1
|
Alvarez J, Boklund A, Dippel S, Dórea F, Figuerola J, Herskin MS, Michel V, Miranda Chueca MÁ, Nannoni E, Nielsen SS, Nonno R, Riber AB, Stegeman JA, Ståhl K, Thulke H, Tuyttens F, Winckler C, Brugerolles C, Wolff T, Parys A, Lindh E, Latorre‐Margalef N, Rameix Welti M, Dürrwald R, Trebbien R, Van der Werf S, Gisslén M, Monne I, Fusaro A, Guinat C, Bortolami A, Alexakis L, Enkirch T, Svartstrom O, Willgert K, Baldinelli F, Preite L, Grant M, Broglia A, Melidou A. Preparedness, prevention and control related to zoonotic avian influenza. EFSA J 2025; 23:e9191. [PMID: 39882189 PMCID: PMC11775931 DOI: 10.2903/j.efsa.2025.9191] [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] [Indexed: 01/31/2025] Open
Abstract
A risk assessment framework was developed to evaluate the zoonotic potential of avian influenza (AI), focusing on virus mutations linked to phenotypic traits related to mammalian adaptation identified in the literature. Virus sequences were screened for the presence of these mutations and their geographical, temporal and subtype-specific trends. Spillover events to mammals (including humans) and human seroprevalence studies were also reviewed. Thirty-four mutations associated with five phenotypic traits (increased receptor specificity, haemagglutinin stability, neuraminidase specificity, enhanced polymerase activity and evasion of innate immunity) were shortlisted. AI viruses (AIVs) carrying multiple adaptive mutations and traits belonged to both low and highly pathogenic subtypes, mainly to A(H9N2), A(H7N9), A(H5N6) and A(H3N8), were sporadic and primarily detected in Asia. In the EU/EEA, H5Nx viruses of clade 2.3.4.4b, which have increased opportunities for evolution due to widespread circulation in birds and occasional cases/outbreaks in mammals, have acquired the highest number of zoonotic traits. Adaptive traits, such as enhanced polymerase activity and immune evasion, were frequently acquired, while receptor-specific mutations remained rare. Globally, human cases remain rare, with the majority overall due to A(H5N1), A(H5N6), A(H7N9) and A(H9N2) that are among the subtypes that tend to have a higher number of adaptive traits. The main drivers of mammalian adaptation include virus and host characteristics, and external factors increasing AIV exposure of mammals and humans to wild and domestic birds (e.g. human activities and ecological factors). Comprehensive surveillance of AIVs targeting adaptive mutations with whole genome sequencing in animals and humans is essential for early detection of zoonotic AIVs and efficient implementation of control measures. All preparedness, preventive and control measures must be implemented under a One Health framework and tailored to the setting and the epidemiological situation; in particular, enhanced monitoring, biosecurity, genomic surveillance and global collaboration are critical for mitigating the zoonotic risks of AIV.
Collapse
Affiliation(s)
| | | | - Julio Alvarez
- EFSA Panel on Animal Health and Animal Welfare members
| | | | - Sabine Dippel
- EFSA Panel on Animal Health and Animal Welfare members
| | | | | | | | | | | | | | | | - Romolo Nonno
- EFSA Panel on Animal Health and Animal Welfare members
| | - Anja B. Riber
- EFSA Panel on Animal Health and Animal Welfare members
| | | | - Karl Ståhl
- EFSA Panel on Animal Health and Animal Welfare members
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Liparulo TS, Shoemaker JE. Mathematical Modeling Suggests That Monocyte Activity May Drive Sex Disparities during Influenza Infection. Viruses 2024; 16:837. [PMID: 38932131 PMCID: PMC11209518 DOI: 10.3390/v16060837] [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: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
In humans, females of reproductive age often experience a more severe disease during influenza A virus infection, which may be due to differences in their innate immune response. Sex-specific outcomes to influenza infection have been recapitulated in mice, enabling researchers to study viral and immune dynamics in vivo in order to identify immune mechanisms that are differently regulated between the sexes. This study is based on the hypothesis that sex-specific outcomes emerge due to differences in the rates/speeds that select immune components respond. Using publicly available sex-specific murine data, we utilized dynamic mathematical models of the innate immune response to identify candidate mechanisms that may lead to increased disease severity in female mice. We implemented a large computational screen using the Bayesian information criterion (BIC), wherein the goodness of fit of the competing model scenarios is balanced against complexity (i.e., the number of parameters). Our results suggest that having sex-specific rates for proinflammatory monocyte induction by interferon and monocyte inhibition of virus replication provides the simplest (lowest BIC) explanation for the difference observed in the male and female immune responses. Markov-chain Monte Carlo (MCMC) analysis and global sensitivity analysis of the top performing scenario were performed to provide rigorous estimates of the sex-specific parameter distributions and to provide insight into which parameters most affect innate immune responses. Simulations using the top-performing model suggest that monocyte activity could be a key target to reduce influenza disease severity in females. Overall, our Bayesian statistical and dynamic modeling approach suggests that monocyte activity and induction parameters are sex-specific and may explain sex-differences in influenza disease immune dynamics.
