mechanism of antiviral resistance. In this study, we profiled resistance to the anti-influenza drug laninamivir in AIVs with substitutions known to impart resistance to other NAIs. We found 10 substitutions that conferred resistance to all four NAIs tested. On the other hand, we found that the remaining 26 NA substitutions were susceptible to at least one or more NAIs and showed for a small selection that in vitro data predicted in vivo behavior. Therefore, our findings highlight the usefulness of screening resistance markers in NA enzyme inhibition assays and animal models of AIV infections.
Collapse
Affiliation(s)
- Ju Hwan Jeong
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Republic of Korea
| | - Won-Suk Choi
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Republic of Korea
| | - Khristine Joy C Antigua
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Republic of Korea
| | - Young Ki Choi
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Republic of Korea
| | - Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yun Hee Baek
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Republic of Korea
| | - Min-Suk Song
- Department of Microbiology, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Republic of Korea
| |
Collapse
|
15
|
Gattani A, Kumar A, Agrawal A, Khan MH, Mahawar M, Bag S, Rajak KK, Singh RK, Singh P. An electrochemical impedance sensor for monitoring of gallic acid inhibited neuraminidase activity of PPR HN protein. Microchem J 2020. [DOI: 10.1016/j.microc.2020.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
16
|
Qorri B, Harless W, Szewczuk MR. Novel Molecular Mechanism of Aspirin and Celecoxib Targeting Mammalian Neuraminidase-1 Impedes Epidermal Growth Factor Receptor Signaling Axis and Induces Apoptosis in Pancreatic Cancer Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4149-4167. [PMID: 33116404 PMCID: PMC7550724 DOI: 10.2147/dddt.s264122] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
Background Aspirin (acetylsalicylic acid) and celecoxib have been used as potential anti-cancer therapies. Aspirin exerts its therapeutic effect in both cyclooxygenase (COX)-dependent and -independent pathways to reduce tumor growth and disable tumorigenesis. Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, reduces factors that cause inflammation and pain. The question is whether aspirin and celecoxib have other molecular targets of equal or more therapeutic efficacy with significant anti-cancer preventive benefits. Aim Here, we propose that aspirin and celecoxib exert their anti-cancer effects by targeting and inhibiting mammalian neuraminidase-1 (Neu-1). Neu-1 has been reported to regulate the activation of several receptor tyrosine kinases (RTKs) and TOLL-like receptors and their downstream signaling pathways. Neu-1 in complex with matrix metalloproteinase-9 (MMP-9) and G protein-coupled receptors (GPCRs) has been reported to be tethered to RTKs at the ectodomain. Materials and Methods The WST-1 cell viability assay, Caspase 3/7 assay, and Annexin V assay were used to evaluate the cell viability and detect apoptotic and necrotic cells following treatment in MiaPaCa-2, PANC-1 and the gemcitabine-resistant PANC-1 variant (PANC-1 GemR) cells. Microscopic imaging, lectin cytochemistry, and flow cytometry were used to detect levels of α-2,3 sialic acid. Epidermal growth factor (EGF)-stimulated live cell sialidase assays and neuraminidase assays were used to detect Neu-1 activity. Immunocytochemistry was used to detect levels of EGFR and phosphorylated EGFR (pEGFR) following treatment. Results For the first time, aspirin and celecoxib were shown to significantly inhibit Neu-1 sialidase activity in a dose- and time-dependent manner following stimulation with EGF. Aspirin blocked Neu-1 desialylation of α-2,3-sialic acid expression following 30 min stimulation with EGF. Aspirin and celecoxib significantly and dose-dependently inhibited isolated neuraminidase (Clostridium perfringens) activity on fluorogenic substrate 2ʹ-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MUNANA). Aspirin inhibited phosphorylation of the EGFR in EGF-stimulated