1
|
Chakraborty S, Chauhan A. Fighting the flu: a brief review on anti-influenza agents. Biotechnol Genet Eng Rev 2024; 40:858-909. [PMID: 36946567 DOI: 10.1080/02648725.2023.2191081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
The influenza virus causes one of the most prevalent and lethal infectious viral diseases of the respiratory system; the disease progression varies from acute self-limiting mild fever to disease chronicity and death. Although both the preventive and treatment measures have been vital in protecting humans against seasonal epidemics or sporadic pandemics, there are several challenges to curb the influenza virus such as limited or poor cross-protection against circulating virus strains, moderate protection in immune-compromised patients, and rapid emergence of resistance. Currently, there are four US-FDA-approved anti-influenza drugs to treat flu infection, viz. Rapivab, Relenza, Tamiflu, and Xofluza. These drugs are classified based on their mode of action against the viral replication cycle with the first three being Neuraminidase inhibitors, and the fourth one targeting the viral polymerase. The emergence of the drug-resistant strains of influenza, however, underscores the need for continuous innovation towards development and discovery of new anti-influenza agents with enhanced antiviral effects, greater safety, and improved tolerability. Here in this review, we highlighted commercially available antiviral agents besides those that are at different stages of development including under clinical trials, with a brief account of their antiviral mechanisms.
Collapse
Affiliation(s)
| | - Ashwini Chauhan
- Department of Microbiology, Tripura University, Agartala, India
| |
Collapse
|
2
|
Ahmad M, Ahmed I, Akhtar T, Amir M, Parveen S, Narayan E, Iqbal H, Rehman SU. Strategies and innovations for combatting diseases in animals (Review). WORLD ACADEMY OF SCIENCES JOURNAL 2024; 6:55. [DOI: 10.3892/wasj.2024.270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Affiliation(s)
- Muhammad Ahmad
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Ishtiaq Ahmed
- La Trobe Rural Health School, Albury‑Wodonga Campus, La Trobe University, Wodonga, Victoria 3690, Australia
| | - Tayyaba Akhtar
- Department of Epidemiology and Public Health, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Muhammad Amir
- School of Health and Society, Faculty of Arts, Social Sciences and Humanities, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Shakeela Parveen
- Department of Zoology, Government Sadiq College Women University, Bahawalpur 63100, Pakistan
| | - Edward Narayan
- School of Agriculture and Food Sustainability, The University of Queensland, Gatton, Queensland 4343, Australia
| | - Hafiz Iqbal
- Facultad de Agronomía, Campus Ciencias Agropecuarias, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, C.P. 66050, Mexico
| | - Saif Ur Rehman
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
3
|
Bialy D, Richardson S, Chrzastek K, Bhat S, Polo N, Freimanis G, Iqbal M, Shelton H. Recombinant A(H6N1)-H274Y avian influenza virus with dual drug resistance does not require permissive mutations to retain the replicative fitness in vitro and in ovo. Virology 2024; 590:109954. [PMID: 38086284 DOI: 10.1016/j.virol.2023.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024]
Abstract
The possible emergence of drug-resistant avian flu raises concerns over the limited effectiveness of currently approved antivirals (neuraminidase inhibitors - NAIs) in the hypothetical event of a zoonotic spillover. Our study demonstrated that the recombinant avian A(H6N1) viruses showed reduced inhibition (RI) by multiple NAI drugs following the introduction of point mutations found predominantly in the neuraminidase gene (NA) of NAI-resistant human influenza strains (E119V, R292K and H274Y; N2 numbering). Moreover, A(H6N1)-H274Y showed increased replication efficiency in vitro, and a fitness advantage over wild-type (WT) when co-inoculated into embryonated hen's eggs. The results presented in our study together with the zoonotic potential of the A(H6N1) virus as evidenced by the human infection from 2013, highlight the need for enhanced monitoring of NAI resistance-associated signatures in circulating LPAI (low pathogenic avian influenza) globally.
Collapse
Affiliation(s)
- Dagmara Bialy
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom.
| | - Samuel Richardson
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| | - Klaudia Chrzastek
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| | - Sushant Bhat
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| | - Noemi Polo
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| | - Graham Freimanis
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| | - Munir Iqbal
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| | - Holly Shelton
- The Pirbright Institute, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom
| |
Collapse
|
4
|
Li Y, Liu HY, Yang MJ, Liu D, Song JQ, Lao Z, Chen Y, Yang Y. Preparation of eicosavalent triazolylsialoside-conjugated human serum albumin as a dual hemagglutinin/neuraminidase inhibitor and virion adsorbent for the prevention of influenza infection. Carbohydr Res 2023; 532:108918. [PMID: 37586142 DOI: 10.1016/j.carres.2023.108918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
A triazolylsialoside-human serum albumin conjugate was prepared as a multivalent hemagglutinin and neuraminidase inhibitor using a di-(N-succinimidyl) adipate strategy. Matrix-Assisted Laser Desorption/Ionization-Time of Flight-Mass Spectrometry (MALDI-TOF-MS) indicated that five tetravalent sialyl galactosides were grafted onto the protein backbone resulting in an eicosavalent triazolylsialoside-protein complex. Compared with monomeric sialic acid, molecular interaction studies showed that the synthetic pseudo-glycoprotein bound tightly not only to hemagglutinin (HA)/neuraminidase (NA) but also to mutated drug-resistant NA on the surface of the influenza virus with a dissociation constant (KD) in the 1 μM range, attributed to the cluster effect. Moreover, this glycoconjugate exhibited potent antiviral activity against a broad spectrum of virus strains and showed no cytotoxicity towards Human Umbilical Vein Endothelial Cells (HUVECs) and Madin-Darby canine kidney (MDCK) cells at high concentrations. Further mechanistic studies demonstrated this multivalent sialyl conjugate showed strong capture and trapping of influenza virions, thus disrupting the ability of the influenza virus to infect host cells. This research lays the experimental foundation for the development of new antiviral agents based on multivalent sialic acid-protein conjugates.
Collapse
Affiliation(s)
- Yang Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Han-Yu Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Ming-Jiang Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Dong Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Jia-Qi Song
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Zhiqi Lao
- Department of Medical Laboratory, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China.
| | - Yue Chen
- Department of Medical Laboratory, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, China.
| | - Yang Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China.
| |
Collapse
|
5
|
Kiso M, Furusawa Y, Uraki R, Imai M, Yamayoshi S, Kawaoka Y. In vitro and in vivo characterization of SARS-CoV-2 strains resistant to nirmatrelvir. Nat Commun 2023; 14:3952. [PMID: 37402789 PMCID: PMC10319741 DOI: 10.1038/s41467-023-39704-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023] Open
Abstract
Nirmatrelvir, an oral antiviral agent that targets a SARS-CoV-2 main protease (3CLpro), is clinically useful against infection with SARS-CoV-2 including its omicron variants. Since most omicron subvariants have reduced sensitivity to many monoclonal antibody therapies, potential SARS-CoV-2 resistance to nirmatrelvir is a major public health concern. Several amino acid substitutions have been identified as being responsible for reduced susceptibility to nirmatrelvir. Among them, we selected L50F/E166V and L50F/E166A/L167F in the 3CLpro because these combinations of substitutions are unlikely to affect virus fitness. We prepared and characterized delta variants possessing Nsp5-L50F/E166V and Nsp5-L50F/E166A/L167F. Both mutant viruses showed decreased susceptibility to nirmatrelvir and their growth in VeroE6/TMPRSS2 cells was delayed. Both mutant viruses showed attenuated phenotypes in a male hamster infection model, maintained airborne transmissibility, and were outcompeted by wild-type virus in co-infection experiments in the absence of nirmatrelvir, but less so in the presence of the drug. These results suggest that viruses possessing Nsp5-L50F/E166V and Nsp5-L50F/E166A/L167F do not become dominant in nature. However, it is important to closely monitor the emergence of nirmatrelvir-resistant SARS-CoV-2 variants because resistant viruses with additional compensatory mutations could emerge, outcompete the wild-type virus, and become dominant.
Collapse
Affiliation(s)
- Maki Kiso
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yuri Furusawa
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Ryuta Uraki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, Tokyo, Japan.
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, USA.
| |
Collapse
|
6
|
Farah EM, Amine S, Ahmad S, Nonlaopon K, Allali K. Theoretical and numerical results of a stochastic model describing resistance and non-resistance strains of influenza. EUROPEAN PHYSICAL JOURNAL PLUS 2022; 137:1169. [PMID: 36310610 PMCID: PMC9589591 DOI: 10.1140/epjp/s13360-022-03302-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/18/2022] [Indexed: 06/16/2023]
Abstract
In this world, there are several acute viral infections. One of them is influenza, a respiratory disease caused by the influenza virus. Stochastic modelling of infectious diseases is now a popular topic in the current century. Several stochastic epidemiological models have been constructed in the research papers. In the present article, we offer a stochastic two-strain influenza epidemic model that includes both resistant and non-resistance strains. We demonstrate both the existence and uniqueness of the global positive solution using the stochastic Lyapunov function theory. The extinction of our research sickness results from favourable circumstances. Additionally, the infection's persistence in the mean is demonstrated. Finally, to demonstrate how well our theoretical analysis performs, various noise disturbances are simulated numerically.
Collapse
Affiliation(s)
- El Mehdi Farah
- Laboratory of Mathematics , Computer Science and Applications, Faculty of Sciences and Technologies, Hassan II University of Casablanca, PO Box 146, 20650 Mohammedia, Morocco
| | - Saida Amine
- Laboratory of Mathematics , Computer Science and Applications, Faculty of Sciences and Technologies, Hassan II University of Casablanca, PO Box 146, 20650 Mohammedia, Morocco
| | - Shabir Ahmad
- Department of Mathematics, University of Malakand, Chakdara, Dir Lower, Khyber Pakhtunkhwa Pakistan
| | - Kamsing Nonlaopon
- Present Address: Department of Mathematics, Faculty of Science, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Karam Allali
- Laboratory of Mathematics , Computer Science and Applications, Faculty of Sciences and Technologies, Hassan II University of Casablanca, PO Box 146, 20650 Mohammedia, Morocco
| |
Collapse
|
7
|
Ju H, Hou L, Zhao F, Zhang Y, Jia R, Guizzo L, Bonomini A, Zhang J, Gao Z, Liang R, Bertagnin C, Kong X, Ma X, Kang D, Loregian A, Huang B, Liu X, Zhan P. Iterative Optimization and Structure-Activity Relationship Studies of Oseltamivir Amino Derivatives as Potent and Selective Neuraminidase Inhibitors via Targeting 150-Cavity. J Med Chem 2022; 65:11550-11573. [PMID: 35939763 DOI: 10.1021/acs.jmedchem.1c01970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With our continuous endeavors in seeking neuraminidase (NA) inhibitors, we reported herein three series of novel oseltamivir amino derivatives with the goal of exploring the druggable chemical space inside the 150-cavity of influenza virus NAs. Among them, around half of the compounds in series C were demonstrated to be better inhibitors against both wild-type and oseltamivir-resistant group-1 NAs than oseltamivir carboxylate (OSC). Notably, compounds 12d, 12e, 15e, and 15i showed more potent or equipotent antiviral activity against H1N1, H5N1, and H5N8 viruses compared to OSC in cellular assays. Furthermore, compounds 12e and 15e exhibited high metabolic stability in human liver microsomes (HLMs) and low inhibitory effect on main cytochrome P450 (CYP) enzymes, as well as low acute/subacute toxicity and certain antiviral efficacy in vivo. Also, pharmacokinetic (PK) and molecular docking studies were performed. Overall, 12e and 15e possess great potential to serve as anti-influenza candidates and are worthy of further investigation.