Collapse
Affiliation(s)
- Tatum S. Liparulo
- Department of Chemical & Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Jason E. Shoemaker
- Department of Chemical & Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
3
|
Hook JL, Bhattacharya J. The pathogenesis of influenza in intact alveoli: virion endocytosis and its effects on the lung's air-blood barrier. Front Immunol 2024; 15:1328453. [PMID: 38343548 PMCID: PMC10853445 DOI: 10.3389/fimmu.2024.1328453] [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: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
Lung infection by influenza A virus (IAV) is a major cause of global mortality from lung injury, a disease defined by widespread dysfunction of the lung's air-blood barrier. Endocytosis of IAV virions by the alveolar epithelium - the cells that determine barrier function - is central to barrier loss mechanisms. Here, we address the current understanding of the mechanistic steps that lead to endocytosis in the alveolar epithelium, with an eye to how the unique structure of lung alveoli shapes endocytic mechanisms. We highlight where future studies of alveolar interactions with IAV virions may lead to new therapeutic approaches for IAV-induced lung injury.
Collapse
Affiliation(s)
- Jaime L. Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jahar Bhattacharya
- Department of Medicine, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, United States
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
4
|
Kirk NM, Liang Y, Ly H. Comparative Pathology of Animal Models for Influenza A Virus Infection. Pathogens 2023; 13:35. [PMID: 38251342 PMCID: PMC10820042 DOI: 10.3390/pathogens13010035] [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: 10/18/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Animal models are essential for studying disease pathogenesis and to test the efficacy and safety of new vaccines and therapeutics. For most diseases, there is no single model that can recapitulate all features of the human condition, so it is vital to understand the advantages and disadvantages of each. The purpose of this review is to describe popular comparative animal models, including mice, ferrets, hamsters, and non-human primates (NHPs), that are being used to study clinical and pathological changes caused by influenza A virus infection with the aim to aid in appropriate model selection for disease modeling.
Collapse
Affiliation(s)
| | | | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN 55108, USA; (N.M.K.); (Y.L.)
| |
Collapse
|
5
|
Bauer L, Benavides FFW, Veldhuis Kroeze EJB, de Wit E, van Riel D. The neuropathogenesis of highly pathogenic avian influenza H5Nx viruses in mammalian species including humans. Trends Neurosci 2023; 46:953-970. [PMID: 37684136 PMCID: PMC10591965 DOI: 10.1016/j.tins.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023]
Abstract
Circulation of highly pathogenic avian influenza (HPAI) H5Nx viruses of the A/Goose/Guangdong/1/96 lineage in birds regularly causes infections of mammals, including humans. In many mammalian species, infections are associated with severe neurological disease, a unique feature of HPAI H5Nx viruses compared with other influenza A viruses. Here, we provide an overview of the neuropathogenesis of HPAI H5Nx virus infection in mammals, centered on three aspects: neuroinvasion, neurotropism, and neurovirulence. We focus on in vitro studies, as well as studies on naturally or experimentally infected mammals. Additionally, we discuss the contribution of viral factors to the neuropathogenesis of HPAI H5Nx virus infections and the efficacy of intervention strategies to prevent neuroinvasion or the development of neurological disease.