cells. Aspirin dose- and time-dependently induced CellEvent caspase-3/7+ cells as well as apoptosis and necrosis on PANC-1 cells. Conclusion These findings signify a novel multimodality mechanism(s) of action for aspirin and celecoxib, specifically targeting and inhibiting Neu-1 activity, regulating EGF-induced growth receptor activation and inducing apoptosis and necrosis in a dose- and time-dependent manner. Repurposing aspirin and celecoxib as anti-cancer agents may also upend other critical targets involved in multistage tumorigenesis regulated by mammalian neuraminidase-1. Significance These findings may be the missing link connecting the anti-cancer efficacy of NSAIDs to the role of glycosylation in inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | | | - Myron R Szewczuk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
17
|
Márquez-Domínguez L, Reyes-Leyva J, Herrera-Camacho I, Santos-López G, Scior T. Five Novel Non-Sialic Acid-Like Scaffolds Inhibit In Vitro H1N1 and H5N2 Neuraminidase Activity of Influenza a Virus. Molecules 2020; 25:molecules25184248. [PMID: 32947893 PMCID: PMC7571124 DOI: 10.3390/molecules25184248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 01/04/2023] Open
Abstract
Neuraminidase (NA) of influenza viruses enables the virus to access the cell membrane. It degrades the sialic acid contained in extracellular mucin. Later, it is responsible for releasing newly formed virions from the membrane of infected cells. Both processes become key functions within the viral cycle. Therefore, it is a therapeutic target for research of the new antiviral agents. Structure–activity relationships studies have revealed which are the important functional groups for the receptor–ligand interaction. Influenza virus type A NA activity was inhibited by five scaffolds without structural resemblance to sialic acid. Intending small organic compound repositioning along with drug repurposing, this study combined in silico simulations of ligand docking into the known binding site of NA, along with in vitro bioassays. The five proposed scaffolds are N-acetylphenylalanylmethionine, propanoic 3-[(2,5-dimethylphenyl) carbamoyl]-2-(piperazin-1-yl) acid, 3-(propylaminosulfonyl)-4-chlorobenzoic acid, ascorbic acid (vitamin C), and 4-(dipropylsulfamoyl) benzoic acid (probenecid). Their half maximal inhibitory concentration (IC50) was determined through fluorometry. An acidic reagent 2′-O-(4-methylumbelliferyl)-α-dN-acetylneuraminic acid (MUNANA) was used as substrate for viruses of human influenza H1N1 or avian influenza H5N2. Inhibition was observed in millimolar ranges in a concentration-dependent manner. The IC50 values of the five proposed scaffolds ranged from 6.4 to 73 mM. The values reflect a significant affinity difference with respect to the reference drug zanamivir (p < 0.001). Two compounds (N-acetyl dipeptide and 4-substituted benzoic acid) clearly showed competitive mechanisms, whereas ascorbic acid reflected non-competitive kinetics. The five small organic molecules constitute five different scaffolds with moderate NA affinities. They are proposed as lead compounds for developing new NA inhibitors which are not analogous to sialic acid.
Collapse
Affiliation(s)
- Luis Márquez-Domínguez
- Laboratorio de Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla 74630, Mexico; (L.M.-D.); (J.R.-L.)
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico
- Laboratorio de Simulaciones Computacionales Moleculares, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico
| | - Julio Reyes-Leyva
- Laboratorio de Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla 74630, Mexico; (L.M.-D.); (J.R.-L.)
| | - Irma Herrera-Camacho
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico;
| | - Gerardo Santos-López
- Laboratorio de Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla 74630, Mexico; (L.M.-D.); (J.R.-L.)
- Correspondence: (G.S.-L.); (T.S.); Tel.: +52-244-444-0122 (G.S.-L.)
| | - Thomas Scior
- Laboratorio de Simulaciones Computacionales Moleculares, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico
- Correspondence: (G.S.-L.); (T.S.); Tel.: +52-244-444-0122 (G.S.-L.)
