1
|
Cargnin Faccin F, Perez DR. Pandemic preparedness through vaccine development for avian influenza viruses. Hum Vaccin Immunother 2024; 20:2347019. [PMID: 38807261 PMCID: PMC11141480 DOI: 10.1080/21645515.2024.2347019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Influenza A viruses pose a significant threat to global health, impacting both humans and animals. Zoonotic transmission, particularly from swine and avian species, is the primary source of human influenza outbreaks. Notably, avian influenza viruses of the H5N1, H7N9, and H9N2 subtypes are of pandemic concern through their global spread and sporadic human infections. Preventing and controlling these viruses is critical due to their high threat level. Vaccination remains the most effective strategy for influenza prevention and control in humans, despite varying vaccine efficacy across strains. This review focuses specifically on pandemic preparedness for avian influenza viruses. We delve into vaccines tested in animal models and summarize clinical trials conducted on H5N1, H7N9, and H9N2 vaccines in humans.
Collapse
Affiliation(s)
- Flavio Cargnin Faccin
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Daniel R. Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
2
|
Mihaylova NM, Manoylov IK, Nikolova MH, Prechl J, Tchorbanov AI. DNA and protein-generated chimeric molecules for delivery of influenza viral epitopes in mouse and humanized NSG transfer models. Hum Vaccin Immunother 2024; 20:2292381. [PMID: 38193304 DOI: 10.1080/21645515.2023.2292381] [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: 10/14/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Purified subunit viral antigens are weakly immunogenic and stimulate only the antibody but not the T cell-mediated immune response. An alternative approach to inducing protective immunity with small viral peptides may be the targeting of viral epitopes to immunocompetent cells by DNA and protein-engineered vaccines. This review will focus on DNA and protein-generated chimeric molecules carrying engineered fragments specific for activating cell surface co-receptors for inducing protective antiviral immunity. Adjuvanted protein-based vaccine or DNA constructs encoding simultaneously T- and B-cell peptide epitopes from influenza viral hemagglutinin, and scFvs specific for costimulatory immune cell receptors may induce a significant increase of anti-influenza antibody levels and strong CTL activity against virus-infected cells in a manner that mimics the natural infection. Here we summarize the development of several DNA and protein chimeric constructs carrying influenza virus HA317-41 fragment. The generated engineered molecules were used for immunization in intact murine and experimentally humanized NSG mouse models.
Collapse
Affiliation(s)
- Nikolina M Mihaylova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Iliyan K Manoylov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Maria H Nikolova
- National Reference Laboratory of Immunology, National Center of Infectious and Parasitic Diseases, Sofia, Bulgaria
| | | | - Andrey I Tchorbanov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
- National Institute of Immunology, Sofia, Bulgaria
| |
Collapse
|
3
|
Russell CA, Fouchier RAM, Ghaswalla P, Park Y, Vicic N, Ananworanich J, Nachbagauer R, Rudin D. Seasonal influenza vaccine performance and the potential benefits of mRNA vaccines. Hum Vaccin Immunother 2024; 20:2336357. [PMID: 38619079 PMCID: PMC11020595 DOI: 10.1080/21645515.2024.2336357] [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: 11/13/2023] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Influenza remains a public health threat, partly due to suboptimal effectiveness of vaccines. One factor impacting vaccine effectiveness is strain mismatch, occurring when vaccines no longer match circulating strains due to antigenic drift or the incorporation of inadvertent (eg, egg-adaptive) mutations during vaccine manufacturing. In this review, we summarize the evidence for antigenic drift of circulating viruses and/or egg-adaptive mutations occurring in vaccine strains during the 2011-2020 influenza seasons. Evidence suggests that antigenic drift led to vaccine mismatch during four seasons and that egg-adaptive mutations caused vaccine mismatch during six seasons. These findings highlight the need for alternative vaccine development platforms. Recently, vaccines based on mRNA technology have demonstrated efficacy against SARS-CoV-2 and respiratory syncytial virus and are under clinical evaluation for seasonal influenza. We discuss the potential for mRNA vaccines to address strain mismatch, as well as new multi-component strategies using the mRNA platform to improve vaccine effectiveness.
Collapse
Affiliation(s)
- Colin A. Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ron A. M. Fouchier
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
4
|
Pelz L, Dogra T, Marichal-Gallardo P, Hein MD, Hemissi G, Kupke SY, Genzel Y, Reichl U. Production of antiviral "OP7 chimera" defective interfering particles free of infectious virus. Appl Microbiol Biotechnol 2024; 108:97. [PMID: 38229300 DOI: 10.1007/s00253-023-12959-6] [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: 08/21/2023] [Revised: 10/26/2023] [Accepted: 11/05/2023] [Indexed: 01/18/2024]
Abstract
Defective interfering particles (DIPs) of influenza A virus (IAV) are suggested for use as broad-spectrum antivirals. We discovered a new type of IAV DIP named "OP7" that carries point mutations in its genome segment (Seg) 7 instead of a deletion as in conventional DIPs (cDIPs). Recently, using genetic engineering tools, we generated "OP7 chimera DIPs" that carry point mutations in Seg 7 plus a deletion in Seg 1. Together with cDIPs, OP7 chimera DIPs were produced in shake flasks in the absence of infectious standard virus (STV), rendering UV inactivation unnecessary. However, only part of the virions harvested were OP7 chimera DIPs (78.7%) and total virus titers were relatively low. Here, we describe the establishment of an OP7 chimera DIP production process applicable for large-scale production. To increase total virus titers, we reduced temperature from 37 to 32 °C during virus replication. Production of almost pure OP7 chimera DIP preparations (99.7%) was achieved with a high titer of 3.24 log10(HAU/100 µL). This corresponded to an 11-fold increase relative to the initial process. Next, this process was transferred to a stirred tank bioreactor resulting in comparable yields. Moreover, DIP harvests purified and concentrated by steric exclusion chromatography displayed an increased interfering efficacy in vitro. Finally, a perfusion process with perfusion rate control was established, resulting in a 79-fold increase in total virus yields compared to the original batch process in shake flasks. Again, a very high purity of OP7 chimera DIPs was obtained. This process could thus be an excellent starting point for good manufacturing practice production of DIPs for use as antivirals. KEY POINTS: • Scalable cell culture-based process for highly effective antiviral OP7 chimera DIPs • Production of almost pure OP7 chimera DIPs in the absence of infectious virus • Perfusion mode production and purification train results in very high titers.
Collapse
Affiliation(s)
- Lars Pelz
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Tanya Dogra
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Pavel Marichal-Gallardo
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Marc Dominique Hein
- Otto Von Guericke University Magdeburg, Bioprocess Engineering, Magdeburg, Germany
| | - Ghada Hemissi
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Sascha Young Kupke
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Otto Von Guericke University Magdeburg, Bioprocess Engineering, Magdeburg, Germany
| |
Collapse
|
5
|
Aydillo T, Balsera-Manzanero M, Rojo-Fernandez A, Escalera A, Salamanca-Rivera C, Pachón J, Del Mar Muñoz-García M, Sánchez-Cordero MJ, Sánchez-Céspedes J, García-Sastre A, Cordero E. Concomitant administration of seasonal influenza and COVID-19 mRNA vaccines. Emerg Microbes Infect 2024; 13:2292068. [PMID: 38054302 PMCID: PMC10798284 DOI: 10.1080/22221751.2023.2292068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/03/2023] [Indexed: 12/07/2023]
Abstract
Current clinical guidelines support the concomitant administration of seasonal influenza vaccines and COVID-19 mRNA boosters vaccine. Whether dual vaccination may impact vaccine immunogenicity due to an interference between influenza or SARS-CoV-2 antigens is unknown. We aimed to understand the impact of mRNA COVID-19 vaccines administered concomitantly on the immune response to influenza vaccines. For this, 128 volunteers were vaccinated during the 22-23 influenza season. Three groups of vaccination were assembled: FLU vaccine only (46, 35%) versus volunteers that received the mRNA bivalent COVID-19 vaccines concomitantly to seasonal influenza vaccines, FluCOVID vaccine in the same arm (42, 33%) or different arm (40, 31%), respectively. Sera and whole blood were obtained the day of vaccination, +7, and +28 days after for antibody and T cells response quantification. As expected, side effects were increased in individuals who received the FluCOVID vaccine as compared to FLU vaccine only based on the known reactogenicity of mRNA vaccines. In general, antibody levels were high at 4 weeks post-vaccination and differences were found only for the H3N2 virus when administered in different arms compared to the other groups at day 28 post-vaccination. Additionally, our data showed that subjects that received the FluCOVID vaccine in different arm tended to have better antibody induction than those receiving FLU vaccines for H3N2 virus in the absence of pre-existing immunity. Furthermore, no notable differences in the influenza-specific cellular immune response were found for any of the vaccination groups. Our data supports the concomitant administration of seasonal influenza and mRNA COVID-19 vaccines.
Collapse
Affiliation(s)
- Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn School of Medicine at Mount Sinai, Global Health and Emerging Pathogens Institute, New York, USA
| | - Maria Balsera-Manzanero
- Viral Diseases and Infections in Immunodeficiencies Research Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
| | - Amaya Rojo-Fernandez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn School of Medicine at Mount Sinai, Global Health and Emerging Pathogens Institute, New York, USA
| | - Alba Escalera
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Celia Salamanca-Rivera
- Unit of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Sevilla, Spain
- Department of Preventive Medicine, University of Seville, Spain
| | - Jerónimo Pachón
- Viral Diseases and Infections in Immunodeficiencies Research Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Department of Medicine, School of Medicine, University of Sevilla, Sevilla, Spain
| | | | | | - Javier Sánchez-Céspedes
- Viral Diseases and Infections in Immunodeficiencies Research Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Unit of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Sevilla, Spain
- CIBERINFEC, CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn School of Medicine at Mount Sinai, Global Health and Emerging Pathogens Institute, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, USA
| | - Elisa Cordero
- Viral Diseases and Infections in Immunodeficiencies Research Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Unit of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Sevilla, Spain
- CIBERINFEC, CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, School of Medicine, University of Sevilla, Sevilla, Spain
| |
Collapse
|
6
|
van der Pol S, Postma MJ, Boersma C. Antivirals to prepare for surges in influenza cases: an economic evaluation of baloxavir marboxil for the Netherlands. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2024; 25:1557-1567. [PMID: 38483666 PMCID: PMC11512865 DOI: 10.1007/s10198-024-01683-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 02/06/2024] [Indexed: 10/27/2024]
Abstract
OBJECTIVES We perform a cost-effectiveness analysis (CEA) and budget impact analysis (BIA) of baloxavir marboxil compared to current care in the Netherlands for patients at risk of influenza-related complications, including patients with comorbidities and the elderly. METHODS In the CEA, a decision tree model was developed to assess the cost-effectiveness of baloxavir marboxil for a cohort of 52-year-olds from a societal perspective. A lifetime horizon was taken by incorporating the quality-adjusted life expectancy. The BIA included different epidemiological scenarios, estimating different plausible epidemiological scenarios for seasonal influenza considering the whole Dutch population with an increased risk of influenza complications. RESULTS The base-case ICER was estimated to be €8,300 per QALY. At the willingness-to-pay threshold of €20,000 per QALY, the probability of being cost effective was 58%. The base-case expected budget impact was €5.7 million on average per year, ranging from €1.5 million to €10.5 million based on the severity of the influenza epidemic and vaccine effectiveness. CONCLUSION In the Netherlands, baloxavir is a cost-effective treatment option for seasonal influenza, with a base-case ICER of €8,300 per QALY for the population aged 60 years and over and patients at high risk of influenza-related complications. For a large part, this ICER is driven by the reduction of the illness duration of influenza and productivity gains in the working population.
Collapse
Affiliation(s)
- Simon van der Pol
- Health-Ecore, Utrechtseweg 60, 3704 HE, Zeist, The Netherlands.