Collapse
Affiliation(s)
- Lisa Bauer
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Emmie de Wit
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Debby van Riel
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
6
|
Abdelwhab EM, Mettenleiter TC. Zoonotic Animal Influenza Virus and Potential Mixing Vessel Hosts. Viruses 2023; 15:980. [PMID: 37112960 PMCID: PMC10145017 DOI: 10.3390/v15040980] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Influenza viruses belong to the family Orthomyxoviridae with a negative-sense, single-stranded segmented RNA genome. They infect a wide range of animals, including humans. From 1918 to 2009, there were four influenza pandemics, which caused millions of casualties. Frequent spillover of animal influenza viruses to humans with or without intermediate hosts poses a serious zoonotic and pandemic threat. The current SARS-CoV-2 pandemic overshadowed the high risk raised by animal influenza viruses, but highlighted the role of wildlife as a reservoir for pandemic viruses. In this review, we summarize the occurrence of animal influenza virus in humans and describe potential mixing vessel or intermediate hosts for zoonotic influenza viruses. While several animal influenza viruses possess a high zoonotic risk (e.g., avian and swine influenza viruses), others are of low to negligible zoonotic potential (e.g., equine, canine, bat and bovine influenza viruses). Transmission can occur directly from animals, particularly poultry and swine, to humans or through reassortant viruses in "mixing vessel" hosts. To date, there are less than 3000 confirmed human infections with avian-origin viruses and less than 7000 subclinical infections documented. Likewise, only a few hundreds of confirmed human cases caused by swine influenza viruses have been reported. Pigs are the historic mixing vessel host for the generation of zoonotic influenza viruses due to the expression of both avian-type and human-type receptors. Nevertheless, there are a number of hosts which carry both types of receptors and can act as a potential mixing vessel host. High vigilance is warranted to prevent the next pandemic caused by animal influenza viruses.
Collapse
Affiliation(s)
- Elsayed M. Abdelwhab
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Thomas C. Mettenleiter
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| |
Collapse
|
7
|
Dutta AK, Gazi MS, Uddin SJ. A systemic review on medicinal plants and their bioactive constituents against avian influenza and further confirmation through in-silico analysis. Heliyon 2023; 9:e14386. [PMID: 36925514 PMCID: PMC10011005 DOI: 10.1016/j.heliyon.2023.e14386] [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: 08/06/2022] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Background Avian influenza or more commonly known as bird flu is a widespread infectious disease in poultry. This review aims to accumulate information of different natural plant sources that can aid in combating this disease. Influenza virus (IV) is known for its ability to mutate and infect different species (including humans) and cause fatal consequences. Methods Total 33 plants and 4 natural compounds were identified and documented. Molecular docking was performed against the target viral protein neuraminidase (NA), with some plant based natural compounds and compared their results with standard drugs Oseltamivir and Zanamivir to obtain novel drug targets for influenza in chickens. Results It was seen that most extracts exhibit their action by interacting with viral hemagglutinin or neuraminidase and inhibit viral entry or release from the host cell. Some plants also interacted with the viral RNA replication or by reducing proinflammatory cytokines. Ethanol was mostly used for extraction. Among all the plants Theobroma cacao, Capparis Sinaica Veil, Androgarphis paniculate, Thallasodendron cillatum, Sinularia candidula, Larcifomes officinalis, Lenzites betulina, Datronia molis, Trametes gibbose exhibited their activity with least concentration (below 10 μg/ml). The dockings results showed that some natural compounds (5,7- dimethoxyflavone, Aloe emodin, Anthocyanins, Quercetin, Hemanthamine, Lyocrine, Terpenoid EA showed satisfactory binding affinity and binding specificity with viral neuraminidase compared to the synthetic drugs. Conclusion This review clusters up to date information of effective herbal plants to bolster future influenza treatment research in chickens. The in-silico analysis also suggests some potential targets for future drug development but these require more clinical analysis.
Collapse
Affiliation(s)
- Ashit Kumar Dutta
- Pharmacy Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh
| | - Md Shamim Gazi
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh
| |
Collapse
|
8
|
Viral subversion of the cell polarity regulator Scribble. Biochem Soc Trans 2023; 51:415-426. [PMID: 36606695 PMCID: PMC9987997 DOI: 10.1042/bst20221067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
Scribble is a scaffolding protein that regulates key events such as cell polarity, tumorigenesis and neuronal signalling. Scribble belongs to the LAP family which comprise of 16 Leucine Rich Repeats (LRR) at the N-terminus, two LAP Specific Domains (LAPSD) and four PSD-95/Discs-large/ZO-1 (PDZ) domains at the C-terminus. The four PDZ domains have been shown to be key for a range of protein-protein interactions and have been identified to be crucial mediators for the vast majority of Scribble interactions, particularly via PDZ Binding Motifs (PBMs) often found at the C-terminus of interacting proteins. Dysregulation of Scribble is associated with poor prognosis in viral infections due to subversion of multiple cell signalling pathways by viral effector proteins. Here, we review the molecular details of the interplay between Scribble and viral effector proteins that provide insight into the potential modes of regulation of Scribble mediated polarity signalling.