| |
Collapse
|
18
|
Fedorov AY, Zhirnov OP. [Method for evaluating the neuraminidase activity of receptordestroying enzyme (RDE) compounds using the influenza virus.]. Vopr Virusol 2020; 65:113-118. [PMID: 32515567 DOI: 10.36233/0507-4088-2020-65-2-113-118] [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: 03/12/2020] [Accepted: 03/31/2020] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The classic hemagglutination inhibition reaction (RTGA) is used to determine the level of antiviral antibodies in human and animal serum specimens. During the performance of RTGA the tested sera must be treated with a receptor-destroying enzyme (RDE) to remove serum glycans that degrade the accuracy of the RTGA results. To optimize the amounts of RDE compounds used, it is necessary to know their real neuraminidase activity. This article describes a simple and economical method for testing the neuraminidase activity of receptordestroying compounds using standard reagents and laboratory equipment. Aims of investigation. Design of an improved simple and convenient method for evaluating the neuramin1idase activity using the flu virus. MATERIAL AND METHODS Here, we propose a convenient method for evaluating the activity of neuraminidase by double-fold dilution procedure with human or animal erythrocytes followed by hemagglutination assay with influenza A virus. RESULTS AND DISCUSSION The method is based on the ability of neuraminidase to hydrolyze sialic acid residues on the cell surface of erythrocytes, that deprives red blood cells to be agglutinated with the flu virus, since these sialic glycans provide virus attachment and hemagglutination. CONCLUSION The designed method allows the accurate measurement of the receptor-destroying (neuraminidase) activity of RDE compounds and the comparison of the compounds with each other. This test is necessary to optimize the RTGA protocol when monitoring blood sera of animals and humans after influenza infection and/or Acute Respiratory diseases (ARD). The designed method can be included in the guidelines of regulations for the RTGA protocol, which is used in different laboratories to monitor the epidemic process of influenza and ARD infections.
Collapse
Affiliation(s)
- A Y Fedorov
- D.I. Ivanovsky Institute of Virology, National Research Centre for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russia.,I.M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - O P Zhirnov
- D.I. Ivanovsky Institute of Virology, National Research Centre for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russia.,Russian-German Academy of Medical and Biotechnological Sciences, Skolkovo Innovation Center, Moscow, 121205, Russia
| |
Collapse
|
19
|
Chen J, Feng S, Xu Y, Huang X, Zhang J, Chen J, An X, Zhang Y, Ning X. Discovery and characterization of a novel peptide inhibitor against influenza neuraminidase. RSC Med Chem 2020; 11:148-154. [PMID: 33479615 PMCID: PMC7433756 DOI: 10.1039/c9md00473d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/13/2019] [Indexed: 01/11/2023] Open
Abstract
Neuraminidase, an abundant glycoprotein on the influenza virus surface, plays crucial roles in virus replication. Targeting neuraminidase could be a splendid way for the prevention of the spread of influenza infections. Herein, we have identified an octapeptide (errKPAQP) from a synthesized peptide library, originating from mimicking the binding pocket of oseltamivir in neuraminidase, as a potent peptide neuraminidase inhibitor. The docking-based virtual studies showed that errKPAQP exhibited a strong binding affinity (a docking score of -20.03) and nanomolar affinity (11 nM) to influenza neuraminidase, and can inhibit neuraminidase activity at a concentration as low as 4.25 μM, leading to effective protection of MDCK cells from influenza virus-induced death and replication. Furthermore, errKPAQP presented low hemolytic activity, minimal cytotoxicity, and good pharmacokinetic characteristics, which are imperative for an anti-influenza drug. Importantly, errKPAQP was capable of reducing influenza virus-induced inflammation, the serious damage to the lung tissues, and mortality rates in infected mice, indicating that it could protect against the lethal challenge of influenza viruses in vivo. Therefore, we have developed a novel neuraminidase peptide inhibitor with advantageous biological properties and high inhibitory activity towards neuraminidase, and it can serve as a promising anti-influenza drug.