Collapse
Affiliation(s)
- Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Lingxin Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ying Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ruifang Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Laura Guizzo
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Anna Bonomini
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ruipeng Liang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Xiujie Kong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Xiuli Ma
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 202 North Gongye Road, 250100 Jinan, Shandong, P. R. China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Bing Huang
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 202 North Gongye Road, 250100 Jinan, Shandong, P. R. China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| |
Collapse
|
8
|
Predicting Permissive Mutations That Improve the Fitness of A(H1N1)pdm09 Viruses Bearing the H275Y Neuraminidase Substitution. J Virol 2022; 96:e0091822. [PMID: 35867563 PMCID: PMC9364793 DOI: 10.1128/jvi.00918-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Oseltamivir-resistant influenza viruses arise due to amino acid mutations in key residues of the viral neuraminidase (NA). These changes often come at a fitness cost; however, it is known that permissive mutations in the viral NA can overcome this cost. This result was observed in former seasonal A(H1N1) viruses in 2007 which expressed the H275Y substitution (N1 numbering) with no apparent fitness cost and lead to widespread oseltamivir resistance. Therefore, this study aims to predict permissive mutations that may similarly enable fit H275Y variants to arise in currently circulating A(H1N1)pdm09 viruses. The first approach in this study utilized in silico analyses to predict potentially permissive mutations. The second approach involved the generation of a virus library which encompassed all possible NA mutations while keeping H275Y fixed. Fit variants were then selected by serially passaging the virus library either through ferrets by transmission or passaging once in vitro. The fitness impact of selected substitutions was further evaluated experimentally. The computational approach predicted three candidate permissive NA mutations which, in combination with each other, restored the replicative fitness of an H275Y variant. The second approach identified a stringent bottleneck during transmission between ferrets; however, three further substitutions were identified which may improve transmissibility. A comparison of fit H275Y variants in vitro and in experimentally infected animals showed a statistically significant correlation in the variants that were positively selected. Overall, this study provides valuable tools and insights into potential permissive mutations that may facilitate the emergence of a fit H275Y A(H1N1)pdm09 variant. IMPORTANCE Oseltamivir (Tamiflu) is the most widely used antiviral for the treatment of influenza infections. Therefore, resistance to oseltamivir is a public health concern. This study is important as it explores the different evolutionary pathways available to current circulating influenza viruses that may lead to widespread oseltamivir resistance. Specifically, this study develops valuable experimental and computational tools to evaluate the fitness landscape of circulating A(H1N1)pmd09 influenza viruses bearing the H275Y mutation. The H275Y substitution is most commonly reported to confer oseltamivir resistance but also leads to loss of virus replication and transmission fitness, which limits its spread. However, it is known from previous influenza seasons that influenza viruses can evolve to overcome this loss of fitness. Therefore, this study aims to prospectively predict how contemporary A(H1N1)pmd09 influenza viruses may evolve to overcome the fitness cost of bearing the H275Y NA substitution, which could result in widespread oseltamivir resistance.
Collapse
|
9
|
Fast and Efficient Synthesis of Racemic Baloxavir Catalyzed by Strong Solid Acid under Microwave Conditions. CRYSTALS 2022. [DOI: 10.3390/cryst12070891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The compound (±)-12aR-12-[(11S)-7,8-difluoro-6,11-dihydrodibenzo[b,e]thiepin-11-yl]-7-hydroxy-3,4,12,12a-tetrahydro-1H-[1,4]oxazino[3,4-c]pyrido[2,1-f][1,2,4]triazine-6,8-dione is the intermediate of baloxavir marboxil. In the literature, traditional heating methods and common acid catalysts are used, which result in long reaction times and a low yield. Therefore, finding an efficient and environmentally friendly synthetic route is necessary. In this study, (±)-12aR-12-[(11S)-7,8-difluoro-6,11-dihydrodibenzo[b,e]thiepin-11-yl]-7-benzyloxy-3,4,12,12a-tetrahydro-1h-[1,4]oxazino[3,4-c]pyrido[2,1-f][1,2,4]triazine-6,8-dione (compound 3) was synthesized using a sulfonate resin solid acid catalyst (HND-580) under microwave conditions. The benzyl group was removed without further purification, and an intermediate, racemic baloxavir, was obtained under microwave irradiation. The total yield of the two steps was 78%. This method greatly reduces the reaction time and improves production efficiency.
Collapse
|
10
|
Identification of a permissive secondary mutation that restores the enzymatic activity of oseltamivir resistance mutation H275Y. J Virol 2022; 96:e0198221. [PMID: 35045267 DOI: 10.1128/jvi.01982-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many oseltamivir resistance mutations exhibit fitness defects in the absence of drug pressure that hinders their propagation in hosts. Secondary permissive mutations can rescue fitness defects and facilitate the segregation of resistance mutations in viral populations. Previous studies have identified a panel of permissive or compensatory mutations in neuraminidase (NA) that restore the growth defect of the predominant oseltamivir resistance mutation (H275Y) in H1N1 influenza A. In prior work, we identified a hyperactive mutation (Y276F) that increased NA activity by approximately 70%. While Y276F had not been previously identified as a permissive mutation, we hypothesized that Y276F may counteract the defects caused by H275Y by buffering its reduced NA expression and enzyme activity. In this study we measured the relative fitness, NA activity, and surface expression, as well as sensitivity to oseltamivir, for several oseltamivir resistance mutations including H275Y in the wildtype or Y276F genetic background. Our results demonstrate that Y276F selectively rescues the fitness defect of H275Y by restoring its NA surface expression and enzymatic activity, elucidating the local compensatory structural impacts of Y276F on the adjacent H275Y. Importance The potential for influenza A virus (IAV) to cause pandemics makes understanding evolutionary mechanisms that impact drug resistance critical for developing surveillance and treatment strategies. Oseltamivir is the most widely used therapeutic strategy to treat IAV infections, but mutations in IAV can lead to drug resistance. The main oseltamivir resistance mutation, H275Y, occurs in the neuraminidase (NA) protein of IAV and reduces drug binding as well as NA function. Here, we identify a new helper mutation, Y276F that can rescue the functional defects of H275Y and contribute to the evolution of drug resistance in IAV.
Collapse
|
11
|
Singh S, Verma DK, Thakur M, Tripathy S, Patel AR, Shah N, Utama GL, Srivastav PP, Benavente-Valdés JR, Chávez-González ML, Aguilar CN. Supercritical fluid extraction (SCFE) as green extraction technology for high-value metabolites of algae, its potential trends in food and human health. Food Res Int 2021; 150:110746. [PMID: 34865764 DOI: 10.1016/j.foodres.2021.110746] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
Application of high-value algal metabolites (HVAMs) in cosmetics, additives, pigments, foods and medicines are very important. These HVAMs can be obtained from the cultivation of micro- and macro-algae. These metabolites can benefit human and animal health in a physiological and nutritional manner. However, because of conventional extraction methods and their energy and the use of pollutant solvents, the availability of HVAMs from algae remains insufficient. Receiving their sustainability and environmental benefits have recently made green extraction technologies for HVAM extractions more desirable. But very little information is available about the technology of green extraction of algae from these HVAM. This review, therefore, highlights the supercritical fluid extraction (SCFE) as principal green extraction technologyand theirideal parameters for extracting HVAMs. In first, general information is provided concerning the HVAMs and their components of macro and micro origin. The review also includes a description of SCFE technology's properties, instrumentation operation, solvents used, and the merits and demerits. Moreover, there are several HVAMs associated with their numerous high-level biological activities which include high-level antioxidant, anti-inflammatory, anticancer and antimicrobial activity and have potential health-beneficial effects in humans since they are all HVAMs, such as foods and nutraceuticals. Finally, it provides future insights, obstacles, and suggestions for selecting the right technologies for extraction.
Collapse
Affiliation(s)
- Smita Singh
- Department of Nutrition and Dietetics, University Institute of Applied Health Sciences, Chandigarh University, Chandigarh 140413, Punjab, India.
| | - Deepak Kumar Verma
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| | - Mamta Thakur
- Department of Food Technology, School of Sciences, ITM University, Gwalior 474001, Madhya Pradesh, India.
| | - Soubhagya Tripathy
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Ami R Patel
- Division of Dairy Microbiology, Mansinhbhai Institute of Dairy and Food Technology-MIDFT, Dudhsagar Dairy Campus, Mehsana 384 002, Gujarat, India
| | - Nihir Shah
- Division of Dairy Microbiology, Mansinhbhai Institute of Dairy and Food Technology-MIDFT, Dudhsagar Dairy Campus, Mehsana 384 002, Gujarat, India
| | - Gemilang Lara Utama
- Faculty of Agro-Industrial Technology, Universitas Padjadjaran, Sumedang 45363, Indonesia; Center for Environment and Sustainability Science, Universitas Padjadjaran, Bandung 40132, Indonesia
| | - Prem Prakash Srivastav
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Juan Roberto Benavente-Valdés
- Bioprocesses and Bioproducts Research Group, Food Research Department, School of Chemistry, Autonomous University of Coahuila, Saltillo Campus, 25280 Coahuila, Mexico
| | - Mónica L Chávez-González
- Bioprocesses and Bioproducts Research Group, Food Research Department, School of Chemistry, Autonomous University of Coahuila, Saltillo Campus, 25280 Coahuila, Mexico
| | - Cristobal Noe Aguilar
- Bioprocesses and Bioproducts Research Group, Food Research Department, School of Chemistry, Autonomous University of Coahuila, Saltillo Campus, 25280 Coahuila, Mexico.
| |
Collapse
|
12
|
Jia Y, Xiang Y, Guo S, Guo L, Guo L, Cheng Z, Zhang Y, Zhang L, Long E. Analysis on the risk of respiratory virus transmission by air conditioning system operation based on experimental evidence. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:56376-56391. [PMID: 34053044 PMCID: PMC8164484 DOI: 10.1007/s11356-021-14495-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/17/2021] [Indexed: 05/03/2023]
Abstract
It is important to know whether SARS-CoV-2 is spread through the air conditioning systems. Taking the central air conditioning system as an example, we analyze the mechanism and potential health risk of respiratory virus transmission in air-conditioned rooms and propose a method to study the risk of virus transmission in central air conditioning systems by investigating the data from medical experiments. The virus carrying capacity and the decay characteristics of indoor pathogen droplets are studied in this research. Additionally, the effects of air temperature and relative humidity on the virus survival in the air or on surfaces are investigated. The removal efficiency of infectious droplet nuclei by using an air conditioning filter was then determined. Thus, the transmission risk during the operation of the centralized air conditioning system is evaluated. The results show that the indoor temperature and humidity are controlled in the range of 20-25 °C and 40-70% by central air conditioning during the epidemic period, which not only benefits the health and comfort of residents, but also weakens the vitality of the virus. The larger the droplet size, the longer the viruses survive. Since the filter efficiency of the air conditioning filter increases with the increase in particle size, increasing the number of air changes of the circulating air volume can accelerate the removal of potential pathogen particles. Therefore, scientific operation of centralized air conditioning systems during the epidemic period has more advantages than disadvantages.
Collapse
Affiliation(s)
- Yonghong Jia
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Yue Xiang
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Shurui Guo
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Lei Guo
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Luyao Guo
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Zhu Cheng
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Yin Zhang
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Solid Waste Treatment Technology, Sichuan Environmental Protection Key Laboratory of Pollution Control for Heavy Metals, Sichuan Academy of Environmental Sciences, Chengdu, China
| | - Enshen Long
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Hayden FG, Asher J, Cowling BJ, Hurt AC, Ikematsu H, Kuhlbusch K, Lemenuel-Diot A, Du Z, Meyers LA, Piedra PA, Takazono T, Yen HL, Monto AS. Reducing influenza virus transmission: the value of antiviral treatment. Clin Infect Dis 2021; 74:532-540. [PMID: 34245250 PMCID: PMC8834654 DOI: 10.1093/cid/ciab625] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Indexed: 11/16/2022] Open
Abstract
Prompt antiviral treatment has the potential to reduce influenza virus transmission to close contacts, but rigorous data on the magnitude of treatment effects on transmission are limited. Animal model data indicate that rapid reductions in viral replication after antiviral treatment reduce the risk of transmission. Observational and clinical trial data with oseltamivir and other neuraminidase inhibitors indicate that prompt treatment of household index patients seems to reduce the risk of illness in contacts, although the magnitude of the reported effects has varied widely across studies. In addition, the potential risk of transmitting drug-resistant variants exists with all approved classes of influenza antivirals. A controlled trial examining baloxavir treatment efficacy to reduce transmission, including the risk of transmitting virus with reduced baloxavir susceptibility, is currently in progress. If reduced transmission risk is confirmed, modeling studies indicate that early treatment could have major epidemiologic benefits in seasonal and pandemic influenza.