Collapse
|
9
|
Vakili K, Fathi M, Yaghoobpoor S, Sayehmiri F, Nazerian Y, Nazerian A, Mohamadkhani A, Khodabakhsh P, Réus GZ, Hajibeygi R, Rezaei-Tavirani M. The contribution of gut-brain axis to development of neurological symptoms in COVID-19 recovered patients: A hypothesis and review of literature. Front Cell Infect Microbiol 2022; 12:983089. [PMID: 36619768 PMCID: PMC9815719 DOI: 10.3389/fcimb.2022.983089] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/25/2022] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota undergoes significant alterations in response to viral infections, particularly the novel SARS-CoV-2. As impaired gut microbiota can trigger numerous neurological disorders, we suggest that the long-term neurological symptoms of COVID-19 may be related to intestinal microbiota disorders in these patients. Thus, we have gathered available information on how the virus can affect the microbiota of gastrointestinal systems, both in the acute and the recovery phase of the disease, and described several mechanisms through which this gut dysbiosis can lead to long-term neurological disorders, such as Guillain-Barre syndrome, chronic fatigue, psychiatric disorders such as depression and anxiety, and even neurodegenerative diseases such as Alzheimer's and Parkinson's disease. These mechanisms may be mediated by inflammatory cytokines, as well as certain chemicals such as gastrointestinal hormones (e.g., CCK), neurotransmitters (e.g., 5-HT), etc. (e.g., short-chain fatty acids), and the autonomic nervous system. In addition to the direct influences of the virus, repurposed medications used for COVID-19 patients can also play a role in gut dysbiosis. In conclusion, although there are many dark spots in our current knowledge of the mechanism of COVID-19-related gut-brain axis disturbance, based on available evidence, we can hypothesize that these two phenomena are more than just a coincidence and highly recommend large-scale epidemiologic studies in the future.
Collapse
Affiliation(s)
- Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayehmiri
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Nazerian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ashraf Mohamadkhani
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gislaine Z. Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ramtin Hajibeygi
- Department of Cardiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Penn R, Tregoning JS, Flight KE, Baillon L, Frise R, Goldhill DH, Johansson C, Barclay WS. Levels of Influenza A Virus Defective Viral Genomes Determine Pathogenesis in the BALB/c Mouse Model. J Virol 2022; 96:e0117822. [PMID: 36226985 PMCID: PMC9645217 DOI: 10.1128/jvi.01178-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022] Open
Abstract
Defective viral genomes (DVGs), which are generated by the viral polymerase in error during RNA replication, can trigger innate immunity and are implicated in altering the clinical outcome of infection. Here, we investigated the impact of DVGs on innate immunity and pathogenicity in a BALB/c mouse model of influenza virus infection. We generated stocks of influenza viruses containing the internal genes of an H5N1 virus that contained different levels of DVGs (indicated by different genome-to-PFU ratios). In lung epithelial cells, the high-DVG stock was immunostimulatory at early time points postinfection. DVGs were amplified during virus replication in myeloid immune cells and triggered proinflammatory cytokine production. In the mouse model, infection with the different virus stocks produced divergent outcomes. The high-DVG stock induced an early type I interferon (IFN) response that limited viral replication in the lungs, resulting in minimal weight loss. In contrast, the virus stock with low levels of DVGs replicated to high titers and amplified DVGs over time, resulting in elevated levels of proinflammatory cytokines accompanied by rapid weight loss and increased morbidity and mortality. Our results suggest that the timing and levels of immunostimulatory DVGs generated during infection contribute to H5N1 pathogenesis. IMPORTANCE Mammalian infections with highly pathogenic avian influenza viruses (HPAIVs) cause severe disease associated with excessive proinflammatory cytokine production. Aberrant replication products, such as defective viral genomes (DVGs), can stimulate the antiviral response, and cytokine induction is associated with their emergence in vivo. We show that stocks of a recombinant virus containing HPAIV internal genes that differ in their amounts of DVGs have vastly diverse outcomes in a mouse model. The high-DVG stock resulted in extremely mild disease due to suppression of viral replication. Conversely, the stock that contained low DVGs but rapidly accumulated DVGs over the course of infection led to severe disease. Therefore, the timing of DVG amplification and proinflammatory cytokine production impact disease outcome, and these findings demonstrate that not all DVG generation reduces viral virulence. This study also emphasizes the crucial requirement to examine the quality of virus preparations regarding DVG content to ensure reproducible research.