Collapse
Affiliation(s)
- Jianmei Chen
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| | - Shujun Feng
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| | - Yurui Xu
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| | - Xinyu Huang
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| | - Jikang Zhang
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| | - Jiao Chen
- Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , Jiangsu 210028 , China
| | - Xueying An
- State Key Laboratory of Pharmaceutical Biotechnology , Department of Sports Medicine and Adult Reconstructive Surgery , Nanjing Drum Tower Hospital , The Affiliated Hospital of Nanjing University Medical School , 321 Zhongshan Road , Nanjing 210008 , Jiangsu , PR China
| | - Yu Zhang
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| | - Xinghai Ning
- Department of Biomedical Engineering , Nanjing National Laboratory of Microstructures , College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China . ;
- Chemistry and Biomedicine Innovation Center , Nanjing University , China
| |
Collapse
|
20
|
Suttie A, Tok S, Yann S, Keo P, Horm SV, Roe M, Kaye M, Sorn S, Holl D, Tum S, Barr IG, Hurt AC, Greenhill AR, Karlsson EA, Vijaykrishna D, Deng YM, Dussart P, Horwood PF. The evolution and genetic diversity of avian influenza A(H9N2) viruses in Cambodia, 2015 - 2016. PLoS One 2019; 14:e0225428. [PMID: 31815945 PMCID: PMC6901181 DOI: 10.1371/journal.pone.0225428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/04/2019] [Indexed: 11/18/2022] Open
Abstract
Low pathogenic A(H9N2) subtype avian influenza viruses (AIVs) were originally detected in Cambodian poultry in 2013, and now circulate endemically. We sequenced and characterised 64 A(H9N2) AIVs detected in Cambodian poultry (chickens and ducks) from January 2015 to May 2016. All A(H9) viruses collected in 2015 and 2016 belonged to a new BJ/94-like h9-4.2.5 sub-lineage that emerged in the region during or after 2013, and was distinct to previously detected Cambodian viruses. Overall, there was a reduction of genetic diversity of H9N2 since 2013, however two genotypes were detected in circulation, P and V, with extensive reassortment between the viruses. Phylogenetic analysis showed a close relationship between A(H9N2) AIVs detected in Cambodian and Vietnamese poultry, highlighting cross-border trade/movement of live, domestic poultry between the countries. Wild birds may also play a role in A(H9N2) transmission in the region. Some genes of the Cambodian isolates frequently clustered with zoonotic A(H7N9), A(H9N2) and A(H10N8) viruses, suggesting a common ecology. Molecular analysis showed 100% of viruses contained the hemagglutinin (HA) Q226L substitution, which favours mammalian receptor type binding. All viruses were susceptible to the neuraminidase inhibitor antivirals; however, 41% contained the matrix (M2) S31N substitution associated with resistance to adamantanes. Overall, Cambodian A(H9N2) viruses possessed factors known to increase zoonotic potential, and therefore their evolution should be continually monitored.
Collapse
Affiliation(s)
- Annika Suttie
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
- School of Health and Life Sciences, Federation University, Churchill, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Songha Tok
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Sokhoun Yann
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Ponnarath Keo
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Srey Viseth Horm
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Merryn Roe
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Matthew Kaye
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - San Sorn
- National Animal Health and Production Research Institute, General Directorate of Animal Health and Production, Cambodian Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Davun Holl
- National Animal Health and Production Research Institute, General Directorate of Animal Health and Production, Cambodian Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Sothyra Tum
- National Animal Health and Production Research Institute, General Directorate of Animal Health and Production, Cambodian Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Ian G. Barr
- School of Health and Life Sciences, Federation University, Churchill, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Aeron C. Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Andrew R. Greenhill
- School of Health and Life Sciences, Federation University, Churchill, Australia
| | - Erik A. Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Dhanasekaran Vijaykrishna
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria Australia
| | - Yi-Mo Deng
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
- * E-mail: (PH); (PD)
| | - Paul F. Horwood
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia
- * E-mail: (PH); (PD)
| |
Collapse
|
21
|
Lai CC, Cheng YC, Chen PW, Lin TH, Tzeng TT, Lu CC, Lee MS, Hu AYC. Process development for pandemic influenza VLP vaccine production using a baculovirus expression system. J Biol Eng 2019; 13:78. [PMID: 31666806 PMCID: PMC6813129 DOI: 10.1186/s13036-019-0206-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
Background Influenza viruses cause hundreds of thousands of respiratory diseases worldwide each year, and vaccination is considered the most effective approach for preventing influenza annual epidemics or pandemics. Since 1950, chicken embryonated eggs have been used as the main method for producing seasonal influenza vaccines. However, this platform has the main drawback of a lack of scale-up flexibility, and thus, egg-based vaccine manufacturers cannot supply sufficient doses within a short period for use for pandemic prevention. As a result, strategies for reducing the manufacturing time and increasing production capacity are urgently needed. Non-virion vaccine methods have been considered an alternative strategy against an influenza pandemic, and the purpose of maintaining an immunogenic capsule structure with infectious properties appears to be met by the virus-like particle (VLP) platform. Results An influenza H7N9-TW VLP production platform using insect cells, which included the expression of hemagglutinin (HA), NA, and M1 proteins, was established. To scale up H7N9-TW VLP production, several culture conditions were optimized to obtain a higher production yield. A high level of dissolved oxygen (DO) could be critical to H7N9-TW VLP production. If the DO was maintained at a high level, the HA titer obtained in the spinner flask system with ventilation was similar to that obtained in a shake flask. In this study, the HA titer in a 5-L bioreactor with a well-controlled DO level was substantially improved by 128-fold (from 4 HA units (HAU)/50 μL to 512 HAU/50 μL). Conclusions In this study, a multigene expression platform and an effective upstream process were developed. Notably, a high H7N9-TW VLP yield was achieved using a two-step production strategy while a high DO level was maintained. The upstream process, which resulted in high VLP titers, could be further used for large-scale influenza VLP vaccine production.