Collapse
Affiliation(s)
- Frederick G Hayden
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, USA
| | | | - Benjamin J Cowling
- School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | | | | | | - Zhanwei Du
- School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Lauren Ancel Meyers
- Department of Integrative Biology and Statistics & Data Sciences, University of Texas, Austin, Texas, USA
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, USA
| | - Takahiro Takazono
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hui-Ling Yen
- School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Arnold S Monto
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Goldhill DH, Yan A, Frise R, Zhou J, Shelley J, Gallego Cortés A, Miah S, Akinbami O, Galiano M, Zambon M, Lackenby A, Barclay WS. Favipiravir-resistant influenza A virus shows potential for transmission. PLoS Pathog 2021; 17:e1008937. [PMID: 34061908 PMCID: PMC8195362 DOI: 10.1371/journal.ppat.1008937] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 06/11/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022] Open
Abstract
Favipiravir is a nucleoside analogue which has been licensed to treat influenza in the event of a new pandemic. We previously described a favipiravir resistant influenza A virus generated by in vitro passage in presence of drug with two mutations: K229R in PB1, which conferred resistance at a cost to polymerase activity, and P653L in PA, which compensated for the cost of polymerase activity. However, the clinical relevance of these mutations is unclear as the mutations have not been found in natural isolates and it is unknown whether viruses harbouring these mutations would replicate or transmit in vivo. Here, we infected ferrets with a mix of wild type p(H1N1) 2009 and corresponding favipiravir-resistant virus and tested for replication and transmission in the absence of drug. Favipiravir-resistant virus successfully infected ferrets and was transmitted by both contact transmission and respiratory droplet routes. However, sequencing revealed the mutation that conferred resistance, K229R, decreased in frequency over time within ferrets. Modelling revealed that due to a fitness advantage for the PA P653L mutant, reassortment with the wild-type virus to gain wild-type PB1 segment in vivo resulted in the loss of the PB1 resistance mutation K229R. We demonstrated that this fitness advantage of PA P653L in the background of our starting virus A/England/195/2009 was due to a maladapted PA in first wave isolates from the 2009 pandemic. We show there is no fitness advantage of P653L in more recent pH1N1 influenza A viruses. Therefore, whilst favipiravir-resistant virus can transmit in vivo, the likelihood that the resistance mutation is retained in the absence of drug pressure may vary depending on the genetic background of the starting viral strain.
Collapse
Affiliation(s)
- Daniel H. Goldhill
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Ada Yan
- Department of Infectious Disease Epidemiology, Imperial College, London, United Kingdom
| | - Rebecca Frise
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Jie Zhou
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Jennifer Shelley
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Ana Gallego Cortés
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | | | | | | | | | | | - Wendy S. Barclay
- Department of Infectious Disease, Imperial College, London, United Kingdom
| |
Collapse
|
15
|
Differential Viral-Host Immune Interactions Associated with Oseltamivir-Resistant H275Y and Wild-Type H1N1 A(pdm09) Influenza Virus Pathogenicity. Viruses 2020; 12:v12080794. [PMID: 32721992 PMCID: PMC7472233 DOI: 10.3390/v12080794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/26/2022] Open
Abstract
Oseltamivir is a common therapy against influenza A virus (IAV) infections. The acquisition of oseltamivir resistance (OR) mutations, such as H275Y, hampers viral fitness. However, OR H1N1 viruses have demonstrated the ability to spread throughout different populations. The objective of this work was to compare the fitness of two strains of OR (R6 and R7) containing the H275Y mutation, and a wild-type (F) pandemic influenza A (H1N1) 2009 (pdm09) virus both in vitro and in vivo in mice and to select one OR strain for a comparison with F in ferrets. R6 showed faster replication and pathogenicity than R7 in vitro and in mice. Subsequently, R6 was selected for the fitness comparison with the F strain in ferrets. Ferrets infected with the F virus showed more severe clinical signs, histopathological lung lesions, and viral quantification when compared to OR R6-infected animals. More importantly, differential viral kinetics correlated with differential pro-inflammatory host immune responses in the lungs of infected ferrets, where OR-infected animals developed a protective higher expression of type I IFN and Retinoid acid Inducible Gene I (RIG-I) genes early after infection, resulting in the development of milder disease. These results suggest the presence of early specific viral-host immune interactions relevant in the development of influenza-associated lung pathology.
Collapse
|
16
|
Nandhini P, Sistla S. Genetic sequencing of influenza A (H1N1) pdm09 isolates from South India, collected between 2011 and 2015 to detect mutations affecting virulence and resistance to oseltamivir. Indian J Med Microbiol 2020; 38:324-337. [PMID: 33154243 DOI: 10.4103/ijmm.ijmm_20_83] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background Influenza A viruses evolve continuously and the two surface antigens, hemagglutinin (HA) and neuraminidase (NA) have been the target proteins for research as they are vital components in determining the virulence, immune effectiveness, pathogenicity, transmission and resistance. Methods Both HA and NA (partial genes) of 45 pandemic influenza A(H1N1)pdm09 isolates were sequenced. Phylogenetic analysis was performed with reference to representative global isolates retrieved from Influenza Virus Resource (IVR), GISAID EpiFluTM and GenBank and evolutionary analyses. Nucleotide and amino acid sequences were aligned using ClustalW/ Clustal Omega/MEGA version 6 with reference to vaccine strain (A/California/07/2009). Results All the isolates clustered along with the clade 7 virus, irrespective of the year of isolation. The study isolates exhibited 98.5% and 98.8% nucleotide homology to the reference strain A/California/07/2009(H1N1) for HA and NA, respectively. Overall, there was limited genetic diversity observed over a period of 3 years (2012-2015). Two samples collected from expired patients had D239N (D222G or D225G) mutation in HA. This mutation which is associated with dual-binding specificity of the virus has been well-correlated with severe disease outcomes. All the study isolates possessed H274 residue and 7 strains had N295S, the next most common mutation found in oseltamivir-resistant variants. Conclusion In this study, although H274Y mutation associated with oseltamivir resistance has not been noted, significant mutations have been noted in both HA and NA genes including D239N, N295S, V106I, Q136K, N248D, V267A. In both HA and NA gene analysis, multiple mutations were found more in 2015 strains when compared to 2012 strains. Hence such accumulation of mutations has to be monitored continuously to determine the efficacy of annual flu vaccines and anti-influenza drugs.
Collapse
MESH Headings
- Adult
- Aged
- Amino Acids/analysis
- Amino Acids/genetics
- Antiviral Agents/pharmacology
- Child, Preschool
- Drug Resistance, Viral/genetics
- Female
- Hemagglutinins/chemistry
- Hemagglutinins/genetics
- Humans
- India/epidemiology
- Infant
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza, Human/epidemiology
- Influenza, Human/virology
- Male
- Middle Aged
- Mutation
- Neuraminidase/chemistry
- Neuraminidase/genetics
- Oseltamivir/pharmacology
- Phylogeny
- RNA, Viral/chemistry
- RNA, Viral/isolation & purification
- Virulence
- Young Adult
Collapse
Affiliation(s)
- P Nandhini
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Sujatha Sistla
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
17
|
Low replicative fitness of neuraminidase inhibitor-resistant H7N9 avian influenza a virus with R292K substitution in neuraminidase in cynomolgus macaques compared with I222T substitution. Antiviral Res 2020; 178:104790. [PMID: 32272175 DOI: 10.1016/j.antiviral.2020.104790] [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: 10/06/2019] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 12/21/2022]
Abstract
Human cases of H7N9 influenza A virus infection have been increasing since 2013. The first choice of treatment for influenza is neuraminidase (NA) inhibitors (NAIs), but there is a concern that NAI-resistant viruses are selected in the presence of NAIs. In our previous study, an H7N9 virus carrying AA substitution of threonine (T) for isoleucine (I) at residue 222 in NA (NA222T, N2 numbering) and an H7N9 virus carrying AA substitution of lysine (K) for arginine (R) at residue 292 in NA (NA292K, N2 numbering) were found in different macaques that had been infected with A/Anhui/1/2013 (H7N9) and treated with NAIs. In the present study, the variant with NA292K showed not only resistance to NAIs but also lower replication activity in MDCK cells than did the virus with wild-type NA, whereas the variant with NA222T, which was less resistant to NAIs, showed replication activity similar to that of the wild-type virus. Next, we examined the pathogenicity of these H7N9 NAI-resistant viruses in macaques. The variants caused clinical signs similar to those caused by the wild-type virus with similar replication potency. However, the virus with NA292K was replaced within 7 days by that with NA292R (same as the wild-type) in nasal samples from macaques infected with the virus with NA292K, i.e. the so-called revertant (wild-type virus) became dominant in the population in the absence of an NAI. These results suggest that the clinical signs observed in macaques infected with the NA292K virus are caused by the NA292K virus and the NA292R virus and that the virus with NA292K may not replicate continuously in the upper respiratory tract of patients without treatment as effectively as the wild-type virus.
Collapse
|
18
|
Uehara T, Hayden FG, Kawaguchi K, Omoto S, Hurt AC, De Jong MD, Hirotsu N, Sugaya N, Lee N, Baba K, Shishido T, Tsuchiya K, Portsmouth S, Kida H. Treatment-Emergent Influenza Variant Viruses With Reduced Baloxavir Susceptibility: Impact on Clinical and Virologic Outcomes in Uncomplicated Influenza. J Infect Dis 2019; 221:346-355. [DOI: 10.1093/infdis/jiz244] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Background
Single-dose baloxavir rapidly reduces influenza virus titers and symptoms in patients with uncomplicated influenza, but viruses with reduced in vitro susceptibility due to amino acid substitutions at position 38 of polymerase acidic protein (PA/I38X) sometimes emerge.
Methods
We evaluated the kinetics, risk factors, and effects on clinical and virologic outcomes of emergence of PA/I38X-substituted viruses.
Results
Viruses containing PA/I38X substitutions were identified 3–9 days after baloxavir treatment in 9.7% (36/370) of patients, of whom 85.3% had transient virus titer rises. Median time to sustained cessation of infectious virus detection was 192, 48, and 96 hours in the baloxavir recipients with PA/I38X-substituted viruses, without PA/I38X-substituted viruses, and placebo recipients, respectively. The corresponding median times to alleviation of symptoms were 63.1, 51.0, and 80.2 hours, respectively. After day 5, symptom increases occurred in 11.5%, 8.0%, and 13.0%, respectively, and in 8.9% of oseltamivir recipients. Variant virus emergence was associated with lower baseline neutralizing antibody titers.
Conclusions
The emergence of viruses with PA/I38X substitutions following baloxavir treatment was associated with transient rises in infectious virus titers, prolongation of virus detectability, initial delay in symptom alleviation, and uncommonly with symptom rebound. The potential transmissibility of PA/I38X-substituted viruses requires careful study.
Clinical Trial Registration
NCT02954354.