- Department of Health Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Maarten J Postma
- Health-Ecore, Utrechtseweg 60, 3704 HE, Zeist, The Netherlands
- Department of Health Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Economics, Econometrics & Finance, University of Groningen, Groningen, The Netherlands
| | - Cornelis Boersma
- Health-Ecore, Utrechtseweg 60, 3704 HE, Zeist, The Netherlands
- Department of Health Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Management Sciences, Open University, Heerlen, The Netherlands
| |
Collapse
|
7
|
Xu S, Yang J. A deterministic model for homologous antibody dependant enhancement on influenza infection. Infect Dis Model 2024; 9:1265-1275. [PMID: 39183947 PMCID: PMC11342263 DOI: 10.1016/j.idm.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Antibody dependant enhancement refers that viral infectivity was unexpectedly enhanced at low antibody concentration compared to when antibodies were absent, such as Dengue, Zika and influenza virus. To mathematically describe switch from enhancement to neutralisation with increase of antibody concentration, one hyperbolic tangent variant is used as switching function in existed models. However, switching function with hyperbolic tangent contains four parameters, and does not always increase with antibody concentration. To address this problem, we proposed a monotonically increasing Logistical function variant as switching function, which only contains position parameter and magnitude parameter. Analysing influenza viral titre estimated from 21 focus reduction assay (FRA) datasets from neutralisation group (viral titre lower than negative control on all serial dilutions) and 20 FRA dataset from enhancement group (viral titre higher than negative control on high serial dilution), switching function with Logistic function performs better than existed model independent of both groups and exhibited different behaviour/character; specifically, magnitude parameter estimated from enhancement group is lower, but position parameter estimated from enhancement group is higher. A lower magnitude parameter refers that enhancement group more rapidly switches from enhancement to neutralisation with increase of antibody concentration, and a higher position parameter indicates that enhancement group provides a larger antibody concentration interval corresponding to enhancement. Integrating estimated neutralisation kinetics with viral replication, we demonstrated that antibody-induced bistable influenza kinetics exist independent of both groups. However, comparing with neutralisation group, enhancement group provides higher threshold value of antibody concentration corresponding to influenza infectivity. This explains the observed phenomenon that antibody dependent enhancement enhances susceptibility, severity, and mortality to influenza infection. On population level, antibody dependant enhancement can promote H1N1 and H3N2 influenza virus cooperate to sustain long-term circulation on human populations according to antigenic seniority theory.
Collapse
Affiliation(s)
- Shilian Xu
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, 3086, Australia
- Department of Mathematical and Physical Sciences, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Jiaru Yang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- The Institute for Tropical Medicine, School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, China
| |
Collapse
|
8
|
Çalışkan DM, Kumar S, Hinse S, Schughart K, Wiewrodt R, Fischer S, Krueger V, Wiebe K, Barth P, Mellmann A, Ludwig S, Brunotte L. Molecular characterisation of influenza B virus from the 2017/18 season in primary models of the human lung reveals improved adaptation to the lower respiratory tract. Emerg Microbes Infect 2024; 13:2402868. [PMID: 39248230 PMCID: PMC11421153 DOI: 10.1080/22221751.2024.2402868] [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: 05/24/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
The 2017/18 influenza season was characterized by unusual high numbers of severe infections and hospitalizations. Instead of influenza A viruses, this season was dominated by infections with influenza B viruses of the Yamagata lineage. While this IBV/Yam dominance was associated with a vaccine mismatch, a contribution of virus intrinsic features to the clinical severity of the infections was speculated. Here, we performed a molecular and phenotypic characterization of three IBV isolates from patients with severe flu symptoms in 2018 and compared it to an IBV/Yam isolate from 2016 using experimental models of increasing complexity, including human lung explants, lung organoids, and alveolar macrophages. Viral genome sequencing revealed the presence of clade but also isolate specific mutations in all viral genes, except NP, M1, and NEP. Comparative replication kinetics in different cell lines provided further evidence for improved replication fitness, tolerance towards higher temperatures, and the development of immune evasion mechanisms by the 2018 IBV isolates. Most importantly, immunohistochemistry of infected human lung explants revealed an impressively altered cell tropism, extending from AT2 to AT1 cells and macrophages. Finally, transcriptomics of infected human lung explants demonstrated significantly reduced amounts of type I and type III IFNs by the 2018 IBV isolate, supporting the existence of additional immune evasion mechanisms. Our results show that the severeness of the 2017/18 Flu season was not only the result of a vaccine mismatch but was also facilitated by improved adaptation of the circulating IBV strains to the environment of the human lower respiratory tract.
Collapse
Affiliation(s)
- Duygu Merve Çalışkan
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Sriram Kumar
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Saskia Hinse
- Institute of Virology, University of Münster, Münster, Germany
| | - Klaus Schughart
- Institute of Virology, University of Münster, Münster, Germany
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rainer Wiewrodt
- Department of Medicine A, Haematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- Department of Respiratory Medicine and Thoracic Oncology, Foundation Mathias Spital, Rheine and Ibbenbueren, Germany
| | - Stefan Fischer
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- Department of Respiratory Medicine and Thoracic Oncology, Foundation Mathias Spital, Rheine and Ibbenbueren, Germany
| | - Vera Krueger
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- Department of Respiratory Medicine and Thoracic Oncology, Foundation Mathias Spital, Rheine and Ibbenbueren, Germany
| | - Karsten Wiebe
- Department of Thoracic Surgery, University Hospital Münster, Muenster, Germany
| | - Peter Barth
- Gerhard-Domagk-Institute of Pathology, University of Münster, Muenster, Germany
| | | | - Stephan Ludwig
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Linda Brunotte
- Institute of Virology, University of Münster, Münster, Germany
| |
Collapse
|
9
|
Wang X, Pu F, Yang X, Feng X, Zhang J, Duan K, Nian X, Ma Z, Ma XX, Yang XM. Immunosuppressants exert antiviral effects against influenza A(H1N1)pdm09 virus via inhibition of nucleic acid synthesis, mRNA splicing, and protein stability. Virulence 2024; 15:2301242. [PMID: 38170681 PMCID: PMC10854267 DOI: 10.1080/21505594.2023.2301242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Influenza A virus (IAV) poses a threat to patients receiving immunosuppressive medications since they are more susceptible to infection with severe symptoms, and even death. Understanding the direct effects of immunosuppressants on IAV infection is critical for optimizing immunosuppression in these patients who are infected or at risk of influenza virus infection. We profiled the effects of 10 immunosuppressants, explored the antiviral mechanisms of immunosuppressants, and demonstrated the combined effects of immunosuppressants with the antiviral drug oseltamivir in IAV-infected cell models. We found that mycophenolic acid (MPA) strongly inhibits viral RNA replication via depleting cellular guanosine pool. Treatment with 6-Thioguanine (6-TG) promoted viral protein degradation through a proteasomal pathway. Filgotinib blocked mRNA splicing of matrix protein 2, resulting in decreased viral particle assembly. Furthermore, combined treatment with immunosuppressants and oseltamivir inhibits IAV viral particle production in an additive or synergic manner. Our results suggest that MPA, 6-TG, and filgotinib could be the preferential choices for patients who must take immunosuppressants but are at risk of influenza virus infection.
Collapse
Affiliation(s)
- Xin Wang
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Feiyang Pu
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Xuanye Yang
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Xili Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
- Wuhan Institute of Biological Products Co, Ltd, Wuhan, China
| | - Kai Duan
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
- Wuhan Institute of Biological Products Co, Ltd, Wuhan, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
- Wuhan Institute of Biological Products Co, Ltd, Wuhan, China
| | - Zhongren Ma
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Xiao-Xia Ma
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Xiao-Ming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan, China
- China National Biotech Group Company Limited, Beijing, China
| |
Collapse
|
10
|
Hamza H, Ghosh M, Löffler MW, Rammensee HG, Planz O. Identification and relative abundance of naturally presented and cross-reactive influenza A virus MHC class I-restricted T cell epitopes. Emerg Microbes Infect 2024; 13:2306959. [PMID: 38240239 PMCID: PMC10854457 DOI: 10.1080/22221751.2024.2306959] [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: 12/13/2023] [Accepted: 01/14/2024] [Indexed: 02/10/2024]
Abstract
Cytotoxic T lymphocytes are key for controlling viral infection. Unravelling CD8+ T cell-mediated immunity to distinct influenza virus strains and subtypes across prominent HLA types is relevant for combating seasonal infections and emerging new variants. Using an immunopeptidomics approach, naturally presented influenza A virus-derived ligands restricted to HLA-A*24:02, HLA-A*68:01, HLA-B*07:02, and HLA-B*51:01 molecules were identified. Functional characterization revealed multifunctional memory CD8+ T cell responses for nine out of sixteen peptides. Peptide presentation kinetics was optimal around 12 h post infection and presentation of immunodominant epitopes shortly after infection was not always persistent. Assessment of immunogenic epitopes revealed that they are highly conserved across the major zoonotic reservoirs and may contain a single substitution in the vicinity of the anchor residues. These findings demonstrate how the identified epitopes promote T cell pools, possibly cross-protective in individuals and can be potential targets for vaccination.
Collapse
Affiliation(s)
- Hazem Hamza
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Virology Laboratory, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Michael Ghosh
- Institute for Immunology, University of Tübingen, Tübingen, Germany
| | - Markus W Löffler
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, Tübingen, Germany
- Centre for Clinical Transfusion Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Cluster of Excellence CMFI (EXC2124) "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Oliver Planz
- Institute for Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Blackmore EN, Lloyd-Smith JO. Reply to Uzoigwe: Modeling and the historical record. Proc Natl Acad Sci U S A 2024; 121:e2417598121. [PMID: 39480846 DOI: 10.1073/pnas.2417598121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Affiliation(s)
- Elizabeth N Blackmore
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520
| | - James O Lloyd-Smith
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095
| |
Collapse
|
12
|
Wang H, Fu H, Zhai L, Le J, Guo B, Zhou Y, Ji C, Li D, Zhang Y. Nanocrystalline alloy-mediated delivery of mosaic epitope peptides for universal influenza vaccine. J Mater Chem B 2024. [PMID: 39494737 DOI: 10.1039/d4tb00742e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Seasonal influenza infection poses great threat to public health systems. The flu vaccine remains the most effective method to reduce transmission and mortality. However, its effectiveness is limited due to the challenges in protecting against all influenza variants, as well as the weaker immune response observed in the adult population. Here, combining machine learning, synchrotron small angle X-ray scattering, we design an adjuvanted influenza vaccine composing mosaic epitope peptides selected from the hemagglutinin proteins of influenza A and B virus. These epitopes share similar physiochemical properties cognate to host antimicrobial peptides (AMPs) allowing them to form supramolecular assembly with poly(I:C), a synthetic toll-like receptor 3 (TLR3) agonist, through electrostatic interaction. The poly(I:C) is arranged into columnar lattice with the average inter-poly(I:C) distance commensurate with TLR3 and thereby capable of inducing multivalent TLR3 binding and hyperactivating the downstream inflammatory pathway. Interestingly, multiple AMP-like epitopes (Ampitopes) with compatible lattice parameter can co-crystalize into the same lattice to form 'alloy'-like composite with better poly(I:C) arrangement which allows the co-delivery of mosaic Ampitopes. The designed Ampitope-poly(I:C) nanocrystalline (and alloy) successfully activates interferon regulatory factor 3 (IRF3)-mediated pathway in antigen presenting cells. The intramuscular delivery of the nanocrystalline to the mice strongly trigger IL-6 and IFN-α release, which well-mimics the cytokines release pattern in influenza infected patients. After the third boost, the antigen-specific T cell response is 55 times higher compared to the free Ampitopes treatment group. Together, this vaccine offers a versatile way of eliciting strong and broad anti-flu protection.
Collapse
Affiliation(s)
- Hongyu Wang
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Han Fu
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Liyan Zhai
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Jiaqing Le
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Bohan Guo
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Yuyang Zhou
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Dapeng Li
- Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, Zhejiang 310012, China
- Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang 310012, China
| | - Yue Zhang
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang 310030, China.