Collapse
Affiliation(s)
- Rebecca Penn
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - John S. Tregoning
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Katie E. Flight
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Laury Baillon
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Rebecca Frise
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Daniel H. Goldhill
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Wendy S. Barclay
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Verma V, Dileepan M, Huang Q, Phan T, Hu WS, Ly H, Liang Y. Influenza A virus activates cellular Tropomyosin receptor kinase A (TrkA) signaling to promote viral replication and lung inflammation. PLoS Pathog 2022; 18:e1010874. [PMID: 36121891 PMCID: PMC9521937 DOI: 10.1371/journal.ppat.1010874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/29/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
Influenza A virus (IAV) infection causes acute respiratory disease with potential severe and deadly complications. Viral pathogenesis is not only due to the direct cytopathic effect of viral infections but also to the exacerbated host inflammatory responses. Influenza viral infection can activate various host signaling pathways that function to activate or inhibit viral replication. Our previous studies have shown that a receptor tyrosine kinase TrkA plays an important role in the replication of influenza viruses in vitro, but its biological roles and functional mechanisms in influenza viral infection have not been characterized. Here we show that IAV infection strongly activates TrkA in vitro and in vivo. Using a chemical-genetic approach to specifically control TrkA kinase activity through a small molecule compound 1NMPP1 in a TrkA knock-in (TrkA KI) mouse model, we show that 1NMPP1-mediated TrkA inhibition completely protected mice from a lethal IAV infection by significantly reducing viral loads and lung inflammation. Using primary lung cells isolated from the TrkA KI mice, we show that specific TrkA inhibition reduced IAV viral RNA synthesis in airway epithelial cells (AECs) but not in alveolar macrophages (AMs). Transcriptomic analysis confirmed the cell-type-specific role of TrkA in viral RNA synthesis, and identified distinct gene expression patterns under the TrkA regulation in IAV-infected AECs and AMs. Among the TrkA-activated targets are various proinflammatory cytokines and chemokines such as IL6, IL-1β, IFNs, CCL-5, and CXCL9, supporting the role of TrkA in mediating lung inflammation. Indeed, while TrkA inhibitor 1NMPP1 administered after the peak of IAV replication had no effect on viral load, it was able to decrease lung inflammation and provided partial protection in mice. Taken together, our results have demonstrated for the first time an important biological role of TrkA signaling in IAV infection, identified its cell-type-specific contribution to viral replication, and revealed its functional mechanism in virus-induced lung inflammation. This study suggests TrkA as a novel host target for therapeutic development against influenza viral disease.
Collapse
Affiliation(s)
- Vikram Verma
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, United States of America
| | - Mythili Dileepan
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, United States of America
| | - Qinfeng Huang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, United States of America
| | - Thu Phan
- Department of Chemical Engineering and Material Sciences, College of Science and Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wei-Shou Hu
- Department of Chemical Engineering and Material Sciences, College of Science and Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, United States of America
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, United States of America
| |
Collapse
|
12
|
Structural Basis of the Avian Influenza NS1 Protein Interactions with the Cell Polarity Regulator Scribble. Viruses 2022; 14:v14030583. [PMID: 35336989 PMCID: PMC8954747 DOI: 10.3390/v14030583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Scribble is a highly conserved regulator of cell polarity, a process that enables the generation of asymmetry at the cellular and tissue level in higher organisms. Scribble acts in concert with Disc-large (Dlg) and Lethal-2-giant larvae (Lgl) to form the Scribble polarity complex, and its functional dysregulation is associated with poor prognosis during viral infections. Viruses have been shown to interfere with Scribble by targeting Scribble PDZ domains to subvert the network of interactions that enable normal control of cell polarity via Scribble, as well as the localisation of the Scribble module within the cell. The influenza A virus NS1 protein was shown to bind to human Scribble (SCRIB) via its C-terminal PDZ binding motif (PBM). It was reported that the PBM sequence ESEV is a virulence determinant for influenza A virus H5N1 whilst other sequences, such as ESKV, KSEV and RSKV, demonstrated no affinity towards Scribble. We now show, using isothermal titration calorimetry (ITC), that ESKV and KSEV bind to SCRIB PDZ domains and that ESEV unexpectedly displayed an affinity towards all four PDZs and not just a selected few. We then define the structural basis for the interactions of SCRIB PDZ1 domain with ESEV and ESKV PBM motifs, as well as SCRIB PDZ3 with the ESKV PBM motif. These findings will serve as a platform for understanding the role of Scribble PDZ domains and their interactions with different NS1 PBMs and the mechanisms that mediate cell polarity within the context of the pathogenesis of influenza A virus.