Collapse
Affiliation(s)
| | | | | | | | - Aeron C Hurt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Menno D De Jong
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, the Netherlands
| | | | - Norio Sugaya
- Department of Pediatrics, Keiyu Hospital, Yokohama, Japan
| | - Nelson Lee
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Canada
| | | | | | | | | | - Hiroshi Kida
- Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| |
Collapse
|
19
|
Salata C, Sgarabotto D, Del Vecchio C, Solimbergo E, Marini G, Nicolè S, Franchin E, Parolin C, Calistri A, Palù G. Antiviral treatment and virological monitoring of oseltamivir-resistant influenza virus A(H1N1)pdm09 in a patient with chronic B lymphocytic leukemia. J Infect Chemother 2019; 25:543-546. [PMID: 31014561 DOI: 10.1016/j.jiac.2018.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/16/2018] [Accepted: 11/12/2018] [Indexed: 11/16/2022]
Abstract
We report the virological monitoring and the antiviral therapy adopted for the treatment of a patient affected by chronic B lymphocytic leukemia, who experienced a severe pneumonia with long-term shedding of influenza virus A(H1N1)pdm09, characterized by an early development of oseltamivir resistance.
Collapse
Affiliation(s)
- Cristiano Salata
- Department of Molecular Medicine, University of Padova, Padova, Italy; Microbiology and Virology Unit, Azienda Ospedaliera di Padova, Padova, Italy
| | - Dino Sgarabotto
- Division of Infectious Diseases, Azienda Ospedaliera di Padova, Padova, Italy
| | - Claudia Del Vecchio
- Department of Molecular Medicine, University of Padova, Padova, Italy; Microbiology and Virology Unit, Azienda Ospedaliera di Padova, Padova, Italy
| | - Erica Solimbergo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giulia Marini
- Division of Infectious Diseases, Azienda Ospedaliera di Padova, Padova, Italy
| | - Stefano Nicolè
- Division of Infectious Diseases, Azienda Ospedaliera di Padova, Padova, Italy
| | - Elisa Franchin
- Department of Molecular Medicine, University of Padova, Padova, Italy; Microbiology and Virology Unit, Azienda Ospedaliera di Padova, Padova, Italy
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padova, Padova, Italy; Microbiology and Virology Unit, Azienda Ospedaliera di Padova, Padova, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy; Microbiology and Virology Unit, Azienda Ospedaliera di Padova, Padova, Italy.
| |
Collapse
|
20
|
Sosa-Hernández JE, Escobedo-Avellaneda Z, Iqbal HMN, Welti-Chanes J. State-of-the-Art Extraction Methodologies for Bioactive Compounds from Algal Biome to Meet Bio-Economy Challenges and Opportunities. Molecules 2018; 23:E2953. [PMID: 30424551 PMCID: PMC6278541 DOI: 10.3390/molecules23112953] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/01/2018] [Accepted: 11/04/2018] [Indexed: 02/05/2023] Open
Abstract
Over the years, significant research efforts have been made to extract bioactive compounds by applying different methodologies for various applications. For instance, the use of bioactive compounds in several commercial sectors such as biomedical, pharmaceutical, cosmeceutical, nutraceutical and chemical industries, has promoted the need of the most suitable and standardized methods to extract these bioactive constituents in a sophisticated and cost-effective manner. In practice, several conventional extraction methods have numerous limitations, e.g., lower efficacy, high energy cost, low yield, etc., thus urges for new state-of-the-art extraction methodologies. Thus, the optimization along with the integration of efficient pretreatment strategies followed by traditional extraction and purification processes, have been the primary goal of current research and development studies. Among different sources, algal biome has been found as a promising and feasible source to extract a broader spectrum of bioactive compounds with point-of-care application potentialities. As evident from the literature, algal bio-products includes biofuels, lipids, polyunsaturated fatty acids, pigments, enzymes, polysaccharides, and proteins. The recovery of products from algal biomass is a matter of constant development and progress. This review covers recent advancements in the extraction methodologies such as enzyme-assisted extraction (EAE), supercritical-fluid extraction (SFE), microwave-assisted extraction (MAE) and pressurized-liquid extraction (PLF) along with their working mechanism for extracting bioactive compounds from algal-based sources to meet bio-economy challenges and opportunities. A particular focus has been given to design characteristics, performance evaluation, and point-of-care applications of different bioactive compounds of microalgae. The previous and recent studies on the anticancer, antibacterial, and antiviral potentialities of algal-based bioactive compounds have also been discussed with particular reference to the mechanism underlying the effects of these active constituents with the related pathways. Towards the end, the information is also given on the possible research gaps, future perspectives and concluding remarks.
Collapse
Affiliation(s)
- Juan Eduardo Sosa-Hernández
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología FEMSA, Ave. Eugenio Garza Sada 2501, C.P. 64849 Monterrey, N.L., Mexico.
| | | | | | | |
Collapse
|
21
|
Abstract
INTRODUCTION Influenza continues to be a major public health concern. Antivirals play an important role in limiting the burden of disease and preventing infection and/or transmission. The developments of such agents are heavily dependent on pre-clinical evaluation where animal models are used to answer questions that cannot be easily addressed in human clinical trials. There are numerous animal models available to study the potential benefits of influenza antivirals but each animal model has its own pros and cons. Areas covered: In this review, the authors describe the advantages and disadvantages of using mice, ferrets, guinea pigs, cotton rats, golden hamsters and non-human primates to evaluate influenza therapeutics. Expert opinion: Animals used for evaluating influenza therapeutics differ in their susceptibility to influenza virus infection, their ability to display clinical signs of illness following viral infection and in their practical requirements such as housing. Therefore, defining the scientific question being asked and the data output required will assist in selecting the most appropriate animal model.
Collapse
Affiliation(s)
- Edin J Mifsud
- a WHO Collaborating Centre for Reference and Research on Influenza , VIDRL, Peter Doherty Institute for Infection and Immunity , Melbourne , Australia
| | - Celeste Mk Tai
- a WHO Collaborating Centre for Reference and Research on Influenza , VIDRL, Peter Doherty Institute for Infection and Immunity , Melbourne , Australia
| | - Aeron C Hurt
- a WHO Collaborating Centre for Reference and Research on Influenza , VIDRL, Peter Doherty Institute for Infection and Immunity , Melbourne , Australia.,b Department of Microbiology and Immunology , University of Melbourne , Melbourne , Victoria , Australia
| |
Collapse
|
22
|
Iqbal HMN. The Quest for Materials-Based Hydrogels with Antimicrobial and Antiviral Potentialities. Open Virol J 2018; 12:69-79. [PMID: 30288196 PMCID: PMC6142656 DOI: 10.2174/1874357901812010069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 03/14/2018] [Accepted: 05/14/2018] [Indexed: 11/22/2022] Open
Abstract
In recent years, the Antimicrobial Resistance (AMR) or Multidrug Resistance (MDR) and viral infections have become serious health issues, globally. Finally, after decades of negligence, the AMR/MDR and viral infection issues have now captured a worldwide attention of the global leaders, public health community, legalization authorities, academia, research-based organizations, and medicinal sector of the modern world, alike. Aiming to resolve these issues, various methodological approaches have been exploited, in the past several years. Among them, biomaterials-based therapeutic hydrogels are of supreme interests for an enhanced and efficient delivery in the current biomedical sector. Depending on the regulatory authorities and practices, the antibiotics consumption was expedited than ever before driven by rising and increasing access, across the globe. Though the emergence of AMR/MDR in microorganisms and emergence/reemergence of viral infections are considered as a natural phenomenon, however, these concerning issues have been driven by those mentioned above faulty human behavior. In this context, many scientists, around the globe, are working at wider spectrum to resolve this problematic issue, efficiently. A proper understanding of biological mechanisms is essential to combat this global threat to the living beings. In this review, an effort has been made to highlight the potent features of materials based hydrogels possessing antimicrobial and antiviral potentialities. The information is also given on the potential research activities, and possible mechanisms of actions of hydrogels are discussed with a closeup look at the future recommendations.
Collapse
Affiliation(s)
- Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N.L., CP 64849, Mexico
| |
Collapse
|
23
|
Deecke L, Dobrovolny HM. Intermittent treatment of severe influenza. J Theor Biol 2018; 442:129-138. [PMID: 29355540 DOI: 10.1016/j.jtbi.2018.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/30/2017] [Accepted: 01/15/2018] [Indexed: 12/17/2022]
Abstract
Severe, long-lasting influenza infections are often caused by new strains of the virus. The long duration of these infections leads to an increased opportunity for the emergence of drug resistant mutants. This is particularly problematic since for new strains there is often no vaccine, so drug treatment is the first line of defense. One strategy for trying to minimize drug resistance is to apply drugs periodically. During treatment phases the wild-type virus decreases, but resistant virus might increase; when there is no treatment, wild-type virus will hopefully out-compete the resistant virus, driving down the number of resistant virus. A stochastic model of severe influenza is combined with a model of drug resistance to simulate long-lasting infections and intermittent treatment with two types of antivirals: neuraminidase inhibitors, which block release of virions; and adamantanes, which block replication of virions. Each drug's ability to reduce emergence of drug resistant mutants is investigated. We find that cell regeneration is required for successful implementation of intermittent treatment and that the optimal cycling parameters change with regeneration rate.
Collapse
Affiliation(s)
- Lucas Deecke
- Institut für Theoretische Physik, Universität zu Köln, Cologne, Germany
| | - Hana M Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, USA.
| |
Collapse
|
24
|
Dobrovolny HM, Beauchemin CAA. Modelling the emergence of influenza drug resistance: The roles of surface proteins, the immune response and antiviral mechanisms. PLoS One 2017; 12:e0180582. [PMID: 28700622 PMCID: PMC5503263 DOI: 10.1371/journal.pone.0180582] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/16/2017] [Indexed: 12/16/2022] Open
Abstract
The emergence of influenza drug resistance has become of particular interest as current planning for an influenza pandemic involves using massive amounts of antiviral drugs. We use semi-stochastic simulations to examine the emergence of drug resistant mutants during the course of a single infection within a patient in the presence and absence of antiviral therapy. We specifically examine three factors and their effect on the emergence of drug-resistant mutants: antiviral mechanism, the immune response, and surface proteins. We find that adamantanes, because they act at the start of the replication cycle to prevent infection, are less likely to produce drug-resistant mutants than NAIs, which act at the end of the replication cycle. A mismatch between surface proteins and internal RNA results in drug-resistant mutants being less likely to emerge, and emerging later in the infection because the mismatch gives antivirals a second chance to prevent propagation of the mutation. The immune response subdues slow growing infections, further reducing the probability that a drug resistant mutant will emerge and yield a drug-resistant infection. These findings improve our understanding of the factors that contribute to the emergence of drug resistance during the course of a single influenza infection.
Collapse
Affiliation(s)
- Hana M. Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, United States of America
- Department of Physics, Ryerson University, Toronto, ON, Canada
| | - Catherine A. A. Beauchemin
- Department of Physics, Ryerson University, Toronto, ON, Canada
- Interdisciplinary Theoretical Science (iTHES) Research Group at RIKEN, Wako, Japan
| |
Collapse
|
25
|
Rath B, Chen X, Spies V, Muehlhans S, Obermeier P, Tief F, Seeber L, Karsch K, Milde J, Skopnik H, Schweiger B, Duwe SC. Prospective surveillance of antiviral resistance in hospitalized infants less than 12 months of age with A(H3N2) influenza infection and treated with oseltamivir. Antivir Ther 2017; 22:515-522. [PMID: 28205506 DOI: 10.3851/imp3141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Infants exhibit elevated influenza virus loads and prolonged viral shedding, which may increase the risk for resistance development, especially in cases of suboptimal exposure to antiviral therapy. METHODS We performed a prospective surveillance of hospitalized infants undergoing oseltamivir therapy during the 2008-2009 and 2011-2012 influenza seasons at two paediatric hospitals in Germany. A total of 37 infants less than 1 year of age with laboratory confirmed influenza A(H3N2) infection received oseltamivir as per physician's order for 5 days (2008-2009 season: 2 mg/kg twice daily; 2011-2012 season: 2.0 mg/kg; 2.5 mg/kg and 3.0 mg/kg twice daily for infants <1 month; 2-3 months and 4-12 months, respectively). Virus load, the susceptibility to neuraminidase inhibitors (NAIs), and the presence of molecular markers of resistance to NAIs was assessed for influenza viruses recovered from respiratory samples collected at baseline and during follow-up visits. RESULTS Overall, 73% of the infants continued to shed viral RNA detectable by reverse transcription (RT)-PCR after dose number 10 of oseltamivir; 12 infants shed viruses, 2 of them (both 9 months of age) shed resistant viruses. Resistance was characterized by ≥1,000-fold increase of 50% inhibitory concentration (IC50) for oseltamivir, up to 50-fold for zanamivir and elevated Km values when compared to susceptible A(H3N2) strains. Sanger sequencing revealed the selection of the NA-R292K substitution in both instances (after dose number 10 on day 6). CONCLUSIONS Our data suggest that it may be relevant to monitor antiviral resistance systematically in all infants, considering that the European Medicines Agency has recently extended the licensure for oseltamivir to include full-term infants.