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
13
|
Marchenko VY, Panova AS, Kolosova NP, Gudymo AS, Svyatchenko SV, Danilenko AV, Vasiltsova NN, Egorova ML, Onkhonova GS, Zhestkov PD, Zinyakov NG, Andreychuk DB, Chvala IA, Kosenko MN, Moiseeva AA, Boldyrev ND, Shadrinova KN, Perfilieva ON, Ryzhikov AB. Characterization of H5N1 avian influenza virus isolated from bird in Russia with the E627K mutation in the PB2 protein. Sci Rep 2024; 14:26490. [PMID: 39489822 PMCID: PMC11532466 DOI: 10.1038/s41598-024-78175-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
Currently A(H5Nx) avian influenza viruses are globally widespread and continue to evolve. Since their emergence in 2020 novel highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b reassortant viruses have become predominant in the world and caused multiple infections in mammals. It was shown that some of A(H5N1) viruses mostly isolated from mammals contain an E627K mutation in the PB2 protein which can lead to adaptation of influenza viruses to mammalian cells. In 2023 in Russia we have isolated two highly pathogenic avian influenza A(H5N1) clade 2.3.4.4b viruses from birds one of which contained an E627K mutation in the PB2 protein. This virus had increased virulence in mice. Limited airborne transmission of the virus with the PB2-E627K mutation was observed between ferrets, in which infectious virus was detected in the nasal washings of the three of the twelve recipient ferrets, and clinical symptoms of the disease were observed in one case. Both viruses showed dominant binding to avian-type sialoside receptors, which was most likely the reason for the limited transmissibility. Thus, this study indicates a possible limited increase in the pandemic potential of A(H5N1) 2.3.4.4b viruses and highlights the importance of continuous avian influenza surveillance for pandemic preparedness and response.
Collapse
Affiliation(s)
- Vasiliy Yu Marchenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia.
| | - Anastasia S Panova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Natalia P Kolosova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Andrey S Gudymo
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Svetlana V Svyatchenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Alexey V Danilenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Natalia N Vasiltsova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Marina L Egorova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Galina S Onkhonova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Pavel D Zhestkov
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Nikolay G Zinyakov
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Dmitriy B Andreychuk
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Ilya A Chvala
- Federal Centre for Animal Health FGBI "ARRIAH", Rosselkhoznadzor, Vladimir, 600901, Russia
| | - Maksim N Kosenko
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Anastasia A Moiseeva
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Nikita D Boldyrev
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Kiunnei N Shadrinova
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Olga N Perfilieva
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| | - Alexander B Ryzhikov
- FBRI State Research Centre of Virology and Biotechnology "Vector", Rospotrebnadzor, Koltsovo, Novosibirsk, 630559, Russia
| |
Collapse
|
14
|
Li YY, Liang GD, Chen ZX, Zhang K, Liang JL, Jiang LR, Yang SZ, Jiang F, Liu SW, Yang J. A small molecule compound targeting hemagglutinin inhibits influenza A virus and exhibits broad-spectrum antiviral activity. Acta Pharmacol Sin 2024; 45:2380-2393. [PMID: 38987389 PMCID: PMC11489770 DOI: 10.1038/s41401-024-01331-7] [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: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/12/2024] Open
Abstract
Influenza A virus (IAV) is a widespread pathogen that poses a significant threat to human health, causing pandemics with high mortality and pathogenicity. Given the emergence of increasingly drug-resistant strains of IAV, currently available antiviral drugs have been reported to be inadequate to meet clinical demands. Therefore, continuous exploration of safe, effective and broad-spectrum antiviral medications is urgently required. Here, we found that the small molecule compound J1 exhibited low toxicity both in vitro and in vivo. Moreover, J1 exhibits broad-spectrum antiviral activity against enveloped viruses, including IAV, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human coronavirus OC43 (HCoV-OC43), herpes simplex virus type 1 (HSV-1) and HSV-2. In this study, we explored the inhibitory effects and mechanism of action of J1 on IAV in vivo and in vitro. The results showed that J1 inhibited infection by IAV strains, including H1N1, H7N9, H5N1 and H3N2, as well as by oseltamivir-resistant strains. Mechanistic studies have shown that J1 blocks IAV infection mainly through specific interactions with the influenza virus hemagglutinin HA2 subunit, thereby blocking membrane fusion. BALB/c mice were used to establish a model of acute lung injury (ALI) induced by IAV. Treatment with J1 increased survival rates and reduced viral titers, lung index and lung inflammatory damage in virus-infected mice. In conclusion, J1 possesses significant anti-IAV effects in vitro and in vivo, providing insights into the development of broad-spectrum antivirals against future pandemics.
Collapse
Affiliation(s)
- Yin-Yan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guo-Dong Liang
- Key Laboratory for Candidate Drug Design and Screening Based on Chemical Biology, College of Pharmacy, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Zhi-Xuan Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ke Zhang
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang, 561113, China
| | - Jin-Long Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin-Rui Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Si-Zu Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Feng Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Wen Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jie Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
de la Cueva IS, Gerber JE, Hastie A, Brotons C, Panzer F, Pirçon JY, Talsma P, Eckermann T, Nikic V, Gomez XM, Alsdurf H. Enhanced Safety Surveillance of GSK's Inactivated Quadrivalent Seasonal Influenza Vaccine in Belgium, Germany, and Spain During the 2022/2023 Influenza Season. Drug Saf 2024; 47:1137-1148. [PMID: 38949714 PMCID: PMC11485106 DOI: 10.1007/s40264-024-01456-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Seasonal influenza is prevented through annual vaccination, especially in children and older adults. These vaccines are annually updated based on World Health Organization recommendations and require continuous safety monitoring. OBJECTIVE We assessed the frequency and severity of adverse events within 7 days of administering GSK's inactivated quadrivalent seasonal influenza vaccine (IIV4) in Belgium, Germany, and Spain during the 2022/2023 influenza season. METHODS In this enhanced safety surveillance study, adults who received GSK's IIV4 and parents/guardians/legally acceptable representatives of vaccinated children (aged 6 months-17 years) were invited to complete adverse drug reaction cards reporting adverse events within 7 days post-vaccination. RESULTS In total, 1332 participants (53.6% female) received at least one dose of GSK's IIV4, including 43 children who received two doses. Overall, 97.8% of adverse drug reaction cards were completed and returned in the study. All participants in Belgium were adults, while 54.7% and 7.4% in Spain and Germany, respectively, were pediatric participants aged 6 months-17 years. After Dose 1, across all age groups, 49.8% of participants reported at least one adverse event. The most common adverse events (cumulative frequency >5%) following Dose 1 were injection-site pain (37.6%), fatigue (15.0%), headache (13.2%), injection-site swelling (9.3%), myalgia (7.6%), and injection-site erythema (7.4%). Across all countries, adverse events were most common in adults aged 18-65 years (59.7%), followed by those aged 3-17 years (47.0%), >65 years (35.7%), and 6-35 months (23.5%). After Dose 2, 18.6% of participants reported at least one adverse event, with general disorders and administration site conditions again being the most frequent. CONCLUSIONS Across all age and risk groups for serious disease, no serious adverse events related to GSK's IIV4 were reported within 7 days post-vaccination. This study supports and confirms the acceptable safety profile of GSK's IIV4 across all recommended age groups. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov number: not applicable.
Collapse
Affiliation(s)
| | | | - Andrew Hastie
- GSK, 14200 Shady Grove Road, Rockville, MD, 20850, USA
| | - Carlos Brotons
- Biomedical Research Institute Sant Pau, EAP Sardenya, Barcelona, Spain
| | | | | | - Paul Talsma
- Statistics Department, Vivos Technology Limited (Phastar), London, UK
| | | | - Vanja Nikic
- GSK, 14200 Shady Grove Road, Rockville, MD, 20850, USA
| | - Xavier Martinez Gomez
- Department of Preventive Medicine, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | |
Collapse
|
16
|
Hohensee L, Scheibner D, Luttermann C, Shelton H, Dorhoi A, Abdelwhab EM, Blohm U. PB1-F2 of low pathogenicity H7N7 restricts apoptosis in avian cells. Virus Res 2024; 349:199444. [PMID: 39089370 PMCID: PMC11386312 DOI: 10.1016/j.virusres.2024.199444] [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: 04/15/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/03/2024]
Abstract
Avian influenza viruses (AIV) pose a continuous challenge to global health and economy. While countermeasures exist to control outbreaks in poultry, the persistent circulation of AIV in wild aquatic and shorebirds presents a significant challenge to effective disease prevention efforts. PB1-F2 is a non-structural protein expressed from a second open reading frame (+1) of the polymerase basic 1 (PB1) segment. The sequence and length of the PB1-F2 protein can vary depending on the host of origin. While avian isolates typically carry full-length PB1-F2, isolates from mammals, often express truncated forms. The selective advantage of the full-length PB1-F2 in avian isolates is not fully understood. Most research on the role of PB1-F2 in influenza virus replication has been conducted in mammalian systems, where PB1-F2 interfered with the host immune response and induced apoptosis. Here, we used Low Pathogenicity (LP) AIV H7N7 expressing full-length PB1-F2 as well as a knockout mutant. We found that the full-length PB1-F2 of LPAIV prolonged survival of infected cells by limiting apoptotic cell death. Furthermore, PB1-F2 knockout LPAIV significantly decreased MHC-I expression on fibroblasts, delayed tissue healing and increased phagocytic uptake of infected cells, whereas LPAIV expressing PB1-F2 has limited effects. These findings indicate that full-length PB1-F2 enables AIV to cause prolonged infections without severely harming the avian host. Our observations may explain maintenance of AIV in the natural bird reservoir in absence of severe clinical signs.
Collapse
Affiliation(s)
- Luise Hohensee
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany
| | - David Scheibner
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany; Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany
| | - Christine Luttermann
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany
| | - Holly Shelton
- The Pirbright Institute, Pirbright, Ash Road, Surrey GU24 0NF, United Kingdom
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany
| | - Elsayed M Abdelwhab
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany
| | - Ulrike Blohm
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, Greifswald, Insel Riems 17493, Germany.
| |
Collapse
|
17
|
Panthi S, Hong JY, Satange R, Yu CC, Li LY, Hou MH. Antiviral drug development by targeting RNA binding site, oligomerization and nuclear export of influenza nucleoprotein. Int J Biol Macromol 2024; 282:136996. [PMID: 39486729 DOI: 10.1016/j.ijbiomac.2024.136996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
The quasispecies of the influenza virus poses a significant challenge for developing effective therapies. Current antiviral drugs such as oseltamivir, zanamivir, peramivir and baloxavir marboxil along with seasonal vaccines have limitations due to viral variability caused by antigenic drift and shift as well as the development of drug resistance. Therefore, there is a clear need for novel antiviral agents targeting alternative mechanisms, either independently or in combination with existing modalities, to reduce the impact of influenza virus-related infections. The influenza nucleoprotein (NP) is a key component of the viral ribonucleoprotein complex. The multifaceted nature of the NP makes it an attractive target for antiviral intervention. Recent reports have identified inhibitors that specifically target this protein. Recognizing the importance of developing influenza treatments for potential pandemics, this review explores the structural and functional aspects of NP and highlights its potential as an emerging target for anti-influenza drugs. We discuss various strategies for targeting NP, including RNA binding, oligomerization, and nuclear export, and also consider the potential of NP-based vaccines. Overall, this review provides insights into recent developments and future perspectives on targeting influenza NP for antiviral therapies.
Collapse
Affiliation(s)
- Sankar Panthi
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Jhen-Yi Hong
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Roshan Satange
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Ching-Ching Yu
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Long-Yuan Li
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.
| | - Ming-Hon Hou
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan; Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
18
|
Guzman Ruiz L, Zollner AM, Hoxie I, Küchler J, Hausjell C, Mesurado T, Krammer F, Jungbauer A, Pereira Aguilar P, Klausberger M, Grabherr R. Enhancing NA immunogenicity through novel VLP designs. Vaccine 2024; 42:126270. [PMID: 39197219 DOI: 10.1016/j.vaccine.2024.126270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Current influenza virus vaccines poorly display key neuraminidase (NA) epitopes and do not robustly induce NA-reactive antibodies; instead, they focus on the induction of hemagglutinin (HA)-reactive antibodies. Next-generation influenza vaccines should be optimized in order to activate NA-reactive B cells and to induce a broadly cross-reactive and protective antibody response. We aimed at enhancing the immunogenicity of the NA on vaccines by two strategies: (i) modifying the HA:NA ratio of the vaccine preparation and (ii) exposing epitopes on the lateral surface or beneath the head of the NA by extending the NA stalk. The H1N1 glycoproteins from the influenza virus A/California/04/2009 strain were displayed on human immunodeficiency virus 1 (HIV-1) gag-based virus-like particles (VLP). Using the baculovirus insect cell expression system, we biased the quantity of surface glycoproteins employing two different promoters, the very late baculovirus p10 promoter and the early and late gp64 promoter. This led to a 1:1 to 2:1 HA:NA ratio, which was approximately double or triple the amount of NA as present on the wild-type influenza A virus (HA:NA ratio 3:1 to 5:1). Furthermore, by insertion of 15 amino acids from the A-New York/61/2012 strain (NY12) which prolongates the NA stalk (NA long stalk; NA-LS), we intended to improve the accessibility of the NA. Six different types of VLPs were produced and purified using a platform downstream process based on Capto-Core 700™ followed by Capto-Heparin™ affinity chromatography combined with ultracentrifugation. These VLPs were then tested in a mouse model. Robust titers of antibodies that inhibit the neuraminidase activity were elicited even after vaccination with two low doses (0.3 μg) of the H1N1 VLPs without compromising the anti-HA responses. In conclusion, our results demonstrate the feasibility of the two developed strategies to retain HA immunogenicity and improve NA immunogenicity as a future influenza vaccine candidate.