Collapse
|
13
|
Full Genomic Sequences of H5N1 Highly Pathogenic Avian Influenza Virus in Human Autopsy Specimens Reveal Genetic Variability and Adaptive Changes for Growth in MDCK Cell Cultures. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3890681. [PMID: 34337007 PMCID: PMC8323515 DOI: 10.1155/2021/3890681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/23/2021] [Accepted: 07/13/2021] [Indexed: 11/25/2022]
Abstract
The entire H5N1 highly pathogenic avian influenza viral genomes were identified in the frozen autopsy specimens: the trachea, lung, colon, and intestinal feces from a patient who died of the disease in 2006. Phylogenetic analysis of the viral genomes showed that these viruses belonged to clade 1 and were the reassortants generated from the reassortment of the viruses within the same clade. The sequencing data from the autopsy specimens revealed at least 8 quasispecies of the H5N1 viruses across all 4 specimen types. These sequences were compared to those derived from the virus isolates grown in Madin Darby canine kidney (MDCK) cells. The virus isolates from the trachea, lung, and fecal specimens showed 27 nucleotide substitutions, leading to the changes of 18 amino acid residues. However, there was no change in the amino acid residues that determined the viral virulence. The changes were more commonly observed in the lung, particularly in the HA and NA genes. Our study suggested that the adaptation changes for the viral fitness to survive in a new host species (MDCK cells) might involve many genes, for example, the amino acid substitution 177G or 177W adjacent to the receptor-binding residues in the HA1 globular head and the substitution M315I in PB2. However, a mutation changes near the receptor binding domain may play an important role in determining the cell tropism and is needed to be further explored.
Collapse
|
14
|
Al Khatib HA, Coyle PV, Al Maslamani MA, Al Thani AA, Pathan SA, Yassine HM. Molecular and biological characterization of influenza A viruses isolated from human fecal samples. INFECTION GENETICS AND EVOLUTION 2021; 93:104972. [PMID: 34153546 DOI: 10.1016/j.meegid.2021.104972] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 01/14/2023]
Abstract
Human influenza viruses are occasionally detected in the stools of influenza patients. OBJECTIVES Here, we investigated the molecular and biological characteristics of intestinal influenza viruses and their potential role in virus transmission. METHODS Fecal samples were first screened for the presence of influenza viral RNA using RT-qPCR. Positive fecal samples were subjected to cell culture. Isolated viruses were then sequenced using MiSeq platform. Replication kinetics and receptor binding affinity were also evaluated. RESULTS Influenza RNA was detected in stool samples of 41% (36/87) of influenza A positive patients. Among the 36 stool samples subjected to viral isolation, 5 showed virus growth. Sequence analysis of isolated viruses revealed two distinct mutation patterns in fecal viruses. Set I viruses was able to replicate to higher titers in cell culture despite the limited number of mutations (6 mutations) compared to set II viruses (>10 mutations). Functional analysis of both sets revealed the ability to replicate efficiently in differentiated human bronchial cells. Receptor binding testing has also demonstrated their ability to bind α 2,3 and α 2,6 sialic acid receptors. CONCLUSION The ability of fecal influenza viruses to replicate in intestinal cells and human 3D bronchial cells might suggest their possible contribution in virus transmission.