Collapse
Affiliation(s)
- Chia-Chun Lai
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan.,2College of Life Science, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Yu-Chieh Cheng
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Pin-Wen Chen
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Ting-Hui Lin
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan.,2College of Life Science, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Tsai-Teng Tzeng
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Chia-Chun Lu
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Min-Shi Lee
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Alan Yung-Chih Hu
- National Institute of Infectious Diseases and Vaccinology, NHRI, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| |
Collapse
|
22
|
Horwood PF, Karlsson EA, Horm SV, Ly S, Heng S, Chin S, Darapheak C, Saunders D, Chanthap L, Rith S, Y P, Chea KL, Sar B, Parry A, Ieng V, Tsuyouka R, Deng YM, Hurt AC, Barr IG, Komadina N, Buchy P, Dussart P. Circulation and characterization of seasonal influenza viruses in Cambodia, 2012-2015. Influenza Other Respir Viruses 2019; 13:465-476. [PMID: 31251478 PMCID: PMC6692578 DOI: 10.1111/irv.12647] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 12/13/2018] [Accepted: 04/26/2019] [Indexed: 12/04/2022] Open
Abstract
Background Influenza virus circulation is monitored through the Cambodian influenza‐like illness (ILI) sentinel surveillance system and isolates are characterized by the National Influenza Centre (NIC). Seasonal influenza circulation has previously been characterized by year‐round activity and a peak during the rainy season (June‐November). Objectives We documented the circulation of seasonal influenza in Cambodia for 2012‐2015 and investigated genetic, antigenic, and antiviral resistance characteristics of influenza isolates. Patients/Methods Respiratory samples were collected from patients presenting with influenza‐like illness (ILI) at 11 hospitals throughout Cambodia. First‐line screening was conducted by the National Institute of Public Health and the Armed Forces Research Institute of Medical Sciences. Confirmation of testing and genetic, antigenic and antiviral resistance characterization was conducted by Institute Pasteur in Cambodia, the NIC. Additional virus characterization was conducted by the WHO Collaborating Centre for Reference and Research on Influenza (Melbourne, Australia). Results Between 2012 and 2015, 1,238 influenza‐positive samples were submitted to the NIC. Influenza A(H3N2) (55.3%) was the dominant subtype, followed by influenza B (30.9%; predominantly B/Yamagata‐lineage) and A(H1N1)pdm09 (13.9%). Circulation of influenza viruses began earlier in 2014 and 2015 than previously described, coincident with the emergence of A(H3N2) clades 3C.2a and 3C.3a, respectively. There was high diversity in the antigenicity of A(H3N2) viruses, and to a smaller extent influenza B viruses, during this period, with some mismatches with the northern and southern hemisphere vaccine formulations. All isolates tested were susceptible to the influenza antiviral drugs oseltamivir and zanamivir. Conclusions Seasonal and year‐round co‐circulation of multiple influenza types/subtypes were detected in Cambodia during 2012‐2015.