Collapse
Affiliation(s)
- Leticia Guzman Ruiz
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria; University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Alexander M Zollner
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Irene Hoxie
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, Gustave L. Levy Place, 10029-5674 New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jan Küchler
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Christina Hausjell
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria
| | - Tomas Mesurado
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, Gustave L. Levy Place, 10029-5674 New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Alois Jungbauer
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria
| | - Patricia Pereira Aguilar
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria
| | - Miriam Klausberger
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria
| | - Reingard Grabherr
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
19
|
Zheng R, Yu C, Yao D, Cai M, Zhang L, Ye F, Huang X. Engineering Stimuli-Responsive Materials for Precision Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406439. [PMID: 39444066 DOI: 10.1002/smll.202406439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Over the past decade, precision medicine has garnered increasing attention, making significant strides in discovering new therapeutic drugs and mechanisms, resulting in notable achievements in symptom alleviation, pain reduction, and extended survival rates. However, the limited target specificity of primary drugs and inter-individual differences have often necessitated high-dosage strategies, leading to challenges such as restricted deep tissue penetration rates and systemic side effects. Material science advancements present a promising avenue for these issues. By leveraging the distinct internal features of diseased regions and the application of specific external stimuli, responsive materials can be tailored to achieve targeted delivery, controllable release, and specific biochemical reactions. This review aims to highlight the latest advancements in stimuli-responsive materials and their potential in precision medicine. Initially, we introduce disease-related internal stimuli and capable external stimuli, elucidating the reaction principles of responsive functional groups. Subsequently, we provide a detailed analysis of representative pre-clinical achievements of stimuli responsive materials across various clinical applications, including enhancements in the treatment of cancers, injury diseases, inflammatory diseases, infection diseases, and high-throughput microfluidic biosensors. Finally, we discuss some clinical challenges, such as off-target effects, long-term impacts of nano-materials, potential ethical concerns, and offer insights into future perspectives of stimuli-responsive materials.
Collapse
Affiliation(s)
- Ruixuan Zheng
- Joint Centre of Translational Medicine, Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Wenzhou, Zhejiang, 325000, China
| | - Chang Yu
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Wenzhou, Zhejiang, 325000, China
- Intervention Department, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Dan Yao
- Joint Centre of Translational Medicine, Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Wenzhou, Zhejiang, 325000, China
| | - Mengsi Cai
- Joint Centre of Translational Medicine, Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Wenzhou, Zhejiang, 325000, China
| | - Lexiang Zhang
- Joint Centre of Translational Medicine, Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Fangfu Ye
- Joint Centre of Translational Medicine, Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaoying Huang
- Joint Centre of Translational Medicine, Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
20
|
Huang QJ, Kim R, Song K, Grigorieff N, Munro JB, Schiffer CA, Somasundaran M. Virion-associated influenza hemagglutinin clusters upon sialic acid binding visualized by cryo-electron tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618557. [PMID: 39463923 PMCID: PMC11507849 DOI: 10.1101/2024.10.15.618557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Influenza viruses are enveloped, negative sense single-stranded RNA viruses covered in a dense layer of glycoproteins. Hemagglutinin (HA) accounts for 80-90% of influenza glycoprotein and plays a role in host cell binding and membrane fusion. While previous studies have characterized structures of receptor-free and receptor-bound HA in vitro, the effect of receptor binding on HA organization and structure on virions remains unknown. Here, we used cryo-electron tomography (cryoET) to visualize influenza virions bound to a sialic acid receptor mimic. Overall, receptor binding did not result in significant changes in viral morphology; however, we observed rearrangements of HA trimer organization and orientation. Compared to the even inter-glycoprotein spacing of unliganded HA trimers, receptor binding promotes HA trimer clustering and formation of a triplet of trimers. Subtomogram averaging and refinement yielded 8-10 Å reconstructions that allowed us to visualize specific contacts between HAs from neighboring trimers and identify molecular features that mediate clustering. Taken together, we present new structural evidence that receptor binding triggers clustering of HA trimers, revealing an additional layer of HA dynamics and plasticity.
Collapse
|
21
|
Kong J, Chen L. Gene expression profile analysis of severe influenza-based modulation of idiopathic pulmonary fibrosis. Eur J Med Res 2024; 29:501. [PMID: 39420432 PMCID: PMC11488079 DOI: 10.1186/s40001-024-02107-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND It is known severe influenza infections and idiopathic pulmonary fibrosis (IPF) disease might stimulate each other. Till now, no associated mechanism has been reported. METHOD We collected the genetic pattern of expression of severe influenza (GSE111368) and IPF (GSE70866) from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (C-DEGs) were identified from the two datasets, and using this data, we conducted three forms of analyses, functional annotation, protein-protein interaction (PPI) network and module construction, and hub gene identification and co-expression analysis. RESULTS In all, 174 C-DEGs were selected for additional analyses. Based on our functional analysis, these C-DEGs mediated inflammatory response and cell differentiation. Furthermore, using cytoHubba, we identified 15 genes, namely, MELK, HJURP, BIRC5, TPX2, TK1, CDT1, UBE2C, UHRF1, CCNA2, TYMS, CDCA5, CDCA8, RAD54L, CCNB2, and ITGAM, which served as hub genes to possibly contribute to severe influenza patients with IPF disease as comorbidity. The hub gene expressions were further confirmed using two stand-alone datasets (GSE101702 for severe influenza and GSE10667 for IPF). CONCLUSION Herein, we demonstrated the significance of common pathways and critical genes in severe influenza and IPF etiologies. The identified pathways and genes may be employed as possible therapeutic targets for future therapy against severe influenza patients with IPF.
Collapse
Affiliation(s)
- Jianping Kong
- Department of Nephrology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 211200, China
| | - Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, NO 188 Lingshan North Road, Qixia District, Nanjing, 210046, China.
| |
Collapse
|
22
|
Quach HQ, Haralambieva IH, Goergen KM, Grill DE, Chen J, Ovsyannikova IG, Poland GA, Kennedy RB. Similar humoral responses but distinct CD4 + T cell transcriptomic profiles in older adults elicited by MF59 adjuvanted and high dose influenza vaccines. Sci Rep 2024; 14:24420. [PMID: 39424894 PMCID: PMC11489691 DOI: 10.1038/s41598-024-75250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Older age (≥ 65 years) is associated with impaired responses to influenza vaccination, leading to the preferential recommendation of MF59-adjuvanted (MF59Flu) or high-dose (HDFlu) influenza vaccines for this age group in the United States. Herein, we characterized transcriptomic profiles of CD4+ T cells isolated from 234 recipients (≥ 65 years) of either MF59Flu or HDFlu vaccine, prior to vaccination and 28 days thereafter. We identified 412 and 645 differentially expressed genes (DEGs) in CD4+ T cells of older adults after receiving MF59Flu and HDFlu, respectively. DEGs in CD4+ T cells of MF59Flu recipients were enriched in 14 KEGG pathways, all of which were downregulated. DEGs in CD4+ T cells of HDFlu recipients were enriched in 11 upregulated pathways and 20 downregulated pathways. CD4+ T cells in both vaccine groups shared 50 upregulated genes and 75 downregulated genes, all of which were enriched in 7 KEGG pathways. The remaining 287 and 520 DEGs were specifically associated with MF59Flu and HDFlu, respectively. Unexpectedly, none of these DEGs was significantly correlated with influenza A/H3N2-specific HAI titers, suggesting these DEGs at the individual level may have a limited role in protection against influenza. Our findings emphasize the need for further investigation into other factors influencing immunity against influenza in older adults.
Collapse
Affiliation(s)
- Huy Quang Quach
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Iana H Haralambieva
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Krista M Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Diane E Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Inna G Ovsyannikova
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Gregory A Poland
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard B Kennedy
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
23
|
Elhusseiny MH, Elsayed MM, Mady WH, Mahana O, Bakry NR, Abdelaziz O, Arafa AS, Shahein MA, Eid S, Naguib MM. Genetic features of avian influenza (A/H5N8) clade 2.3.4.4b isolated from quail in Egypt. Virus Res 2024; 350:199482. [PMID: 39396573 PMCID: PMC11532269 DOI: 10.1016/j.virusres.2024.199482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Several genotypes of the highly pathogenic avian influenza (HPAI) virus H5N8 subtype within clade 2.3.4.4b continue to circulate in different species of domestic birds across Egypt. It is believed that quail contribute to virus replication and adaptation to other gallinaceous poultry species and humans. This study provides genetic characterization of the full genome of HPAI H5N8 isolated from quail in Egypt. The virus was isolated from a commercial quail farm associated with respiratory signs. To characterize the genetic features of the detected virus, gene sequencing via Sanger technology and phylogenetic analysis were performed. The results revealed high nucleotide identity with the HPAI H5N8 virus from Egypt, which has multiple basic amino acid motifs PLREKRRKR/GLF at the hemagglutinin (HA) cleavage site. Phylogenetic analysis of the eight gene segments revealed that the quail isolate is grouped with HPAI H5N8 viruses of clade 2.3.4.4b and closely related to the most recent circulating H5N8 viruses in Egypt. Whole-genome characterization revealed amino acid preferences for avian receptors with few mutations, indicating their affinity for human-like receptors and increased virulence in mammals, such as S123P, S133A, T156A and A263T in the HA gene. In addition, the sequencing results revealed a lack of markers associated with influenza antiviral resistance in the neuraminidase and matrix-2 coding proteins. The results of the present study support the spread of HPAIV H5N8 to species other than chickens in Egypt. Therefore, continuous surveillance of AIV in different bird species in Egypt followed by full genomic characterization is needed for better virus control and prevention.