Collapse
Affiliation(s)
| | - Peter V Coyle
- Virology Laboratory, Hamad Medical Corporation, Doha 3050, Qatar.
| | | | - Asmaa A Al Thani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; Department of Biomedical Sciences, College of Health Sciences-QU Health, Qatar University, Doha 2713, Qatar.
| | - Sameer A Pathan
- Emergency Medicine, Hamad Medical Corporation, Doha 3050, Qatar
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; Department of Biomedical Sciences, College of Health Sciences-QU Health, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
15
|
Profiling of Intestinal Microbiota in Patients Infected with Respiratory Influenza A and B Viruses. Pathogens 2021; 10:pathogens10060761. [PMID: 34204203 PMCID: PMC8233933 DOI: 10.3390/pathogens10060761] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/23/2022] Open
Abstract
Little is known about the association between respiratory viral infections and their impact on intestinal microbiota. Here, we compared the effect of influenza types, A and B, and influenza shedding in patients’ stools on the gut microbiota diversity and composition. Deep sequencing analysis was performed for the V4 region of the 16S rRNA gene. Fecal samples were collected from 38 adults with active respiratory influenza infection and 11 age-matched healthy controls. Influenza infection resulted in variations in intestinal bacterial community composition rather than in overall diversity. Overall, infected patients experienced an increased abundance of Bacteroidetes and a corresponding decrease in Firmicutes. Differential abundance testing illustrated that differences in gut microbiota composition were influenza type-dependent, identifying ten differentially abundant operational taxonomic units (OTUs) between influenza A- and influenza B-infected patients. Notably, virus shedding in fecal samples of some patients had significantly reduced gut bacterial diversity (p = 0.023). Further taxonomic analysis revealed that the abundance of Bacteroides fragilis was significantly higher among shedders compared to non-shedders (p = 0.037). These results provide fundamental evidence of the direct effect of influenza infection on gut microbiota diversity, as reported in patients shedding the virus.
Collapse
|
16
|
[The issues in detection of avian-type receptors for influenza viruses]. Uirusu 2021; 71:175-184. [PMID: 37245980 DOI: 10.2222/jsv.71.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Influenza viruses utilize sialic acid-containing glycoconjugates as receptors. The distribution of receptors in host tissues has been investigated in many species to understand the ecology of influenza viruses in nature and the mechanisms of interspecies transmission of the viruses. On the other hand, lectins, which have been widely used to detect these receptor molecules, have many different characteristics from antibodies and thus, require special attention in interpreting the results of lectin staining. In particular, lectins derived from Maackia amurensis, which has been used to detect Siaα2-3Gal, the avian-type receptor for influenza viruses, have been used without fully understanding its characteristics. This led to some confusion in interpreting the distribution of influenza virus receptors in host tissues. How accurately do we know the distribution of avian-type receptors in host animals? In this article, we would like to suggest reviewing the influenza virus receptors by providing issues related to Maackia lectins.
Collapse
|
17
|
Sriwilaijaroen N, Suzuki Y. Host Receptors of Influenza Viruses and Coronaviruses-Molecular Mechanisms of Recognition. Vaccines (Basel) 2020; 8:E587. [PMID: 33036202 PMCID: PMC7712180 DOI: 10.3390/vaccines8040587] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Among the four genera of influenza viruses (IVs) and the four genera of coronaviruses (CoVs), zoonotic αIV and βCoV have occasionally caused airborne epidemic outbreaks in humans, who are immunologically naïve, and the outbreaks have resulted in high fatality rates as well as social and economic disruption and losses. The most devasting influenza A virus (IAV) in αIV, pandemic H1N1 in 1918, which caused at least 40 million deaths from about 500 million cases of infection, was the first recorded emergence of IAVs in humans. Usually, a novel human-adapted virus replaces the preexisting human-adapted virus. Interestingly, two IAV subtypes, A/H3N2/1968 and A/H1N1/2009 variants, and two lineages of influenza B viruses (IBV) in βIV, B/Yamagata and B/Victoria lineage-like viruses, remain seasonally detectable in humans. Both influenza C viruses (ICVs) in γIV and four human CoVs, HCoV-229E and HCoV-NL63 in αCoV and HCoV-OC43 and HCoV-HKU1 in βCoV, usually cause mild respiratory infections. Much attention has been given to CoVs since the global epidemic outbreaks of βSARS-CoV in 2002-2004 and βMERS-CoV from 2012 to present. βSARS-CoV-2, which is causing the ongoing COVID-19 pandemic that has resulted in 890,392 deaths from about 27 million cases of infection as of 8 September 2020, has provoked worldwide investigations of CoVs. With the aim of developing efficient strategies for controlling virus outbreaks and recurrences of seasonal virus variants, here we overview the structures, diversities, host ranges and host receptors of all IVs and CoVs and critically review current knowledge of receptor binding specificity of spike glycoproteins, which mediates infection, of IVs and of zoonotic, pandemic and seasonal CoVs.