Collapse
Affiliation(s)
- Barbara Rath
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany.,Vienna Vaccine Safety Initiative, Berlin, Germany
| | - Xi Chen
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Vera Spies
- Klinikum Worms, Department of Pediatrics and Adolescent Medicine, Worms, Germany
| | - Susann Muehlhans
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Patrick Obermeier
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Franziska Tief
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Lea Seeber
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Katharina Karsch
- Charité University Medical Center, Department of Pediatrics, Berlin, Germany
| | - Jeanette Milde
- Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany
| | - Heino Skopnik
- Klinikum Worms, Department of Pediatrics and Adolescent Medicine, Worms, Germany
| | - Brunhilde Schweiger
- Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany
| | - Susanne C Duwe
- Robert Koch Institute, Division of Influenza Viruses and Other Respiratory Viruses, National Reference Centre for Influenza, Berlin, Germany
| |
Collapse
|
26
|
Antiviral Resistance in Influenza Viruses: Clinical and Epidemiological Aspects. ANTIMICROBIAL DRUG RESISTANCE 2017. [PMCID: PMC7122614 DOI: 10.1007/978-3-319-47266-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
There are three classes of antiviral drugs approved for the treatment of influenza: the M2 ion channel inhibitors (amantadine, rimantadine), neuraminidase (NA) inhibitors (laninamivir, oseltamivir, peramivir, zanamivir), and the protease inhibitor (favipiravir); some of the agents are only available in selected countries [1, 2]. These agents are effective at treating the signs and symptoms of influenza in patients infected with susceptible viruses. Clinical failure has been demonstrated in patients infected with viruses with primary resistance, i.e., antivirals can be present in the virus initially infecting the patient, or resistance may emerge during the course of therapy [3–5]. NA inhibitors are active against all nine NA subtypes recognized in nature [6], including highly pathogenic avian influenza A/H5N1 and recent low-pathogenic avian influenza A/H7N9 viruses [7]. Since seasonal influenza is usually an acute, self-limited illness in which viral clearance usually occurs rapidly due to innate and adaptive host immune responses, the emergence of drug-resistant variants would be anticipated to have limited effect on clinical recovery in otherwise healthy patients, as has been demonstrated clinically [3, 8, 9]. Unfortunately, immunocompromised or immunologically naïve hosts, such as young children and infants or those exposed to novel strains, are more likely to have mutations that confer resistance emergence during therapy; such resistant variants may also result in clinically significant adverse outcomes [10–13].
Collapse
|
27
|
Drug generations that combat influenza A virus infection. BIO-ALGORITHMS AND MED-SYSTEMS 2017. [DOI: 10.1515/bams-2016-0027] [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
AbstractInfluenza viruses are significant human respiratory pathogens that cause infections and unpredictable pandemic outbreaks. M2 ion-channel protein, participating in the transmission of viral genetic materials into infected cells, is considered to be the crucial target for old-generation drugs such as rimantadine and amantadine. Neuraminidase protein, which is responsible for the replication of the influenza virus, is affected by the new generation of drugs, including oseltamivir (Tamiflu) and zanamivir (Relenza). The virus mutations that cause oseltamivir resistance are also described. This review presents the details concerning the treatment of influenza neuraminidase inhibitors against the H5N1 strain. It also describes virus mutations that cause resistance to oseltamivir and presents a new drug, peramivir, which is a neuraminidase inhibitor that was introduced against the H1N1 epidemic. This work specifies the details of the pharmacokinetics, dosing, toxicity, side effects, and efficiency of the drugs being used against influenza A virus infections.
Collapse
|
28
|
Patel P, Bush T, Kojic EM, Overton ET, Henry K, Önen N, Rhame F, Conley L, Brooks JT, Fry A. Duration of Influenza Virus Shedding Among HIV-Infected Adults in the cART Era, 2010-2011. AIDS Res Hum Retroviruses 2016; 32:1180-1186. [PMID: 27174191 PMCID: PMC5175434 DOI: 10.1089/aid.2015.0349] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The duration of influenza virus shedding in HIV-infected adults is unknown and could affect quarantine and treatment recommendations. Participants were monitored for influenza-like illness (ILI), defined as fever and cough or sore throat, using weekly telephone audio computer-assisted self-interviews. Those with ILI were further evaluated at three HIV specialty clinics. For those with influenza, we collected nasopharyngeal washes every 3 days after the date of confirmed influenza infection for 21-28 days; specimens underwent reverse transcriptase - polymerase chain reaction (RT-PCR) and viral culture. Duration of influenza virus shedding was the interval from the date of onset (day 0) of ILI to the date of last culture-positive specimen. Characteristics were compared between patients with and without influenza using Fisher's exact test. We used the Wilcoxon rank-sum test to examine factors that may have affected influenza virus shedding. From October 2010 to April 2011, we enrolled 961 participants in syndromic surveillance and diagnosed 20 patients with influenza whose characteristics were as follows: median age 48 years (interquartile range [IQR]: 43-53), 60% male, 50% non-Hispanic black, 95% had been prescribed combination highly active antiretroviral therapy (cART), 85% were virologically suppressed (HIV RNA <400 copies/ml), median CD4 cell count 317 cells/mm3 (IQR: 190-544), and median follow-up time 21 days (IQR: 19-22). Compared with persons without influenza, persons with influenza were more likely to be older, use injection drugs, and have a lower median CD4 cell count and were less likely to have had an influenza vaccination in the past 12 months. Median durations of shedding, PCR detection, and ILI symptoms were 3 (IQR: 0-5), 10 (IQR: 6-15), and 14 days (IQR: 12-26), respectively. Median days of shedding were similar among patients with and without any prior influenza vaccination (0 vs. 4, p = .448), HIV viral suppression (2 vs. 6, p = .053), and oseltamivir use (5 vs. 0, p = .083). HIV-infected persons on cART in our study shed influenza virus for a similar duration as that reported for HIV-uninfected persons.
Collapse
Affiliation(s)
- Pragna Patel
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Timothy Bush
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - E. Milu Kojic
- Department of Infectious Diseases, Brown University, Providence, Rhode Island
| | - Edgar T. Overton
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Keith Henry
- Hennepin County Medical Center, University of Minnesota, Minneapolis, Minnesota
| | - Nur Önen
- Washington University School of Medicine, St. Louis, Missouri
| | - Frank Rhame
- Abbott-Northwestern Hospital, Minneapolis, Minnesota
| | - Lois Conley
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - John T. Brooks
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Alicia Fry
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
29
|
Romero-Beltran L, Baker SF, Puerto-Solís M, González-Losa R, Conde-Ferraez L, Alvarez-Sánchez LC, Martínez-Sobrido L, Ayora-Talavera G. Mutations at highly conserved residues in influenza A(H1N1)pdm09 virus affect neuraminidase activity. Virus Res 2016; 225:1-9. [PMID: 27596738 DOI: 10.1016/j.virusres.2016.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/17/2016] [Accepted: 09/02/2016] [Indexed: 01/16/2023]
Abstract
Influenza virus neuraminidase (NA) plays a pivotal role during viral growth since its sialidase activity allows the efficient release of nascent virions from infected cells. Consequently, mutations in the NA catalytic site affecting sialic acid (SA) cleavage may influence the biological properties of influenza viruses. This study reports two amino acid substitutions (N386K and P431S) in the NA of the influenza A(H1N1)pdm09 virus that emerged in 2009 in Mexico. The NA sialidase activity to cleave SA-like substrates, and viral growth were examined and the mutant viruses had various deficiencies. NA mutations N386K and P431S together or separately, and in the presence or absence of H275Y were further evaluated using recombinant influenza A/California/04/2009 (pH1N1) viruses containing single, double, or triple mutations. Viral growth was reduced in the presence of mutation P431S alone or combined with N386K and/or H275Y. Substrates hydrolysis was reduced when recombinant pH1N1 viruses were analyzed by NA inhibitory assays. Moreover, elution assays with guinea pig red blood cells indicated an unbalanced hemagglutinin (HA):NA functionality. Altogether, our data underline the functional significance of mutations at highly conserved sites in influenza virus NA glycoprotein and the occurrence of permissive mutations to compensate virus viability in vitro.
Collapse
Affiliation(s)
| | - Steven F Baker
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | | | | | | - Leidi C Alvarez-Sánchez
- Laboratorio de Virología, Universidad Autonoma de Yucatan, Mexico; Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
30
|
Duncan D. Stop the spread: prevention and reduction of influenza among older individuals. Br J Community Nurs 2016; 21:446-450. [PMID: 27594059 DOI: 10.12968/bjcn.2016.21.9.446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Affiliation(s)
- Debbie Duncan
- Senior Lecturer in Adult Nursing, Bucks New University, UK
| |
Collapse
|
31
|
Frise R, Bradley K, van Doremalen N, Galiano M, Elderfield RA, Stilwell P, Ashcroft JW, Fernandez-Alonso M, Miah S, Lackenby A, Roberts KL, Donnelly CA, Barclay WS. Contact transmission of influenza virus between ferrets imposes a looser bottleneck than respiratory droplet transmission allowing propagation of antiviral resistance. Sci Rep 2016; 6:29793. [PMID: 27430528 PMCID: PMC4949428 DOI: 10.1038/srep29793] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/20/2016] [Indexed: 12/19/2022] Open
Abstract
Influenza viruses cause annual seasonal epidemics and occasional pandemics. It is important to elucidate the stringency of bottlenecks during transmission to shed light on mechanisms that underlie the evolution and propagation of antigenic drift, host range switching or drug resistance. The virus spreads between people by different routes, including through the air in droplets and aerosols, and by direct contact. By housing ferrets under different conditions, it is possible to mimic various routes of transmission. Here, we inoculated donor animals with a mixture of two viruses whose genomes differed by one or two reverse engineered synonymous mutations, and measured the transmission of the mixture to exposed sentinel animals. Transmission through the air imposed a tight bottleneck since most recipient animals became infected by only one virus. In contrast, a direct contact transmission chain propagated a mixture of viruses suggesting the dose transferred by this route was higher. From animals with a mixed infection of viruses that were resistant and sensitive to the antiviral drug oseltamivir, resistance was propagated through contact transmission but not by air. These data imply that transmission events with a looser bottleneck can propagate minority variants and may be an important route for influenza evolution.