Collapse
Affiliation(s)
- Mohamed H Elhusseiny
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Moataz M Elsayed
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Wesam H Mady
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Osama Mahana
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Neveen R Bakry
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Ola Abdelaziz
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Abdel-Sattar Arafa
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | | | - Samah Eid
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt
| | - Mahmoud M Naguib
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center (ARC), Giza, Egypt; Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK.
| |
Collapse
|
24
|
Lee AJ, Carson S, Reyne MI, Marshall A, Moody D, Allen DM, Allingham P, Levickas A, Fitzgerald A, Bell SH, Lock J, Coey JD, McSparron C, Nejad BF, Troendle EP, Simpson DA, Courtney DG, Einarsson GG, McKenna JP, Fairley DJ, Curran T, McKinley JM, Gilpin DF, Lemon K, McGrath JW, Bamford CGG. Wastewater monitoring of human and avian influenza A viruses in Northern Ireland: a genomic surveillance study. THE LANCET. MICROBE 2024:100933. [PMID: 39395428 DOI: 10.1016/s2666-5247(24)00175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Influenza A viruses (IAVs) are significant pathogens of humans and other animals. Although endemic in humans and birds, novel IAV strains can emerge, jump species, and cause epidemics, like the latest variant of H5N1. Wastewater-based epidemiology (WBE) has been shown capable of detecting human IAVs. We aimed to assess whether whole-genome sequencing (WGS) of IAVs from wastewater is possible and can be used to discriminate between circulating strains of human and any non-human IAVs, such as those of avian origin. METHODS Using a pan-IAV RT-quantitative PCR assay, six wastewater treatment works (WWTWs) across Northern Ireland were screened from Aug 1 to Dec 5, 2022. A nanopore WGS approach was used to sequence RT-qPCR-positive samples. Phylogenetic analysis of sequences relative to currently circulating human and non-human IAVs was performed. For comparative purposes, clinical data (PCR test results) were supplied by The Regional Virus Laboratory, Belfast Health and Social Care Trust (Belfast, Northern Ireland, UK). FINDINGS We detected a dynamic IAV signal in wastewater from Sept 5, 2022, onwards across Northern Ireland, which did not show a clear positive relationship with the clinical data obtained for the region. Meta (mixed strain) whole-genome sequences were generated from wastewater samples displaying homology to only human and avian IAV strains. The relative proportion of IAV reads of human versus avian origin differed across time and sample site. A diversity in subtypes and lineages was detected (eg, H1N1, H3N2, and several avian). Avian segment 8 related to those found in recent H5N1 clade 2.3.4.4b was identified. INTERPRETATION WBE affords a means to monitor circulating human and avian IAV strains and provide crucial genetic information. As such, WBE can provide rapid, cost-effective, year-round One Health surveillance to help control IAV epidemic and pandemic-related threats. However, optimisation of WBE protocols are necessary to ensure observed wastewater signals not only correlate with clinical case data, but yield information on the wider environmental pan-influenz-ome. FUNDING Department of Health for Northern Ireland.
Collapse
Affiliation(s)
- Andrew J Lee
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK.
| | - Stephen Carson
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Marina I Reyne
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Andrew Marshall
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Daniel Moody
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Danielle M Allen
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Pearce Allingham
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Ashley Levickas
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Arthur Fitzgerald
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Stephen H Bell
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Jonathan Lock
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Jonathon D Coey
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Cormac McSparron
- Geography, School of Natural and Built Environment, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Behnam F Nejad
- Geography, School of Natural and Built Environment, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Evan P Troendle
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - David A Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - David G Courtney
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Gisli G Einarsson
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - James P McKenna
- Regional Virology Laboratory, Belfast Health and Social Care Trust, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - Derek J Fairley
- Regional Virology Laboratory, Belfast Health and Social Care Trust, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - Tanya Curran
- Regional Virology Laboratory, Belfast Health and Social Care Trust, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - Jennifer M McKinley
- Geography, School of Natural and Built Environment, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Deirdre F Gilpin
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Ken Lemon
- Veterinary Sciences Division, Agri-Food and Biosciences Institute, Stormont, Belfast, Northern Ireland, UK
| | - John W McGrath
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK; Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Connor G G Bamford
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK; Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland, UK
| |
Collapse
|
25
|
Shahriar I, Kamra M, Kanduluru AK, Campbell CL, Nguyen TH, Srinivasarao M, Low PS. Targeted recruitment of immune effector cells for rapid eradication of influenza virus infections. Proc Natl Acad Sci U S A 2024; 121:e2408469121. [PMID: 39348541 PMCID: PMC11474073 DOI: 10.1073/pnas.2408469121] [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: 04/28/2024] [Accepted: 07/19/2024] [Indexed: 10/02/2024] Open
Abstract
Despite much research, considerable data suggest that influenza virus remains a serious health problem because i) the effectiveness of current vaccines ranges only from 19% to 60%, ii) available therapies remain ineffective in advanced stages of disease, iii) death rates vary between 25,000 and 72,000/year in the United States, and iv) avian influenza strains are now being transmitted to dairy cattle that in turn are infecting humans. To address these concerns, we have developed zanDR, a bispecific small molecule that binds and inhibits viral neuraminidase expressed on both free virus and virus-infected cells and recruits naturally occurring anti-rhamnose and anti-dinitrophenyl (DNP) antibodies with rhamnose and DNP haptens. Because the neuraminidase inhibition replicates the chemotherapeutic mechanism of zanamivir and oseltamivir, while rhamnose and DNP recruit endogenous antibodies much like an anti-influenza vaccine, zanDR reproduces most of the functions of current methods of protection against influenza virus infections. Importantly, studies on cells in culture demonstrate that both of the above protective mechanisms remain highly functional in the zanDR conjugate, while studies in lethally infected mice with advanced-stage disease establish that a single intranasal dose of zanDR not only yields 100% protection but also reduces lung viral loads faster and ~1,000× more thoroughly than current antiviral therapies. Since zanDR also lowers secretion of proinflammatory cytokines and protects against virus-induced damage to the lungs better than current therapies, we suggest that combining an immunotherapy with a chemotherapy in single pharmacological agent constitutes a promising approach for treating the more challenging forms of influenza.
Collapse
Affiliation(s)
- Imrul Shahriar
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN47907
- Eradivir Inc., West Lafayette, IN47906
| | - Mohini Kamra
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN47907
| | - Ananda Kumar Kanduluru
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN47907
- Eradivir Inc., West Lafayette, IN47906
| | - Charity Lynn Campbell
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN47907
- Eradivir Inc., West Lafayette, IN47906
| | - Thanh Hiep Nguyen
- Department of Biological Sciences, Purdue University, West Lafayette, IN47907
| | - Madduri Srinivasarao
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN47907
- Eradivir Inc., West Lafayette, IN47906
| | - Philip S. Low
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN47907
| |
Collapse
|
26
|
Jin XY, Yang HY, Zhao GY, Dai CX, Zhang ZQ, Zhou DS, Yin Q, Dai EH. Comparative pathogenicity of influenza virus-induced pneumonia mouse model following intranasal and aerosolized intratracheal inoculation. Virol J 2024; 21:240. [PMID: 39354538 PMCID: PMC11446018 DOI: 10.1186/s12985-024-02516-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Infection of mice with mouse-adapted strains of influenza virus has been widely used to establish mouse pneumonia models. Intranasal inoculation is the traditional route for constructing an influenza virus-induced pneumonia mouse model, while intratracheal inoculation has been gradually applied in recent years. In this article, the pathogenicity of influenza virus-induced pneumonia mouse models following intranasal and aerosolized intratracheal inoculation were compared. METHODS By comparing the two ways of influenza inoculation, intranasal and intratracheal, a variety of indices such as survival rate, body weight change, viral titer and load, pathological change, lung wet/dry ratio, and inflammatory factors were investigated. Meanwhile, the transcriptome was applied for the initial exploration of the mechanism underlying the variations in the results between the two inoculation methods. RESULTS The findings suggest that aerosolized intratracheal infection leads to more severe lung injury and higher viral loads in the lungs compared to intranasal infection, which may be influenced by the initial site of infection, sialic acid receptor distribution, and host innate immunity. CONCLUSION Intratracheal inoculation is a better method for modelling severe pneumonia in mice than intranasal infection.
Collapse
Affiliation(s)
- Xiu-Yu Jin
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, People's Republic of China
| | - Hui-Ying Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Guang-Yu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Chen-Xi Dai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Zai-Qing Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Dong-Sheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Er-Hei Dai
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, People's Republic of China.
| |
Collapse
|
27
|
Tan X, Zhang Z, Zhang H, Li J, Yang X, Wang L, Liao X. Comparative study on the incidence of non-COVID-19 viral pneumonia before and after the COVID-19 pandemic: A retrospective analysis based on respiratory non-COVID viral nucleic acid results. JOURNAL OF INTENSIVE MEDICINE 2024; 4:491-495. [PMID: 39310063 PMCID: PMC11411423 DOI: 10.1016/j.jointm.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/26/2024] [Accepted: 02/24/2024] [Indexed: 09/25/2024]
Abstract
Background The impact of the coronavirus disease 2019 (COVID-19) pandemic on the etiology of non-COVID-19 viral pneumonia remains to be identified. We investigated the evolution of non-COVID-19 viral pneumonia in hospitalized patients before and after the COVID-19 pandemic. Methods This is a single-center retrospective study. Patients who came to West China Hospital of Sichuan University diagnosed with non-COVID-19 viral pneumonia from January 1, 2016 to December 31, 2021, were included and divided into pre- and post-COVID-19 groups according to the date of the COVID-19 outbreak in China. The results of 13 viral nucleic acid tests were compared between the two groups. Results A total of 5937 patients (3954 in the pre-COVID-19 group and 1983 in the post-COVID-19 group) were analyzed. Compared with the pre-COVID-19 group, the proportion of patients tested for respiratory non-COVID-19 viral nucleic acid was significantly increased in the post-COVID-19 group (14.78% vs. 22.79%, P <0.05). However, the non-COVID-19 virus-positive rates decreased from 37.9% to 14.6% after the COVID-19 outbreak (P < 0.001). Notably, non-COVID-19 viral pneumonia caused by the influenza A virus H1N1 (InfAH1N1) (2009) dropped to 0% after the pandemic. The top three viruses were InfAH1N1 (2009) (13.9%), human rhinovirus (7.4%), and human adenovirus (3.4%) in the pre-COVID-19 group, and human rhinovirus (3.8%), human respiratory syncytial virus (2.0%), human parainfluenza virus (1.1%) and InfAH3N2 (1.1%) in the post-COVID-19 group. Conclusions The proportion of non-COVID-19 viral pneumonia decreased significantly after the COVID-19 outbreak, among which InfAH1N1 (2009) pneumonia decreased the most dramatically.
Collapse
Affiliation(s)
- Xiaojiao Tan
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Zhang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huan Zhang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jianbo Li
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuewei Yang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lijie Wang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Critical Care Medicine, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
29
|
Ma C, Luo C, Deng F, Yu C, Chen Y, Zhong G, Zhan Y, Nie L, Huang Y, Xia Y, Cai Z, Xu K, Cai H, Wang F, Lu Z, Zeng X, Zhu Y, Liu S. Major vault protein directly enhances adaptive immunity induced by Influenza A virus or indirectly through innate immunity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167441. [PMID: 39069011 DOI: 10.1016/j.bbadis.2024.167441] [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: 01/08/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
As we previously revealed, major vault protein (MVP) is a virus-induced host factor, and its expression is crucial for innate immune responses. Nevertheless, the function of MVP in adaptive immunity is poorly known. Here, we demonstrate that Mvp knockout mice had attenuated antibody responses and reduced survival after rechallenge with homologous influenza A virus (IAV) relative to wild-type mice. Analysis of B cell populations showed that MVP promoted germinal center (GC) responses to develop optimal antiviral humoral immunity. Although MVP-deficient T cells and dendritic cells (DCs) were not intrinsically damaged, MVP promoted activating effector T cells and T follicular helper responses and regulated specific DC subsets. These findings suggest that MVP directs an effective adaptive immune response against IAV by directly engaging in GC reactions or indirectly augmenting cellular immunity via innate immune pathways.
Collapse
Affiliation(s)
- Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Feiyan Deng
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yumeng Chen
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gechang Zhong
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuxin Zhan
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zeng Cai
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huanhuan Cai
- Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China
| | - Zhibing Lu
- Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Xiangtai Zeng
- Department of General Surgery, Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China; Department of General Surgery, Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
30
|
Martinez-Sobrido L, Nogales A. Recombinant Influenza A Viruses Expressing Reporter Genes from the Viral NS Segment. Int J Mol Sci 2024; 25:10584. [PMID: 39408912 PMCID: PMC11476892 DOI: 10.3390/ijms251910584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Studying influenza A viruses (IAVs) requires secondary experimental procedures to detect the presence of the virus in infected cells or animals. The ability to generate recombinant (r)IAV using reverse genetics techniques has allowed investigators to generate viruses expressing foreign genes, including fluorescent and luciferase proteins. These rIAVs expressing reporter genes have allowed for easily tracking viral infections in cultured cells and animal models of infection without the need for secondary approaches, representing an excellent option to study different aspects in the biology of IAV where expression of reporter genes can be used as a readout of viral replication and spread. Likewise, these reporter-expressing rIAVs provide an excellent opportunity for the rapid identification and characterization of prophylactic and/or therapeutic approaches. To date, rIAV expressing reporter genes from different viral segments have been described in the literature. Among those, rIAV expressing reporter genes from the viral NS segment have been shown to represent an excellent option to track IAV infection in vitro and in vivo, eliminating the need for secondary approaches to identify the presence of the virus. Here, we summarize the status on rIAV expressing traceable reporter genes from the viral NS segment and their applications for in vitro and in vivo influenza research.