Collapse
Affiliation(s)
- Paul F Horwood
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia.,Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Erik A Karlsson
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia
| | - Srey Viseth Horm
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia
| | - Sovann Ly
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | - Seng Heng
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | - Savuth Chin
- National Institute of Public Health, Phnom Penh, Cambodia
| | - Chau Darapheak
- National Institute of Public Health, Phnom Penh, Cambodia
| | - David Saunders
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Lon Chanthap
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sareth Rith
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia
| | - Phalla Y
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia
| | - Kim Lay Chea
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia
| | - Borann Sar
- Centers for Disease Control and Prevention, Phnom Penh, Cambodia
| | - Amy Parry
- World Health Organization, Phnom Penh, Cambodia
| | - Vanra Ieng
- World Health Organization, Phnom Penh, Cambodia
| | | | - Yi-Mo Deng
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, Peter Doherty Institute, Melbourne, Victoria, Australia
| | - Aeron C Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, Peter Doherty Institute, Melbourne, Victoria, Australia
| | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, Peter Doherty Institute, Melbourne, Victoria, Australia
| | - Naomi Komadina
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, Peter Doherty Institute, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | - Philippe Buchy
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia.,GlaxoSmithKline Vaccines R&D Intercontinental, Singapore, Singapore
| | - Philippe Dussart
- Virology Unit, Institute Pasteur in Cambodia, Institute Pasteur International Network, Phnom Penh, Cambodia
| |
Collapse
|
23
|
Lvov DK, Burtseva EI, Mukasheva EA, Kolobukhina LV, Trushakova SV, Breslav NV, Feodoritova EL, Merkulova LN, Krasnoslobotsev KG, Morozova EO, Fedyakina IT, Aristova AV, Vartanian RV, Kisteneva LB, Prilipov AG, Alkhovsky SV, Rosatkevich AG, Kruzhkova IS, Belyaev AL, Axselrod EV, Bazarova MV, Smetanina SV. The Activity of Influenza Viruses during 2017-2018 Season in Russia and Countries of the Northern Hemisphere: Conflict by the B-virus Vaccine Component. ACTA ACUST UNITED AC 2019. [DOI: 10.31631/2073-3046-2019-18-3-13-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- D. K. Lvov
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - E. I. Burtseva
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - E. A. Mukasheva
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - L. V. Kolobukhina
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - S. V. Trushakova
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - N. V. Breslav
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - E. L. Feodoritova
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - L. N. Merkulova
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - K. G. Krasnoslobotsev
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - E. O. Morozova
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - I. T. Fedyakina
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - A. V. Aristova
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - R. V. Vartanian
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - L. B. Kisteneva
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - A. G. Prilipov
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - S. V. Alkhovsky
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - A. G. Rosatkevich
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - I. S. Kruzhkova
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - A. L. Belyaev
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - E. V. Axselrod
- D.I. Ivanovsky Institute of Virology of National Research Center of Epidemiology and Microbiology named after honorary academician N.F. Gamaleya of Ministry of Healthcare Russia
| | - M. V. Bazarova
- Clinical Hospital for Infectious Diseases № 1 Department of Healthcare of Moscow
| | - S. V. Smetanina
- Clinical Hospital for Infectious Diseases № 1 Department of Healthcare of Moscow
| |
Collapse
|
24
|
Baculovirus as an efficient vector for gene delivery into mosquitoes. Sci Rep 2018; 8:17778. [PMID: 30542209 PMCID: PMC6290771 DOI: 10.1038/s41598-018-35463-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 11/05/2018] [Indexed: 02/06/2023] Open
Abstract
Efficient gene delivery technologies play an essential role in the gene functional analyses that are necessary for basic and applied researches. Mosquitoes are ubiquitous insects, responsible for transmitting many deadly arboviruses causing millions of human deaths every year. The lack of efficient and flexible gene delivery strategies in mosquitoes are among the major hurdles for the study of mosquito biology and mosquito-pathogen interactions. We found that Autographa californica multiple nucleopolyhedrovirus (AcMNPV), the type baculovirus species, can efficiently transduce mosquito cells without viral propagation, allowing high level gene expression upon inducement by suitable promoters without obvious negative effects on cell propagation and viability. AcMNPV transduces into several mosquito cell types, efficiently than in commonly used mammalian cell lines and classical plasmid DNA transfection approaches. We demonstrated the application of this system by expressing influenza virus neuraminidase (NA) into mosquito hosts. Moreover, AcMNPV can transduce both larvae and adults of essentially all blood-sucking mosquito genera, resulting in bright fluorescence in insect bodies with little or no tissue barriers. Our experiments establish baculovirus as a convenient and powerful gene delivery vector in vitro and in vivo that will greatly benefit research into mosquito gene regulation, development and the study of mosquito-borne viruses.