Collapse
Affiliation(s)
- Rebecca Frise
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | - Konrad Bradley
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | - Neeltje van Doremalen
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | - Monica Galiano
- Public Health England, Colindale, London, United Kingdom
| | - Ruth A. Elderfield
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | - Peter Stilwell
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | - Jonathan W. Ashcroft
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | | | - Shahjahan Miah
- Public Health England, Colindale, London, United Kingdom
| | - Angie Lackenby
- Public Health England, Colindale, London, United Kingdom
| | - Kim L. Roberts
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| | - Christl A. Donnelly
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, United Kingdom
| | - Wendy S. Barclay
- Imperial College London, Faculty of Medicine, Division of Infectious Disease, Norfolk Place, London, W2 1PG, United Kingdom
| |
Collapse
|
32
|
Gaymard A, Charles-Dufant A, Sabatier M, Cortay JC, Frobert E, Picard C, Casalegno JS, Rosa-Calatrava M, Ferraris O, Valette M, Ottmann M, Lina B, Escuret V. Impact on antiviral resistance of E119V, I222L and R292K substitutions in influenza A viruses bearing a group 2 neuraminidase (N2, N3, N6, N7 and N9). J Antimicrob Chemother 2016; 71:3036-3045. [PMID: 27432605 DOI: 10.1093/jac/dkw275] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES While subtype-specific substitutions linked to neuraminidase (NA) inhibitor resistance are well described in human N1 and N2 influenza NAs, little is known about other NA subtypes. The aim of this study was to determine whether the R292K and E119V ± I222L substitutions could be associated with oseltamivir resistance in all group 2 NAs and had an impact on virus fitness. METHODS Reassortant viruses with WT NA or variant N2, N3, N6, N7 or N9 NAs, bearing R292K or E119V ± I222L substitutions, were produced by reverse genetics. The antiviral susceptibility, activity, Km of the NA, mutation stability and in vitro virus fitness in MDCK cells were determined. RESULTS NA activities could be ranked as follows regardless of the substitution: N3 ≥ N6 > N2 ≥ N9 > N7. Using NA inhibitor resistance interpretation criteria used for human N1 or N2, the NA-R292K substitution conferred highly reduced inhibition by oseltamivir and the N6- or N9-R292K substitution conferred reduced inhibition by zanamivir and laninamivir. Viruses with the N3- or N6-E119V substitution showed normal inhibition by oseltamivir, while those with the N2-, N7- or N9-E119V substitution showed reduced inhibition by oseltamivir. Viruses with NA-E119V + I222L substitutions showed reduced inhibition (N3 and N6) or highly reduced inhibition (N2, N7 and N9) by oseltamivir. Viruses bearing the NA-R292K substitution had lower affinity and viruses bearing the NA-E119V substitution had higher affinity for the MUNANA substrate than viruses with corresponding WT NA. CONCLUSIONS NA-R292K and E119V + I222L substitutions conferred reduced inhibition by oseltamivir for all group 2 NAs. Surveillance of NA inhibitor resistance for zoonotic and human influenza viruses and the development of novel antiviral agents with different targets should be continued.
Collapse
Affiliation(s)
- Alexandre Gaymard
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Aymeric Charles-Dufant
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Murielle Sabatier
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Jean-Claude Cortay
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Emilie Frobert
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Caroline Picard
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Jean-Sébastien Casalegno
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Manuel Rosa-Calatrava
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Olivier Ferraris
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Martine Valette
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Michèle Ottmann
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France
| | - Bruno Lina
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France.,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| | - Vanessa Escuret
- Univ Lyon, Université Lyon 1, Faculté de Médecine Lyon Est, CIRI Inserm U1111, équipe Virpath, F-69008, Lyon, France .,Hospices Civils de Lyon, Centre National de Référence virus influenzae France Sud, Laboratoire de Virologie, Groupement Hospitalier Nord, F-69317, Lyon cedex 04, France
| |
Collapse
|
33
|
Kurebayashi Y, Takahashi T, Tamoto C, Sahara K, Otsubo T, Yokozawa T, Shibahara N, Wada H, Minami A, Ikeda K, Suzuki T. High-Efficiency Capture of Drug Resistant-Influenza Virus by Live Imaging of Sialidase Activity. PLoS One 2016; 11:e0156400. [PMID: 27232333 PMCID: PMC4883822 DOI: 10.1371/journal.pone.0156400] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 05/13/2016] [Indexed: 11/18/2022] Open
Abstract
Influenza A and B viruses possess a neuraminidase protein that shows sialidase activity. Influenza virus-specific neuraminidase inhibitors (NAIs) are commonly used for clinical treatment of influenza. However, some influenza A and B viruses that are resistant to NAIs have emerged in nature. NAI-resistant viruses have been monitored in public hygiene surveys and the mechanism underlying the resistance has been studied. Here, we describe a new assay for selective detection and isolation of an NAI-resistant virus in a speedy and easy manner by live fluorescence imaging of viral sialidase activity, which we previously developed, in order to achieve high-efficiency capture of an NAI-resistant virus. An NAI-resistant virus maintains sialidase activity even at a concentration of NAI that leads to complete deactivation of the virus. Infected cells and focuses (infected cell populations) of an oseltamivir-resistant virus were selectively visualized by live fluorescence sialidase imaging in the presence of oseltamivir, resulting in high-efficiency isolation of the resistant viruses. The use of a combination of other NAIs (zanamivir, peramivir, and laninamivir) in the imaging showed that the oseltamivir-resistant virus isolated in 2008 was sensitive to zanamivir and laninamivir but resistant to peramivir. Fluorescence imaging in the presence of zanamivir also succeeded in selective live-cell visualization of cells that expressed zanamivir-resistant NA. Fluorescence imaging of NAI-resistant sialidase activity will be a powerful method for study of the NAI resistance mechanism, for public monitoring of NAI-resistant viruses, and for development of a new NAI that shows an effect on various NAI-resistant mutations.
Collapse
Affiliation(s)
- Yuuki Kurebayashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
| | - Tadanobu Takahashi
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
| | - Chihiro Tamoto
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
| | - Keiji Sahara
- Shizuoka Institute of Environment and Hygiene, Shizuoka-shi, Shizuoka, Japan
| | - Tadamune Otsubo
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, Kure-shi, Hiroshima, Japan
| | - Tatsuya Yokozawa
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
| | - Nona Shibahara
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
- Shizuoka City Institute of Environmental Sciences and Public Health, Shizuoka-shi, Shizuoka, Japan
| | - Hirohisa Wada
- Shizuoka City Institute of Environmental Sciences and Public Health, Shizuoka-shi, Shizuoka, Japan
| | - Akira Minami
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
| | - Kiyoshi Ikeda
- Department of Organic Chemistry, School of Pharmaceutical Sciences, Hiroshima International University, Kure-shi, Hiroshima, Japan
| | - Takashi Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan
| |
Collapse
|
34
|
Ann J, Abed Y, Beaulieu E, Bouhy X, Joly MH, Dubé K, Carbonneau J, Hamelin ME, Mallett C, Boivin G. Impact of a large deletion in the neuraminidase protein identified in a laninamivir-selected influenza A/Brisbane/10/2007 (H3N2) variant on viral fitness in vitro and in ferrets. Influenza Other Respir Viruses 2016; 10:122-6. [PMID: 26526406 PMCID: PMC4746560 DOI: 10.1111/irv.12356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2015] [Indexed: 12/22/2022] Open
Abstract
Viral fitness of a laninamivir‐selected influenza A/Brisbane/10/2007‐like (H3N2) isolate (LRVp9) containing a 237‐amino acid neuraminidase deletion and a P194L hemagglutinin mutation was evaluated in vitro and in ferrets. LRVp9 and the wild‐type (WT) virus showed comparable replication kinetics in MDCK‐ST6GalI cells. Cultured virus was recovered between days 2 and 5 post‐infection in nasal washes (NW) from the 4 WT‐infected ferrets whereas no virus was recovered from the LRVp9‐infected animals. There was a ≥1 log reduction in viral RNA copies/μl of NW for LRVp9 compared to WT at most time points. The large neuraminidase deletion compromises viral infectivity in vivo.
Collapse
Affiliation(s)
- Julie Ann
- CHU de Quebec Research Center (CHUL) and Laval University, Québec City, QC, Canada
| | - Yacine Abed
- CHU de Quebec Research Center (CHUL) and Laval University, Québec City, QC, Canada
| | | | - Xavier Bouhy
- CHU de Quebec Research Center (CHUL) and Laval University, Québec City, QC, Canada
| | | | | | - Julie Carbonneau
- CHU de Quebec Research Center (CHUL) and Laval University, Québec City, QC, Canada
| | - Marie-Eve Hamelin
- CHU de Quebec Research Center (CHUL) and Laval University, Québec City, QC, Canada
| | | | - Guy Boivin
- CHU de Quebec Research Center (CHUL) and Laval University, Québec City, QC, Canada
| |
Collapse
|
35
|
Wilson BA, Garud NR, Feder AF, Assaf ZJ, Pennings PS. The population genetics of drug resistance evolution in natural populations of viral, bacterial and eukaryotic pathogens. Mol Ecol 2016; 25:42-66. [PMID: 26578204 PMCID: PMC4943078 DOI: 10.1111/mec.13474] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/28/2015] [Accepted: 10/08/2015] [Indexed: 01/09/2023]
Abstract
Drug resistance is a costly consequence of pathogen evolution and a major concern in public health. In this review, we show how population genetics can be used to study the evolution of drug resistance and also how drug resistance evolution is informative as an evolutionary model system. We highlight five examples from diverse organisms with particular focus on: (i) identifying drug resistance loci in the malaria parasite Plasmodium falciparum using the genomic signatures of selective sweeps, (ii) determining the role of epistasis in drug resistance evolution in influenza, (iii) quantifying the role of standing genetic variation in the evolution of drug resistance in HIV, (iv) using drug resistance mutations to study clonal interference dynamics in tuberculosis and (v) analysing the population structure of the core and accessory genome of Staphylococcus aureus to understand the spread of methicillin resistance. Throughout this review, we discuss the uses of sequence data and population genetic theory in studying the evolution of drug resistance.
Collapse
Affiliation(s)
| | | | | | - Zoe J. Assaf
- Department of GeneticsStanford UniversityStanfordCA94305USA
| | - Pleuni S. Pennings
- Department of BiologySan Francisco State UniversityRoom 520Hensill Hall1600 Holloway AveSan FranciscoCA94132USA
| |
Collapse
|
36
|
Kawakami T, Hirabayashi Y, Kawakami F, Isobe R, Kaneko N, Mimura Y, Ito T, Furuta K, Shimazaki M, Nakazawa H, Kitano K. Persistent Infection of Drug-resistant Influenza A Virus during Chemotherapy for Malignant Lymphoma. Intern Med 2016; 55:1807-10. [PMID: 27374689 DOI: 10.2169/internalmedicine.55.6327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We herein report the case of an 80-year-old man with malignant lymphoma who became persistently infected with influenza A virus. Although he was repeatedly treated with NA inhibitors, such as oseltamivir or peramivir, nasal cavity swab tests for influenza A antigen continued to be positive for more than 2 months. Virological analyses revealed that he was infected with the NA inhibitor-resistant A (H3N2) virus possessing an R292K substitution in the NA protein. These findings suggest that a drug-resistant influenza virus strain might selectively survive antiviral therapy in elderly patients with refractory malignant lymphoma undergoing multiple chemotherapies.
Collapse
Affiliation(s)
- Toru Kawakami
- Division of Hematology, NHO Matsumoto Medical Center, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
A Balance between Inhibitor Binding and Substrate Processing Confers Influenza Drug Resistance. J Mol Biol 2015; 428:538-553. [PMID: 26656922 DOI: 10.1016/j.jmb.2015.11.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 11/22/2022]
Abstract
The therapeutic benefits of the neuraminidase (NA) inhibitor oseltamivir are dampened by the emergence of drug resistance mutations in influenza A virus (IAV). To investigate the mechanistic features that underlie resistance, we developed an approach to quantify the effects of all possible single-nucleotide substitutions introduced into important regions of NA. We determined the experimental fitness effects of 450 nucleotide mutations encoding positions both surrounding the active site and at more distant sites in an N1 strain of IAV in the presence and absence of oseltamivir. NA mutations previously known to confer oseltamivir resistance in N1 strains, including H275Y and N295S, were adaptive in the presence of drug, indicating that our experimental system captured salient features of real-world selection pressures acting on NA. We identified mutations, including several at position 223, that reduce the apparent affinity for oseltamivir in vitro. Position 223 of NA is located adjacent to a hydrophobic portion of oseltamivir that is chemically distinct from the substrate, making it a hotspot for substitutions that preferentially impact drug binding relative to substrate processing. Furthermore, two NA mutations, K221N and Y276F, each reduce susceptibility to oseltamivir by increasing NA activity without altering drug binding. These results indicate that competitive expansion of IAV in the face of drug pressure is mediated by a balance between inhibitor binding and substrate processing.