Collapse
Affiliation(s)
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, 28130 Madrid, Spain
| |
Collapse
|
31
|
Han Z, Mai Q, Zhao Y, Liu X, Cui M, Li M, Chen Y, Shu Y, Gan J, Pan W, Sun C. Mosaic neuraminidase-based vaccine induces antigen-specific T cell responses against homologous and heterologous influenza viruses. Antiviral Res 2024; 230:105978. [PMID: 39117282 DOI: 10.1016/j.antiviral.2024.105978] [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: 05/29/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.
Collapse
Affiliation(s)
- Zirong Han
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Qianyi Mai
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yangguo Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Xinglai Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Mingting Cui
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Minchao Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yaoqing Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China; Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianhui Gan
- Shenzhen Kangtai Biological Products Co., Ltd, Shenzhen, 518057, China.
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
32
|
Libin KV, Debnath M, Sisodiya S, Rathod SB, Prajapati PB, Lisina KV, Bhuyan R, Evanjelene VK. Bioefficacy, chromatographic profiling and drug-likeness analysis of flavonoids and terpenoids as potential inhibitors of H1N1 influenza viral proteins. Int J Biol Macromol 2024; 281:136125. [PMID: 39357733 DOI: 10.1016/j.ijbiomac.2024.136125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Considering medicinal plants, natural products present in these plants are the best sources of medications for combating viral infection. The possible drug target against viral H1N1 influenza proteins lead to identification of selected secondary metabolites from potential plants Tinospora cordifolia, Ocimum sanctum, and Piper nigrum. On analysis of in vitro cell based antiviral activity of the selected plant extracts, an indication for a possible lead compound against neuraminidase activity was evident. Potent ligands were selected using drug docking and ADMET analysis, and the screened lead metabolites were ultimately identified as terpenoid (Columbin) and, flavonoid (Cubebin, and Apigenin). Among the selected ligands, the drug binding activity of Cubebin with all the 6 proteins of H1N1 influenza type A virus, HA (4r8w), NA (4qn7), M2 (3lbw), PA (4wsb), PB1 (2znl) and PB2 (3wil), was pronounced. In addition, physicochemical and pharmacokinetic parameters linked to absorption, distribution, metabolism, excretion and toxicity (ADMET) have been evaluated and corroborate with our in vitro results. Molecular dynamics modelling indicated Cubebin can be a potential phytochemical in a drug discovery pipeline for the development of neuraminidase inhibitors. Further studies can provide a possibility for an alternative therapy against Influenza viruses.
Collapse
Affiliation(s)
- K V Libin
- Department of Biosciences and Biotechnology, Banasthali Vidyapith Jaipur, Rajasthan 304802, India
| | - Mousumi Debnath
- Department of Biosciences, Manipal University Jaipur, Rajasthan 303007, India.
| | - Smita Sisodiya
- Department of Biosciences, Manipal University Jaipur, Rajasthan 303007, India
| | - Shravan B Rathod
- Department of Chemistry, Smt. S. M. Panchal Science College, Talod, Gujarat, India
| | - Pravin B Prajapati
- Department of Chemistry, Sheth M. N. Science College, Patan, Gujarat, India
| | - K V Lisina
- Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Rajabrata Bhuyan
- Department of Biosciences and Biotechnology, Banasthali Vidyapith Jaipur, Rajasthan 304802, India
| | | |
Collapse
|
33
|
Luo C, Yang Y, Jiang C, Lv A, Zuo W, Ye Y, Ke J. Influenza and the gut microbiota: A hidden therapeutic link. Heliyon 2024; 10:e37661. [PMID: 39315196 PMCID: PMC11417228 DOI: 10.1016/j.heliyon.2024.e37661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/31/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Background The extensive community of gut microbiota significantly influences various biological functions throughout the body, making its characterization a focal point in biomedicine research. Over the past few decades, studies have revealed a potential link between specific gut bacteria, their associated metabolic pathways, and influenza. Bacterial metabolites can communicate directly or indirectly with organs beyond the gut via the intestinal barrier, thereby impacting the physiological functions of the host. As the microbiota increasingly emerges as a 'gut signature' in influenza, gaining a deeper understanding of its role may offer new insights into its pathophysiological relevance and open avenues for novel therapeutic targets. In this Review, we explore the differences in gut microbiota between healthy individuals and those with influenza, the relationship between gut microbiota metabolites and influenza, and potential strategies for preventing and treating influenza through the regulation of gut microbiota and its metabolites, including fecal microbiota transplantation and microecological preparations. Methods We utilized PubMed and Web of Science as our search databases, employing keywords such as "influenza," "gut microbiota," "traditional Chinese medicine," "metabolites," "prebiotics," "probiotics," and "machine learning" to retrieve studies examining the potential therapeutic connections between the modulation of gut microbiota and its metabolites in the treatment of influenza. The search encompassed literature from the inception of the databases up to December 2023. Results Fecal microbiota transplantation (FMT), microbial preparations (probiotics and prebiotics), and traditional Chinese medicine have unique advantages in regulating intestinal microbiota and its metabolites to improve influenza outcomes. The primary mechanism involves increasing beneficial intestinal bacteria such as Bacteroidetes and Bifidobacterium while reducing harmful bacteria such as Proteobacteria. These interventions act directly or indirectly on metabolites such as short-chain fatty acids (SCFAs), amino acids (AAs), bile acids, and monoamines to alleviate lung inflammation, reduce viral load, and exert anti-influenza virus effects. Conclusion The gut microbiota and its metabolites have direct or indirect therapeutic effects on influenza, presenting broad research potential for providing new directions in influenza research and offering references for clinical prevention and treatment. Future research should focus on identifying key strains, specific metabolites, and immune regulation mechanisms within the gut microbiota to accurately target microbiota interventions and prevent respiratory viral infections such as influenza.
Collapse
Affiliation(s)
- Cheng Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Yi Yang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Cheng Jiang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Anqi Lv
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Wanzhao Zuo
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Yuanhang Ye
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Jia Ke
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, 430074, China
| |
Collapse
|
34
|
Ahmadivand S, Fux R, Palić D. Ferritin Vaccine Platform for Animal and Zoonotic Viruses. Vaccines (Basel) 2024; 12:1112. [PMID: 39460279 PMCID: PMC11511493 DOI: 10.3390/vaccines12101112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Viral infections in animals continue to pose a significant challenge, affecting livestock health, welfare, and food safety, and, in the case of zoonotic viruses, threatening global public health. The control of viral diseases currently relies on conventional approaches such as inactivated or attenuated vaccines produced via platforms with inherent limitations. Self-assembling ferritin nanocages represent a novel vaccine platform that has been utilized for several viruses, some of which are currently undergoing human clinical trials. Experimental evidence also supports the potential of this platform for developing commercial vaccines for veterinary viruses. In addition to improved stability and immunogenicity, ferritin-based vaccines are safe and DIVA-compatible, and can be rapidly deployed in response to emerging epidemics or pandemics. This review discusses the structural and functional properties of ferritin proteins, followed by an overview of the design and production of ferritin-based vaccines, the mechanisms of immune responses, and their applications in developing vaccines against animal and zoonotic viruses.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| |
Collapse
|
35
|
Weichel HM, Koch T. [Vaccinations for emerging and re-emerging viral diseases]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024:10.1007/s00108-024-01780-6. [PMID: 39316120 DOI: 10.1007/s00108-024-01780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/07/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Emerging or re-emerging viral diseases have a pandemic potential and threaten global health. Vaccination is of crucial importance in the prevention of emerging and re-emerging viral diseases. OBJECTIVE Description of the current status of vaccine development against Filoviridae, highly pathogenic coronaviruses, smallpox viruses, influenza viruses and arboviruses. MATERIAL AND METHODS Focused literature search. RESULTS The World Health Organization (WHO) regularly publishes a list of infectious diseases that are expected to pose a major threat to humanity as they are could potentially trigger new pandemics; however, in addition to these human-to-human transmissible diseases, some arboviruses also have pandemic potential. In recent years numerous new vaccines, some of which are highly effective, have been licensed against new and re-emerging viral diseases and other promising vaccine candidates are currently in development. CONCLUSION There are still gaps in the development of vaccines in the area of Filoviridae and highly pathogenic coronaviruses. Vaccinations against smallpox viruses have been available for a long time. Developing influenza vaccines against novel strains in a timely manner is a challenge and universal influenza vaccines could be a possible solution. Modern vaccines are available against the arboviruses dengue and Chikungunya fever.
Collapse
Affiliation(s)
- Hanna-Marie Weichel
- Institut für Infektionsforschung und Impfstoffentwicklung (IIRVD), Zentrum für Innere Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
- Abteilung für klinische Immunologie von Infektionskrankheiten, Bernhard-Nocht-Institut für Tropenmedizin, Hamburg, Deutschland
- Ambulanzzentrum für Infektiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hamburg-Lübeck-Borstel-Riems, Hamburg, Deutschland
| | - Till Koch
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hamburg-Lübeck-Borstel-Riems, Hamburg, Deutschland.
- Antibiotic Stewardship Team, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland.
- Klinik für Pneumologie, Infektiologie und Onkologie, Klinikum Itzehoe, Robert Koch-Str. 2, 25524, Itzehoe, Deutschland.
| |
Collapse
|
36
|
Uno N, Ebensen T, Guzman CA, Ross TM. Intranasal administration of octavalent next-generation influenza vaccine elicits protective immune responses against seasonal and pre-pandemic viruses. J Virol 2024; 98:e0035424. [PMID: 39171925 PMCID: PMC11406897 DOI: 10.1128/jvi.00354-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/27/2024] [Indexed: 08/23/2024] Open
Abstract
Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine. IMPORTANCE Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.
Collapse
Affiliation(s)
- Naoko Uno
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, Florida, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
37
|
Rajashekar V, Stern L, Almeida CF, Slobedman B, Abendroth A. The surveillance of viral infections by the unconventional Type I NKT cell. Front Immunol 2024; 15:1472854. [PMID: 39355244 PMCID: PMC11442276 DOI: 10.3389/fimmu.2024.1472854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Type I NKT cells, also known as Invariant Natural Killer T (iNKT) cells, are a subpopulation of unconventional, innate-like T (ILT) cells which can proficiently influence downstream immune effector functions. Type I NKT cells express a semi-invariant αβ T cell receptor (TCR) that recognises lipid-based ligands specifically presented by the non-classical cluster of differentiation (CD1) protein d (CD1d) molecule. Due to their potent immunomodulatory functional capacity, type I NKT cells are being increasingly considered in prophylactic and therapeutic approaches towards various diseases, including as vaccine-adjuvants. As viruses do not encode lipid synthesis, it is surprising that many studies have shown that some viruses can directly impede type I NKT activation through downregulating CD1d expression. Therefore, in order to harness type I NKT cells for potential anti-viral therapeutic uses, it is critical that we fully appreciate how the CD1d-iNKT cell axis interacts with viral immunity. In this review, we examine clinical findings that underpin the importance of type I NKT cell function in viral infections. This review also explores how certain viruses employ immunoevasive mechanisms and directly encode functions to target CD1d expression and type I NKT cell function. Overall, we suggest that the CD1d-iNKT cell axis may hold greater gravity within viral infections than what was previously appreciated.
Collapse
Affiliation(s)
- Varshini Rajashekar
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Lauren Stern
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Catarina F. Almeida
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
38
|
Jalal H, Lee K, Burke DS. Oscillating spatiotemporal patterns of COVID-19 in the United States. Sci Rep 2024; 14:21562. [PMID: 39284868 PMCID: PMC11405777 DOI: 10.1038/s41598-024-72517-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
COVID-19 case rates in the US wax and wane in wave-like patterns over time, but the spatial patterns of these temporal epidemic waves have not been well characterized. By analyzing state- and county-level COVID-19 case rate data for spatiotemporal decomposition modes and oscillatory patterns, we demonstrate that the transmission dynamics of COVID-19 feature recurrent spatiotemporal patterns. In addition to the well-recognized national-level annual mid-winter surges, we demonstrate a prominent but previously unrecognized six-month north-south oscillation in the eastern US (Eastern US COVID-19 Oscillator-EUCO) that gives rise to regional sub-epidemics and travelling epidemic waves. We also demonstrate a second less prominent pattern that oscillates east-west in the northern US (Northern US COVID-19 Oscillator-NUCO). The drivers of these newly recognized oscillatory epidemic patterns remain to be elucidated.