Collapse
|
25
|
Tsedenbal N, Tsend-Ayush A, Badarch D, Jav S, Pagbajab N. Influenza B viruses circulated during last 5 years in Mongolia. PLoS One 2018; 13:e0206987. [PMID: 30439983 PMCID: PMC6237300 DOI: 10.1371/journal.pone.0206987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022] Open
Abstract
Influenza B virus-caused illness has recently been considered as an urgent public health problem due to substantial morbidity, mortality and life-threatening medical complications. In this study, we have reported the main characteristics of influenza B virus in Mongolia, including prevalence, lineages, suitability with vaccine strains and drug susceptibility against the virus. 15768 specimens were tested by qPCR for detecting influenza viruses. From positive specimens for influenza B virus, the clinical isolates were isolated using MDCK cells. Sequencing analysis, hemagglutination inhibition assay and Neuraminidase inhibitor (NAI) drug susceptibility testing were performed for the clinical isolates. Influenza B virus was around in 3.46% of the samples in Mongolia, and B/Victoria clade-1A and B/Yamagata clade-3 lineages were predominant. Importantly, it was confirmed that the lineages corresponded to the vaccine strains. Moreover, drug susceptibility tests revealed that some Mongolian clinical isolates showed reduced susceptibility to antiviral agents. Interestingly, G104R was identified as a novel mutation, which might have a significant role in drug resistance of the virus. These results describe the characteristics of influenza B viruses that have caused respiratory illness in the population of Mongolia between 2013 and 2017.
Collapse
Affiliation(s)
- Naranzul Tsedenbal
- National Influenza Center, National Center for Communicable Diseases, Ministry of Health, Ulaanbaatar, Mongolia
| | - Altansukh Tsend-Ayush
- School of Bio-Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Darmaa Badarch
- National Influenza Center, National Center for Communicable Diseases, Ministry of Health, Ulaanbaatar, Mongolia
| | - Sarantuya Jav
- School of Bio-Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
- * E-mail:
| | - Nymadawa Pagbajab
- National Influenza Center, National Center for Communicable Diseases, Ministry of Health, Ulaanbaatar, Mongolia
- Mongolian Academy of Medical Sciences, Ulaanbaatar, Mongolia
| |
Collapse
|
26
|
Isolation and characterization of H5Nx highly pathogenic avian influenza viruses of clade 2.3.4.4 in Russia. Virology 2018; 525:216-223. [PMID: 30296682 DOI: 10.1016/j.virol.2018.09.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/28/2018] [Accepted: 09/28/2018] [Indexed: 11/23/2022]
Abstract
In 2016-2017, several subtypes of the highly pathogenic avian influenza (HPAI) virus were isolated on the territory of Russia. In the autumn of 2016, during the avian influenza virus surveillance in the territory of the Kamchatka region of Russia the HPAI A(H5N5) influenza virus was isolated. Then, during 2016-2017, multiple outbreaks among wild birds and poultry caused by HPAI A(H5N8) avian influenza virus were recorded in European part of Russia. At the end of 2017, an outbreak among poultry caused by HPAI A(H5N2) influenza virus was recorded in the European part of Russia. Phylogenetic analysis of HA of the A(H5N5), A(H5N8), A(H5N2) showed the strains belong to the clade 2.3.4.4 b. All isolated strains were antigenically closely related to candidate vaccine viruses of clade 2.3.4.4 and showed high virulence in mice. Genetic analysis revealed presence of genetic markers potentially related to high virulence in mice in all studied viruses.
Collapse
|