Collapse
|
38
|
Koelle K, Rasmussen DA. The effects of a deleterious mutation load on patterns of influenza A/H3N2's antigenic evolution in humans. eLife 2015; 4:e07361. [PMID: 26371556 PMCID: PMC4611170 DOI: 10.7554/elife.07361] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 09/14/2015] [Indexed: 11/19/2022] Open
Abstract
Recent phylogenetic analyses indicate that RNA virus populations carry a significant deleterious mutation load. This mutation load has the potential to shape patterns of adaptive evolution via genetic linkage to beneficial mutations. Here, we examine the effect of deleterious mutations on patterns of influenza A subtype H3N2's antigenic evolution in humans. By first analyzing simple models of influenza that incorporate a mutation load, we show that deleterious mutations, as expected, act to slow the virus's rate of antigenic evolution, while making it more punctuated in nature. These models further predict three distinct molecular pathways by which antigenic cluster transitions occur, and we find phylogenetic patterns consistent with each of these pathways in influenza virus sequences. Simulations of a more complex phylodynamic model further indicate that antigenic mutations act in concert with deleterious mutations to reproduce influenza's spindly hemagglutinin phylogeny, co-circulation of antigenic variants, and high annual attack rates. DOI:http://dx.doi.org/10.7554/eLife.07361.001 Each year, up to 15% of the world's population experience symptoms of an influenza infection, also commonly known as flu. The most common culprit is a strain of the virus called influenza type A subtype H3N2. One reason that so many people become infected each year is that this virus evolves rapidly. Within a few years, proteins on the surface of the virus known as antigens become less recognizable to the immune system of a person who has been previously infected. This means that the person can become ill with the virus again because their immune system cannot mount an effective response to the evolved virus strain. Influenza virus strains evolve rapidly because their genetic material accumulates mutations quickly. Although some of these mutations are beneficial to the virus, other mutations are harmful and reduce the ability of the virus to spread. Sometimes beneficial mutations may occur alongside harmful ones, but it is not known how the harmful mutations affect the evolution of the virus. Here, Koelle and Rasmussen used computer models of H3N2 influenza to examine the effect of harmful mutations on the evolution of this virus population. The models show that harmful mutations limit how quickly the antigens can evolve. Also, the presence of these harmful mutations effectively acts as a sieve: they allow only large changes in the antigens to establish in the virus population. The models suggest that there are three routes by which large changes in the antigens on H3N2 viruses may occur. The first is by a single mutation that has a big effect on the antigens in viruses that only carry a few harmful mutations, but these large mutations would not happen very often. Another route may be through more common mutations that have only a small or moderate benefit, which would allow the virus to become more common in the population before it acquires a beneficial mutation with a much greater effect. The third possibility is that a large beneficial mutation may arise in viruses that have many harmful mutations. These harmful mutations may initially limit the ability of the virus to spread, but over time, some of these harmful mutations may then be lost. Koelle and Rasmussen found that the computer models could recreate the patterns of virus evolution that have been observed in real strains of H3N2. Researchers use predictions of influenza evolution to help them decide which virus strains should be included in flu vaccines each year. Koelle and Rasmussen findings indicate that harmful mutations should be considered when making these predictions. DOI:http://dx.doi.org/10.7554/eLife.07361.002
Collapse
Affiliation(s)
- Katia Koelle
- Department of Biology, Duke University, Durham, United States.,Fogarty International Center, National Institutes of Health, Bethesda, United States
| | - David A Rasmussen
- Department of Biology, Duke University, Durham, United States.,Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| |
Collapse
|
39
|
Buhnerkempe MG, Gostic K, Park M, Ahsan P, Belser JA, Lloyd-Smith JO. Mapping influenza transmission in the ferret model to transmission in humans. eLife 2015; 4. [PMID: 26329460 PMCID: PMC4586390 DOI: 10.7554/elife.07969] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/02/2015] [Indexed: 12/27/2022] Open
Abstract
The controversy surrounding 'gain-of-function' experiments on high-consequence avian influenza viruses has highlighted the role of ferret transmission experiments in studying the transmission potential of novel influenza strains. However, the mapping between influenza transmission in ferrets and in humans is unsubstantiated. We address this gap by compiling and analyzing 240 estimates of influenza transmission in ferrets and humans. We demonstrate that estimates of ferret secondary attack rate (SAR) explain 66% of the variation in human SAR estimates at the subtype level. Further analysis shows that ferret transmission experiments have potential to identify influenza viruses of concern for epidemic spread in humans, though small sample sizes and biological uncertainties prevent definitive classification of human transmissibility. Thus, ferret transmission experiments provide valid predictions of pandemic potential of novel influenza strains, though results should continue to be corroborated by targeted virological and epidemiological research. DOI:http://dx.doi.org/10.7554/eLife.07969.001 Every year, thousands of people develop influenza (flu). After being infected by the influenza virus, the immune systems of most people adapt to fight off the virus if it is encountered again. However, there are many different strains of influenza, and new strains constantly evolve. Therefore, although someone may have developed resistance to one previously encountered strain, they can still become ill if another strain infects them. Different strains of the influenza virus have different abilities to spread between people and make them ill. One way that scientists assess whether a particular strain of influenza is a threat to people is by studying ferrets, which develop many of the same flu symptoms as humans. However, questions have been raised over how accurately ferret studies reflect whether a particular virus strain will spread between humans. Controversy has also arisen over experiments in which ferrets are infected with genetically engineered strains of influenza that mimic how a strain that has evolved in birds could adapt to cause a pandemic in humans. In 2014, the United States government suggested that such research should be temporarily stopped until more is known about the risks and usefulness of these studies. Now, Buhnerkempe, Gostic et al. have compared the results of 240 ferret and human studies that aimed to assess how easily strains of influenza spread. Specifically, the studies looked at how often a healthy ferret or human became ill when exposed to an animal or human infected with a particular strain of influenza. The results of the ferret transmission studies matched well with transmission patterns observed in human studies. Ferret studies that assessed how the influenza virus is transmitted through the air via sneezes and coughs were particularly good at predicting how the virus spreads in humans. But Buhnerkempe, Gostic et al. caution that ferret studies are not always accurate, partly because they involve small numbers of animals, which can skew the results. There also needs to be more effort to standardize the procedures and measurements used in ferret studies. Still, the analysis suggests that overall, ferret studies are a useful tool for making an initial prediction of which influenza strains may cause a pandemic in humans, which can then be verified using other methods. DOI:http://dx.doi.org/10.7554/eLife.07969.002
Collapse
Affiliation(s)
- Michael G Buhnerkempe
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, United States
| | - Katelyn Gostic
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, United States
| | - Miran Park
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, United States
| | - Prianna Ahsan
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, United States
| | - Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, United States
| | - James O Lloyd-Smith
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
40
|
Amino acid substitutions in the neuraminidase protein of an H9N2 avian influenza virus affect its airborne transmission in chickens. Vet Res 2015; 46:44. [PMID: 25928577 PMCID: PMC4404070 DOI: 10.1186/s13567-014-0142-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 12/15/2014] [Indexed: 01/21/2023] Open
Abstract
Cases of H9N2 avian influenza virus (AIV) in poultry are increasing throughout many Eurasian countries, and co-infections with other pathogens have resulted in high morbidity and mortality in poultry. Few studies have investigated the genetic factors of virus airborne transmission which determine the scope of this epidemic. In this study, we used specific-pathogen-free chickens housed in isolators to investigate the airborne transmissibility of five recombinant H9N2 AIV rescued by reverse genetic technology. The results show that airborne transmission of A/Chicken/Shandong/01/2008 (SD01) virus was related to the neuraminidase (NA) gene, and four amino acid mutations (D368E, S370L, E313K and G381D) within the head region of the SD01 NA, reduced virus replication in the respiratory tract of chickens, reduced virus NA activity, and resulted in a loss of airborne transmission ability in chickens. Similarly, reverse mutations of these four amino acids in the NA protein of r01/NASS virus, conferred an airborne transmission ability to the recombinant virus. We conclude that these four NA residues may be significant genetic markers for evaluating potential disease outbreak of H9N2 AIV, and propose that immediate attention should be paid to the airborne transmission of this virus.
Collapse
|
41
|
Abstract
The ability of an influenza virus to transmit efficiently from human-to-human is a major factor in determining the epidemiological impact of that strain. The use of a relevant animal model to identify viral determinants of transmission, as well as host and environmental factors affecting transmission efficiency, is therefore critical for public health. The characterization of newly emerging influenza viruses in terms of their potential to transmit in a mammalian host is furthermore an important part of pandemic risk assessment. For these reasons, a guinea pig model of influenza virus transmission was developed in 2006. The guinea pig provides an important alternative to preexisting models for influenza. Most influenza viruses do not readily transmit among mice. Ferrets, while highly relevant, are expensive and can be difficult to obtain in high numbers. Moreover, it is generally accepted that efforts to accurately model human disease are strengthened by the use of multiple animal species. Herein, we provide an overview of influenza virus infectivity, growth, and transmission in the guinea pig and highlight knowledge gained on the topic of influenza virus transmission using the guinea pig model.
Collapse
Affiliation(s)
- Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | |
Collapse
|
42
|
Permissive changes in the neuraminidase play a dominant role in improving the viral fitness of oseltamivir-resistant seasonal influenza A(H1N1) strains. Antiviral Res 2014; 114:57-61. [PMID: 25512229 DOI: 10.1016/j.antiviral.2014.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 11/24/2022]
Abstract
Permissive neuraminidase (NA) substitutions such as R222Q, V234M and D344N have facilitated the emergence and worldwide spread of oseltamivir-resistant influenza A/Brisbane/59/2007 (H1N1)-H275Y viruses. However, the potential contribution of genetic changes in other viral segments on viral fitness remains poorly investigated. A series of recombinant A(H1N1)pdm09 and A/WSN/33 7:1 reassortants containing the wild-type (WT) A/Brisbane/59/2007 NA gene or its single (H275Y) and double (H275Y/Q222R, H275Y/M234V and H275Y/N344D) variants were generated and their replicative properties were assessed in vitro. The Q222R reversion substitution significantly reduced viral titers when evaluated in both A(H1N1)pdm09 and A/WSN/33 backgrounds. The permissive role of the R222Q was further confirmed using A/WSN/33 7:1 reassortants containing the NA gene of the oseltamivir-susceptible or oseltamivir-resistant influenza A/Mississippi/03/2001 strains. Therefore, NA permissive substitutions play a dominant role for improving viral replication of oseltamivir-resistant A (H1N1)-H275Y viruses in vitro.