Collapse
Affiliation(s)
- Hawre Jalal
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, K1G 5Z3, Canada.
| | - Kyueun Lee
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, WA, 98195, USA
| | - Donald S Burke
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| |
Collapse
|
39
|
Cai N, Zhan X, Zhang Q, Di H, Chen C, Hu Y, Yan X. Red Blood Cell-Derived Small Extracellular Vesicles Inhibit Influenza Virus through Surface-Displayed Sialic Acids. Angew Chem Int Ed Engl 2024:e202413946. [PMID: 39275883 DOI: 10.1002/anie.202413946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024]
Abstract
Disrupting the conserved multivalent binding of hemagglutinin (HA) on influenza A virus (IAV) to sialic acids (SAs) on the host cell membrane offers a robust strategy to block viral attachment and infection, irrespective of antigenic evolution or drug resistance. In this study, we exploit red blood cell-derived small extracellular vesicles (RBC sEVs) as nanodecoys by harnessing their high abundance of surface-displayed SAs to interact with IAV through multivalent HA-SA interactions. This high-avidity binding inhibits viral adhesion to the cell surface, effectively preventing both attachment and infection in a dose-dependent manner. Notably, enzymatic removal of SAs from RBC sEVs significantly diminishes their anti-IAV efficacy. Our findings indicate that RBC sEVs possess intrinsic anti-IAV properties due to their native multivalent SAs and hold considerable promise as antiviral therapeutics.
Collapse
Affiliation(s)
- Niangui Cai
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaozhen Zhan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Qingyuan Zhang
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Haonan Di
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Chen Chen
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yunyun Hu
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
40
|
Guo X, Zhao L, Li W, Cao R, Zhong W. The Synergistic Effect of Baloxavir and Neuraminidase Inhibitors against Influenza Viruses In Vitro. Viruses 2024; 16:1467. [PMID: 39339943 PMCID: PMC11437495 DOI: 10.3390/v16091467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Influenza viruses remain a major threat to human health. Four classes of drugs have been approved for the prevention and treatment of influenza infections. Oseltamivir, a neuraminidase inhibitor, is a first-line anti-influenza drug, and baloxavir is part of the newest generation of anti-influenza drugs that targets the viral polymerase. The emergence of drug resistance has reduced the efficacy of established antiviral drugs. Combination therapy is one of the options for controlling drug resistance and enhancing therapeutical efficacies. Here, we evaluate the antiviral effects of baloxavir combined with neuraminidase inhibitors (NAIs) against wild-type influenza viruses, as well as influenza viruses with drug-resistance mutations. The combination of baloxavir with NAIs led to significant synergistic effects; however, the combination of baloxavir with laninamivir failed to result in a synergistic effect on influenza B viruses. Considering the rapid emergence of drug resistance to baloxavir, we believe that these results will be beneficial for combined drug use against influenza.
Collapse
Affiliation(s)
- Xiaojia Guo
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Lei Zhao
- Beijing Sunho Pharmaceutical Co., Ltd., Beijing 102600, China
| | - Wei Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
41
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza induces lung lymphangiogenesis independent of YAP/TAZ activity in lymphatic endothelial cells. Sci Rep 2024; 14:21324. [PMID: 39266641 PMCID: PMC11393066 DOI: 10.1038/s41598-024-72115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases twofold at 7 days post-influenza infection (dpi) and threefold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA.
| | - Senegal Carty
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Fengzhi Shao
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Jhonatan Henao-Vasquez
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alexandra B Ysasi
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Michelle Zeng
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Anne Hinds
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Ming Lo
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Comparative Pathology Laboratory, Boston University National Emerging and Infectious Disease Laboratories, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Matthew R Jones
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alan Fine
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| |
Collapse
|
42
|
Wang M, Chen J, Zhang ZL. Highly-Efficient Selection of Aptamers for Quantitative Fluorescence Detecting Multiple IAV Subtypes. Anal Chem 2024. [PMID: 39259665 DOI: 10.1021/acs.analchem.4c03052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Influenza A virus (IAV) can cause infectious respiratory diseases in humans and animals. IAVs mutate rapidly through antigenic drift and shift, resulting in the emergence of numerous IAV subtypes and significant challenges for IAV detection. Therefore, achieving the simultaneous detection of multiple IAVs is crucial. In this work, three specific aptamers targeting the hemagglutination (HA) protein of the influenza A H5N1, H7N9, and H9N2 viruses were screened using a multichannel magnetic microfluidic chip. The aptamers exhibit nanomolar affinity and excellent specificity for the HA protein of H5N1, H7N9, and H9N2 viruses. Furthermore, three specific aptamers were truncated and labeled with different fluorescence markers to realize fluorescence quantitative detection of influenza A H5N1, H7N9, and H9N2 viruses through an aptamer sandwich assay in 1 h. The limit of detection (LOD) of the developed method is 0.38 TCID50/mL for the H5N1 virus, 0.75 TCID50/mL for the H7N9 virus, and 1.14 TCID50/mL for the H9N2 virus. The detection method has excellent specificity, strong anti-interference ability, and good reproducibility. This work provides a sensitive quantitative detection method for the H5N1, H7N9, and H9N2 viruses, enabling quantitative fluorescence detection for multiple IAV subtypes.
Collapse
Affiliation(s)
- Meng Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Jianjun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
43
|
Pi T, Sun L, Li W, Wang W, Dong M, Xu X, Xu H, Zhao Y. Preparation and characterization of kelp polysaccharide and its research on anti-influenza a virus activity. Int J Biol Macromol 2024; 280:135506. [PMID: 39260640 DOI: 10.1016/j.ijbiomac.2024.135506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/02/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
The beneficial effects of kelp polysaccharide (KPS) have recently attracted attention. In this study, KPS was extracted from kelp using the enzyme hydrolysis combined with freeze-drying, namely, KPS-EF. The structural characterization showed that KPS-EF was a highly sulfated macromolecule with the Mw of 764.2 kDa and the sulfate content of 23.49 %. The antiviral activity of KPS-EF in vitro was verified, and the IC50 value of KPS against the PR8 virus was 0.58 mg/mL. Intranasal administration of KPS-EF significantly inhibited death and weight loss in IAV-infected mice and alleviated virus-induced pneumonia symptoms, meanwhile, KPS-EF (10 mg/kg/day) significantly decreased the production levels of chemokines (CXCL1, RANTES) and inflammatory cytokines (IL-6, TNF-α) in lungs (p < 0.05). KPS-EF could downregulate the activity of viral neuraminidase (NA) primarily in the late stage of viral adsorption with an IC50 value of 0.29 mg/mL. This study provides a theoretical basis for the using KPS as a supplement to NA inhibitors or anti-influenza drugs.
Collapse
Affiliation(s)
- Tianxiang Pi
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Lishan Sun
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Wei Li
- Department of General Surgery, The District Hospital of Qingdao West Coast New Area, Qingdao 266400, China
| | - Wei Wang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Minghui Dong
- Zhongxin Anderson (Guangdong) Biotechnology Co., Ltd., Guangzhou 510000, China
| | - Xinxing Xu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - He Xu
- Lianyungang Baohong Marine Technology Co., Ltd., Lianyungang 222000, China
| | - Yuanhui Zhao
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
44
|
Sanches Santos Rizzo Zuttion M, Parimon T, Bora SA, Yao C, Lagree K, Gao CA, Wunderink RG, Kitsios GD, Morris A, Zhang Y, McVerry BJ, Modes ME, Marchevsky AM, Stripp BR, Soto CM, Wang Y, Merene K, Cho S, Victor BL, Vujkovic-Cvijin I, Gupta S, Cassel SL, Sutterwala FS, Devkota S, Underhill DM, Chen P. Antibiotic use during influenza infection augments lung eosinophils that impair immunity against secondary bacterial pneumonia. J Clin Invest 2024; 134:e180986. [PMID: 39255040 PMCID: PMC11527449 DOI: 10.1172/jci180986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024] Open
Abstract
A leading cause of mortality after influenza infection is the development of a secondary bacterial pneumonia. In the absence of a bacterial superinfection, prescribing antibacterial therapies is not indicated but has become a common clinical practice for those presenting with a respiratory viral illness. In a murine model, we found that antibiotic use during influenza infection impaired the lung innate immunologic defenses toward a secondary challenge with methicillin-resistant Staphylococcus aureus (MRSA). Antibiotics augment lung eosinophils, which have inhibitory effects on macrophage function through the release of major basic protein. Moreover, we demonstrated that antibiotic treatment during influenza infection caused a fungal dysbiosis that drove lung eosinophilia and impaired MRSA clearance. Finally, we evaluated 3 cohorts of hospitalized patients and found that eosinophils positively correlated with antibiotic use, systemic inflammation, and worsened outcomes. Altogether, our work demonstrates a detrimental effect of antibiotic treatment during influenza infection that has harmful immunologic consequences via recruitment of eosinophils to the lungs, thereby increasing the risk of developing a secondary bacterial infection.
Collapse
Affiliation(s)
| | | | | | - Changfu Yao
- Department of Medicine
- Women’s Guild Lung Institute
| | - Katherine Lagree
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Catherine A. Gao
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Richard G. Wunderink
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alison Morris
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Barry R. Stripp
- Department of Medicine
- Women’s Guild Lung Institute
- Department of Biomedical Sciences
| | | | - Ying Wang
- Department of Medicine
- Women’s Guild Lung Institute
| | | | - Silvia Cho
- Department of Medicine
- Women’s Guild Lung Institute
| | | | - Ivan Vujkovic-Cvijin
- Department of Medicine
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suman Gupta
- Department of Medicine
- Women’s Guild Lung Institute
| | | | | | - Suzanne Devkota
- Department of Medicine
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Human Microbiome Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David M. Underhill
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Chen
- Department of Medicine
- Women’s Guild Lung Institute
- Department of Biomedical Sciences
| |
Collapse
|
45
|
Benito AA, Monteagudo LV, Lázaro-Gaspar S, Mazas-Cabetas L, Quílez J. Detection and subtyping of influenza A virus in porcine clinical samples from Spain in 2020. Virology 2024; 600:110223. [PMID: 39278103 DOI: 10.1016/j.virol.2024.110223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
A total of 1019 samples collected on 726 Spanish swine farms suffering from outbreaks of respiratory disease were screened for influenza A viruses (IAVs) using a RT-qPCR method. A subset of positive samples was further analyzed using a subtype-specific RT-qPCR method (n: 142) and Sanger sequencing (n: 64). A total of 19.4% samples from 23% farms tested positive, with infection being most common in suckling (53.6%) and weaning pigs (30.2%). Viruses belonging to four HA subtypes (H1av, H1hu, H1pdm, H3) were detected, with subtypes H1avN2, H1huN2 and H1avN1 accounting for over half of the specimens. An optimized protocol with newly designed primers allowed the detection of H3 viruses in a significant number of samples (21%). A comparison of antigenic positions revealed that circulating strains exhibited mutations with vaccine strains in a significant percentage of amino acid residues, both in the NA protein (27.8-43.3%) and particularly in the HA protein (51-75.3%).