Collapse
|
43
|
Behera AK, Basu S, Cherian SS. Molecular mechanism of the enhanced viral fitness contributed by secondary mutations in the hemagglutinin protein of oseltamivir resistant H1N1 influenza viruses: modeling studies of antibody and receptor binding. Gene 2014; 557:19-27. [PMID: 25479009 DOI: 10.1016/j.gene.2014.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 11/25/2022]
Abstract
The envelope protein hemagglutinin (HA) of influenza viruses is primarily associated with host antibody and receptor interactions. The HA protein is known to maintain a functional balance with neuraminidase (NA), the other major envelope protein. Prior to 2007-2008, human seasonal H1N1 viruses possessing the NA H274Y mutation, which confers oseltamivir resistance, generally had low growth capability. Subsequently, secondary mutations that compensate for the deleterious effect of the NA H274Y mutation have been identified. The molecular mechanism of how the defect could be counteracted by these secondary mutations is not fully understood. We studied here the effect of three such mutations (T86K, K144E and R192K) in the HA protein, which are located at either the HA receptor binding site or in the H1N1 antigenic sites. Molecular docking and dynamics studies showed that, of the three mutations, the R192K mutation could have mediated neutralizing antibody escape and decreased receptor binding affinity, either or both of which may have contributed to increased viral fitness. The study suggests the molecular basis of enhanced viral fitness induced by secondary mutations in the evolution of oseltamivir-resistant influenza strains.
Collapse
Affiliation(s)
- Abhisek Kumar Behera
- Bioinformatics Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Post Box No. 11, Pune 411001, Maharashtra, India.
| | - Sushmita Basu
- Bioinformatics Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Post Box No. 11, Pune 411001, Maharashtra, India.
| | - Sarah S Cherian
- Bioinformatics Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Post Box No. 11, Pune 411001, Maharashtra, India.
| |
Collapse
|
44
|
Animal models for influenza viruses: implications for universal vaccine development. Pathogens 2014; 3:845-74. [PMID: 25436508 PMCID: PMC4282889 DOI: 10.3390/pathogens3040845] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/10/2014] [Accepted: 10/10/2014] [Indexed: 01/22/2023] Open
Abstract
Influenza virus infections are a significant cause of morbidity and mortality in the human population. Depending on the virulence of the influenza virus strain, as well as the immunological status of the infected individual, the severity of the respiratory disease may range from sub-clinical or mild symptoms to severe pneumonia that can sometimes lead to death. Vaccines remain the primary public health measure in reducing the influenza burden. Though the first influenza vaccine preparation was licensed more than 60 years ago, current research efforts seek to develop novel vaccination strategies with improved immunogenicity, effectiveness, and breadth of protection. Animal models of influenza have been essential in facilitating studies aimed at understanding viral factors that affect pathogenesis and contribute to disease or transmission. Among others, mice, ferrets, pigs, and nonhuman primates have been used to study influenza virus infection in vivo, as well as to do pre-clinical testing of novel vaccine approaches. Here we discuss and compare the unique advantages and limitations of each model.
Collapse
|
45
|
Duan S, Govorkova EA, Bahl J, Zaraket H, Baranovich T, Seiler P, Prevost K, Webster RG, Webby RJ. Epistatic interactions between neuraminidase mutations facilitated the emergence of the oseltamivir-resistant H1N1 influenza viruses. Nat Commun 2014; 5:5029. [PMID: 25297528 PMCID: PMC4197134 DOI: 10.1038/ncomms6029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 08/19/2014] [Indexed: 01/06/2023] Open
Abstract
Oseltamivir-resistant H1N1 influenza viruses carrying the H275Y neuraminidase mutation predominated worldwide during the 2007–2009 seasons. While several neuraminidase substitutions were found to be necessary to counteract the adverse effects of H275Y, the order and impact of evolutionary events involved remain elusive. Here, we reconstruct H1N1 neuraminidase phylogeny during 1999–2009, estimate the timing and order of crucial amino acid changes, and evaluate their impact on the biological outcome of the H275Y mutation. Of the twelve neuraminidase substitutions that occurred during 1999–2009, five (chronologically, V234M, R222Q, K329E, D344N, H275Y, and D354G) are necessary for maintaining full neuraminidase function in the presence of the H275Y mutation by altering protein accumulation or enzyme affinity/activity. The sequential emergence and cumulative effects of these mutations clearly illustrate a role for epistasis in shaping the emergence and subsequent evolution of a drug-resistant virus population, which can be useful in understanding emergence of novel viral phenotypes of influenza.
Collapse
Affiliation(s)
- Susu Duan
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Elena A Govorkova
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Justin Bahl
- 1] School of Public Health, The University of Texas Health Science Center at Houston, 1200 Pressler Street, Houston, Texas 77030, USA [2] Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School, 8 College Road, Singapore 169857, Singapore
| | - Hassan Zaraket
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Tatiana Baranovich
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Patrick Seiler
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Kristi Prevost
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Robert G Webster
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 330, Memphis, Tennessee 38105, USA
| |
Collapse
|
46
|
Hai R, Schmolke M, Leyva-Grado VH, Thangavel RR, Margine I, Jaffe EL, Krammer F, Solórzano A, García-Sastre A, Palese P, Bouvier NM. Influenza A(H7N9) virus gains neuraminidase inhibitor resistance without loss of in vivo virulence or transmissibility. Nat Commun 2014; 4:2854. [PMID: 24326875 PMCID: PMC3863970 DOI: 10.1038/ncomms3854] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 11/01/2013] [Indexed: 12/17/2022] Open
Abstract
Without baseline human immunity to the emergent avian influenza A(H7N9) virus, neuraminidase inhibitors are vital for controlling viral replication in severe infections. An amino acid change in the viral neuraminidase associated with drug resistance, NA-R292K (N2 numbering), has been found in some H7N9 clinical isolates. Here we assess the impact of the NA-R292K substitution on antiviral sensitivity and viral replication, pathogenicity and transmissibility of H7N9 viruses. Our data indicate that an H7N9 isolate encoding the NA-R292K substitution is highly resistant to oseltamivir and peramivir and partially resistant to zanamivir. Furthermore, H7N9 reassortants with and without the resistance mutation demonstrate comparable viral replication in primary human respiratory cells, virulence in mice and transmissibility in guinea pigs. Thus, in stark contrast to oseltamivir-resistant seasonal influenza A(H3N2) viruses, H7N9 virus replication and pathogenicity in these models are not substantially altered by the acquisition of high-level oseltamivir resistance due to the NA-R292K mutation. Some clinical isolates of influenza A(H7N9) virus encode a mutation within neuraminidase that could confer resistance to the only class of drugs active against H7N9. Here, the authors show that this mutation does not affect viral replication and pathogenicity while mediating resistance to antivirals in vivo.
Collapse
Affiliation(s)
- Rong Hai
- 1] Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1124, New York, New York 10029, USA [2]
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Severity of clinical disease and pathology in ferrets experimentally infected with influenza viruses is influenced by inoculum volume. J Virol 2014; 88:13879-91. [PMID: 25187553 DOI: 10.1128/jvi.02341-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Ferrets are a valuable model for influenza virus pathogenesis, virus transmission, and antiviral therapy studies. However, the contributions of the volume of inoculum administered and the ferret's respiratory tract anatomy to disease outcome have not been explored. We noted variations in clinical disease outcomes and the volume of inoculum administered and investigated these differences by administering two influenza viruses (A/California/07/2009 [H1N1 pandemic] and A/Minnesota/11/2010 [H3N2 variant]) to ferrets intranasally at a dose of 10(6) 50% tissue culture infective doses in a range of inoculum volumes (0.2, 0.5, or 1.0 ml) and followed viral replication, clinical disease, and pathology over 6 days. Clinical illness and respiratory tract pathology were the most severe and most consistent when the viruses were administered in a volume of 1.0 ml. Using a modified micro-computed tomography imaging method and examining gross specimens, we found that the right main-stem bronchus was consistently larger in diameter than the left main-stem bronchus, though the latter was longer and straighter. These anatomic features likely influence the distribution of the inoculum in the lower respiratory tract. A 1.0-ml volume of inoculum is optimal for delivery of virus to the lower respiratory tract of ferrets, particularly when evaluation of clinical disease is desired. Furthermore, we highlight important anatomical features of the ferret lung that influence the kinetics of viral replication, clinical disease severity, and lung pathology. IMPORTANCE Ferrets are a valuable model for influenza virus pathogenesis, virus transmission, and antiviral therapy studies. Clinical disease in ferrets is an important parameter in evaluating the virulence of novel influenza viruses, and findings are extrapolated to virulence in humans. Therefore, it is highly desirable that the data from different laboratories be accurate and reproducible. We have found that, even when the same virus was administered at similar doses, different investigators reported a range of clinical disease outcomes, from asymptomatic infection to severe weight loss, ocular and nasal discharge, sneezing, and lethargy. We found that a wide range of inoculum volumes was used to experimentally infect ferrets, and we sought to determine whether the variations in disease outcome were the result of the volume of inoculum administered. These data highlight some less explored features of the model, methods of experimental infection, and clinical disease outcomes in a research setting.
Collapse
|
48
|
Hu J, Liu X. Crucial role of PA in virus life cycle and host adaptation of influenza A virus. Med Microbiol Immunol 2014; 204:137-49. [PMID: 25070354 DOI: 10.1007/s00430-014-0349-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/16/2014] [Indexed: 02/01/2023]
Abstract
The PA protein is the third subunit of the polymerase complex of influenza A virus. Compared with the other two polymerase subunits (PB2 and PB1), its precise functions are less defined. However, in recent years, advances in protein expression and crystallization technologies and also the reverse genetics, greatly accelerate our understanding of the essential role of PA in virus infection. Here, we first review the current literature on this remarkably multifunctional viral protein regarding virus life cycle, including viral RNA transcription and replication, viral genome packaging and assembly. We then discuss the various roles of PA in host adaption in avian species and mammals, general virus-host interaction, and host protein synthesis shutoff. We also review the recent findings about the novel proteins derived from PA. Finally, we discuss the prospects of PA as a target for the development of new antiviral approaches and drugs.
Collapse
Affiliation(s)
- Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China
| | | |
Collapse
|
49
|
Bruminhent J, Deziel PJ, Wotton JT, Binnicker MJ, Razonable RR. Prolonged shedding of pandemic influenza A (H1N1) 2009 virus in a pancreas-after-kidney transplant recipient. J Clin Virol 2014; 61:302-4. [PMID: 25081940 DOI: 10.1016/j.jcv.2014.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/04/2014] [Accepted: 07/08/2014] [Indexed: 11/17/2022]
Abstract
Prolonged shedding of influenza virus has been reported in immunocompromised patients. Delayed viral clearance may contribute to antiviral resistance and nosocomial transmission. We report a case of a pancreas-after-kidney transplant recipient who had detectable pandemic influenza A virus for 12 months. Pyrosequencing analysis detected the H275Y mutation, which is associated with resistance to oseltamivir.
Collapse
Affiliation(s)
| | - Paul J Deziel
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA.
| | - Jason T Wotton
- Minnesota Department of Health, St. Paul, MN 55164, USA.
| | - Matthew J Binnicker
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Raymund R Razonable
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
50
|
Hurt AC. The epidemiology and spread of drug resistant human influenza viruses. Curr Opin Virol 2014; 8:22-9. [PMID: 24866471 DOI: 10.1016/j.coviro.2014.04.009] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 01/25/2023]
Abstract
Significant changes in the circulation of antiviral-resistant influenza viruses have occurred over the last decade. The emergence and continued circulation of adamantane-resistant A(H3N2) and A(H1N1)pdm09 viruses mean that the adamantanes are no longer recommended for use. Resistance to the newer class of drugs, the neuraminidase inhibitors, is typically associated with poorer viral replication and transmission. But 'permissive' mutations, that compensated for impairment of viral function in A(H1N1) viruses during 2007/2008, enabled them to acquire the H275Y NA resistance mutation without fitness loss, resulting in their rapid global spread. Permissive mutations now appear to be present in A(H1N1)pdm09 viruses thereby increasing the risk that oseltamivir-resistant A(H1N1)pdm09 viruses may also spread globally, a concerning scenario given that oseltamivir is the most widely used influenza antiviral.
Collapse
Affiliation(s)
- Aeron C Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, North Melbourne, Victoria 3051, Australia.
| |
Collapse
|