Collapse
Affiliation(s)
- Alfredo A Benito
- Exopol S.L., Pol Río Gállego D/14, San Mateo de Gállego, 50840, Zaragoza, Spain
| | - Luis V Monteagudo
- Department of Anatomy, Embriology and Genetics, Faculty of Veterinary Sciences, University of Zaragoza, 50013, Zaragoza, Spain; Agrifood Institute of Aragón (IA2), University of Zaragoza-CITA, 50013, Zaragoza, Spain
| | - Sofía Lázaro-Gaspar
- Exopol S.L., Pol Río Gállego D/14, San Mateo de Gállego, 50840, Zaragoza, Spain
| | - Luna Mazas-Cabetas
- Exopol S.L., Pol Río Gállego D/14, San Mateo de Gállego, 50840, Zaragoza, Spain
| | - Joaquín Quílez
- Department of Animal Pathology, Faculty of Veterinary Sciences, University of Zaragoza, 50013, Zaragoza, Spain; Agrifood Institute of Aragón (IA2), University of Zaragoza-CITA, 50013, Zaragoza, Spain.
| |
Collapse
|
46
|
Hsu D, Jayaraman A, Pucci A, Joshi R, Mancini K, Chen HL, Koslovsky K, Mao X, Choi A, Henry C, Vakil J, Stadlbauer D, Jorquera P, Arunkumar GA, Sanchez-Crespo NE, Wadsworth LT, Bhupathy V, Du E, Avanesov A, Ananworanich J, Nachbagauer R. Safety and immunogenicity of mRNA-based seasonal influenza vaccines formulated to include multiple A/H3N2 strains with or without the B/Yamagata strain in US adults aged 50-75 years: a phase 1/2, open-label, randomised trial. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00493-6. [PMID: 39245055 DOI: 10.1016/s1473-3099(24)00493-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Inclusion of additional influenza A/H3N2 strains in seasonal influenza vaccines could expand coverage against multiple, antigenically distinct, cocirculating A/H3N2 clades and potentially replace the no longer circulating B/Yamagata strain. We aimed to evaluate the safety and immunogenicity of three next-generation seasonal influenza mRNA vaccines with different compositions that encode for haemagglutinins of multiple A/H3N2 strains, with or without the B/Yamagata strain, in adults. METHODS This randomised, open-label, phase 1/2 trial enrolled healthy adults aged 50-75 years across 22 sites in the USA. Participants were randomly assigned (1:1:1:1:1:1:1) via interactive response technology to receive a single dose of mRNA-1011.1 (pentavalent; containing one additional A/H3N2 strain [Newcastle]), mRNA-1011.2 (quadrivalent; B/Yamagata replaced with one additional A/H3N2 strain [Newcastle]), mRNA-1012 at one of two dose levels (pentavalent; B/Yamagata replaced with two additional A/H3N2 strains [Newcastle and Hong Kong]), or one of three quadrivalent mRNA-1010 controls each encoding one of the A/H3N2 study strains. The primary outcomes were safety, evaluated in all randomly assigned participants who received a study vaccination (safety population), and reactogenicity, evaluated in all participants from the safety population who contributed any solicited adverse reaction data (solicited safety population). The secondary outcome was humoral immunogenicity of investigational mRNA vaccines at day 29 versus mRNA-1010 control vaccines based on haemagglutination inhibition antibody (HAI) assay in the per-protocol population. Here, we summarise findings from the planned interim analysis after participants had completed day 29. The study is registered with ClinicalTrials.gov, NCT05827068, and is ongoing. FINDINGS Between March 27 and May 9, 2023, 1183 participants were screened for eligibility, 699 (59·1%) were randomly assigned, and 696 (58·8%) received vaccination (safety population, n=696; solicited safety population, n=694; per-protocol population, n=646). 382 (55%) of the 696 participants in the safety population self-reported as female and 314 (45%) as male. Frequencies of solicited adverse reactions were similar across vaccine groups; 551 (79%) of 694 participants reported at least one solicited adverse reaction within 7 days after vaccination and 83 (12%) of 696 participants reported at least one unsolicited adverse event within 28 days after vaccination. No vaccine-related serious adverse events or deaths were reported. All three next-generation influenza vaccines elicited robust antibody responses against vaccine-matched influenza A and B strains at day 29 that were generally similar to mRNA-1010 controls, and higher responses against additional A/H3N2 strains that were not included within respective mRNA-1010 controls. Day 29 geometric mean fold rises in HAI titres from day 1 against vaccine-matched A/H3N2 strains were 3·0 (95% CI 2·6-3·6; Darwin) and 3·1 (2·6-3·8; Newcastle) for mRNA-1011.1; 3·3 (2·7-4·1; Darwin) and 4·2 (3·4-5·2; Newcastle) for mRNA-1011.2; 3·4 (2·9-4·0; Darwin), 4·5 (3·6-5·5; Newcastle), and 5·1 (4·2-6·2; Hong Kong) for mRNA-1012 50·0 μg; and 2·6 (2·2-3·1; Darwin), 3·7 (3·0-4·6; Newcastle), and 4·1 (3·3-5·1; Hong Kong) for mRNA-1012 62·5 μg. Inclusion of additional A/H3N2 strains did not reduce responses against influenza A/H1N1 or influenza B strains, and removal of B/Yamagata did not affect responses to B/Victoria. INTERPRETATION These data support the continued clinical development of mRNA-based next-generation seasonal influenza vaccines with broadened influenza A/H3N2 strain coverage. FUNDING Moderna.
Collapse
|
47
|
Degtyarev E, Feoktistova S, Volchkov P, Deviatkin A. Complex Evolutionary Dynamics of H5N8 Influenza A Viruses Revealed by Comprehensive Reassortment Analysis. Viruses 2024; 16:1405. [PMID: 39339881 PMCID: PMC11437431 DOI: 10.3390/v16091405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Influenza A viruses (IAVs) circulate among different species and have the potential to cause significant pandemics in humans. This study focuses on reassortment events in the H5N8 subtype of IAV, which poses a serious threat to public health due to its high pathogenicity in birds and potential for cross-species transmission. We retrieved 2359 H5N8 IAV sequences from GISAID, and filtered and analyzed 442 complete genomic sequences for reassortment events using pairwise distance deviation matrices (PDDMs) and pairwise distance correspondence plots (PDCPs). This detailed case study of specific H5N8 viruses revealed previously undescribed reassortment events, highlighting the complex evolutionary history and potential pandemic threat of H5N8 IAVs.
Collapse
Affiliation(s)
- Egor Degtyarev
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| | - Sofia Feoktistova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| | - Pavel Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
- Center for Personalized Medicine, The MCSC Named after A.S. Loginov, 111123 Moscow, Russia
| | - Andrey Deviatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| |
Collapse
|
48
|
Pekarek MJ, Weaver EA. Influenza B Virus Vaccine Innovation through Computational Design. Pathogens 2024; 13:755. [PMID: 39338946 PMCID: PMC11434669 DOI: 10.3390/pathogens13090755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
As respiratory pathogens, influenza B viruses (IBVs) cause a significant socioeconomic burden each year. Vaccine and antiviral development for influenza viruses has historically viewed IBVs as a secondary concern to influenza A viruses (IAVs) due to their lack of animal reservoirs compared to IAVs. However, prior to the global spread of SARS-CoV-2, the seasonal epidemics caused by IBVs were becoming less predictable and inducing more severe disease, especially in high-risk populations. Globally, researchers have begun to recognize the need for improved prevention strategies for IBVs as a primary concern. This review discusses what is known about IBV evolutionary patterns and the effect of the spread of SARS-CoV-2 on these patterns. We also analyze recent advancements in the development of novel vaccines tested against IBVs, highlighting the promise of computational vaccine design strategies when used to target both IBVs and IAVs and explain why these novel strategies can be employed to improve the effectiveness of IBV vaccines.
Collapse
Affiliation(s)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
49
|
Froes F, Timóteo A, Almeida B, Raposo JF, Oliveira J, Carrageta M, Duque S, Morais A. Influenza vaccination in older adults and patients with chronic disorders: A position paper from the Portuguese Society of Pulmonology, the Portuguese Society of Cardiology, the Portuguese Society of Diabetology, the Portuguese Society of Infectious Diseases and Clinical Microbiology, the Portuguese Society of Geriatrics and Gerontology, and the Study Group of Geriatrics of the Portuguese Society of Internal Medicine. Pulmonology 2024; 30:422-436. [PMID: 38129238 DOI: 10.1016/j.pulmoe.2023.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023] Open
Abstract
Influenza affects millions of people worldwide each year and can lead to severe complications, hospitalizations, and even death, especially among vulnerable populations such as older adults and those with chronic medical conditions. Annual vaccination is considered the most effective measure for preventing influenza and its complications. Despite the widespread availability of influenza vaccines, however, vaccination coverage rates remain suboptimal in several countries. Based on the latest scientific evidence and expert opinions on influenza vaccination in older people and patients with chronic disease, the Portuguese Society of Pulmonology (SPP), the Portuguese Society of Diabetology (SPD), the Portuguese Society of Cardiology (SPC), the Portuguese Society of Geriatrics and Gerontology (SPGG), the Study Group of Geriatrics of the Portuguese Society of Internal Medicine (NEGERMI-SPMI), and the Portuguese Society of Infectious Diseases and Clinical Microbiology (SPDIMC) discussed best practices for promoting vaccination uptake and coverage and drew up several recommendations to mitigate the impact of influenza. These recommendations focus on the efficacy and safety of available vaccines; the impact of influenza vaccination on older adults; patients with chronic medical conditions, namely cardiac and respiratory conditions, diabetes, and immunosuppressive diseases; and health care professionals, optimal vaccination timing, and strategies to increase vaccination uptake and coverage. The resulting position paper highlights the critical role that vaccinations play in promoting public health, raising awareness, and encouraging more people to get vaccinated.
Collapse
Affiliation(s)
- F Froes
- Torax Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Portuguese Society of Pulmonology (SPP), Portugal
| | - A Timóteo
- Cardiology Department, Hospital de Santa Marta, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal; NOVA Medical School, Lisboa, Portugal; Portuguese Society of Cardiology (SPC), Portugal
| | - B Almeida
- APDP Diabetes, Lisbon, Portugal; Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - J F Raposo
- NOVA Medical School, Lisboa, Portugal; APDP Diabetes, Lisbon, Portugal; Portuguese Society of Diabetology (SPD), Portugal
| | - J Oliveira
- Infection Control and Prevention and Antimicrobial Resistance Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Portuguese Society of Infectious Diseases and Clinical Microbiology (SPDIMC), Portugal
| | - M Carrageta
- Institute of Preventive Cardiology, Almada, Portugal; Portuguese Society of Geriatrics and Gerontology (SPGG), Portugal
| | - S Duque
- Hospital CUF Descobertas, Lisboa, Portugal; Institute of Preventive Medicine and Public Health, Faculty of Medicine, University of Lisbon, Lisboa, Portugal; Study Group of Geriatrics of the Portuguese Society of Internal Medicine (NEGERMI-SPMI), Portugal
| | - A Morais
- Portuguese Society of Pulmonology (SPP), Portugal; Pulmonology Department, Hospital de São João, Centro Hospitalar Universitário São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; i3S - Instituto de Biologia Molecular e Celular, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
| |
Collapse
|
50
|
Sun R, Zhang X, Hou J, Jia W, Li P, Song C. Development and validation of nomogram for predicting the risk of transferring to the ICU for children with influenza. Eur J Clin Microbiol Infect Dis 2024; 43:1795-1805. [PMID: 39002105 DOI: 10.1007/s10096-024-04898-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
OBJECTIVE Development of a nomogram model for predicting the magnitude of risk of transferring hospitalized children with influenza to the ICU. METHODS In a single-center retrospective study, 318 children with influenza who were hospitalized in our hospital from January 2018 to August 2023 were collected as study subjects. Children with influenza were randomly assigned to the training set and validation set in a ratio of 4:1. In the training set, risk factors were identified using univariate and multivariate logistic regression analyses, and a nomogram model was created on this basis. The validation set was used to evaluate the predictive power of the model. RESULTS Multifactorial logistic regression analysis revealed six independent risk factors for transfer to the ICU in hospitalized children with influenza, including elevated peripheral white blood cell counts, elevated large platelet ratios, reduced mean platelet width, reduced complement C3, elevated serum globulin levels, and reduced total immunoglobulin M levels. Using these six metrics as predictors to construct a nomogram graphical model, the C-index was 0.970 (95% Cl: 0.953-0.988). The areas under the curve for the training and validation sets were 0.966 (95%Cl 0.947-0.985) and 0.919 (95%Cl 0.851-0.986), respectively. CONCLUSION A nomogram for predicting the risk of transferring to the ICU for children with influenza was developed and validated, which demonstrates good calibration and clinical benefits.
Collapse
Affiliation(s)
- Ruiyang Sun
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Xue Zhang
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Jiapu Hou
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Wanyu Jia
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Peng Li
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Chunlan Song
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|