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Shizuoka 422-8526, Japan
| | - Yasuo Suzuki
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Shizuoka 422-8526, Japan
- College of Life and Health Sciences, Chubu University, Kasugai, Aichi 487-8501, Japan
| |
Collapse
|
18
|
Kongsomros S, Thanunchai M, Manopwisedjaroen S, Na-Ek P, Wang SF, Taechalertpaisarn T, Thitithanyanont A. Trogocytosis with monocytes associated with increased α2,3 sialic acid expression on B cells during H5N1 influenza virus infection. PLoS One 2020; 15:e0239488. [PMID: 32946496 PMCID: PMC7500609 DOI: 10.1371/journal.pone.0239488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 09/07/2020] [Indexed: 11/18/2022] Open
Abstract
The immunopathogenesis of H5N1 virus has been studied intensively since it caused cross-species infection and induced high mortality to human. We previously observed the interaction between monocytes and B cells, which increased the susceptibility of B cell to H5N1 virus infection after a co-culture. Levels of α2,3 sialic acid (avian flu receptor) were also significantly increased on B cell surface in this co-culture model with unclear explanation. In this study, we aimed to determine the possible mechanism that responded for this increase in α2,3 sialic acid on B cells. Acquisition of α2,3 SA by B cells via cell contact-dependent trogocytosis was proposed. Results showed that the lack of α2,3 SA was detected on B cell surface, and B cells acquired membrane-bound α2,3 SA molecules from monocytes in H5N1-infected co-cultures. Occurrence of membrane exchange mainly relied on H5N1 infection and cell-cell contact as opposed to a mock infection and transwell. The increase in α2,3 SA on B cell surface mediated by trogocytosis was associated with the enhanced susceptibility to H5N1 infection. These observations thus provide the evidence that H5N1 influenza virus may utilize trogocytosis to expand its cell tropism and spread to immune cells despite the lack of avian flu receptor.
Collapse
Affiliation(s)
- Supasek Kongsomros
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Maytawan Thanunchai
- Department of Clinical Pathology, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | | | - Prasit Na-Ek
- School of Medicine, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Sheng-Fan Wang
- Department of Medical Laboratory Sciences and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | | | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
19
|
Huang Y, Chen S, Yang Z, Guan W, Liu D, Lin Z, Zhang Y, Xu Z, Liu X, Li Y. SARS-CoV-2 Viral Load in Clinical Samples from Critically Ill Patients. Am J Respir Crit Care Med 2020; 201:1435-1438. [PMID: 32293905 PMCID: PMC7258645 DOI: 10.1164/rccm.202003-0572le] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yongbo Huang
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Sibei Chen
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Wenda Guan
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Dongdong Liu
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Zhimin Lin
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Yu Zhang
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Zhiheng Xu
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Yimin Li
- State Key Laboratory of Respiratory DiseasesGuangzhou, China
- Guangzhou Institute of Respiratory HealthGuangzhou, Chinaand
- First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| |
Collapse
|
20
|
Cantan B, Luyt CE, Martin-Loeches I. Influenza Infections and Emergent Viral Infections in Intensive Care Unit. Semin Respir Crit Care Med 2019; 40:488-497. [PMID: 31585475 PMCID: PMC7117087 DOI: 10.1055/s-0039-1693497] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Critically ill patients are admitted to an intensive care unit (ICU) for multiple reasons. In this study, we aim to analyze the current evidence and findings associated with influenza and other emergent viral infections, namely, herpes simplex virus type 1 (HSV-1), Epstein-Barr virus (EBV), and cytomegalovirus (CMV). Among medical conditions, community-acquired respiratory infections are the most frequent reason for ventilatory support in ICUs. Community-acquired pneumonia in a severe form including the need of invasive mechanical ventilation and/or vasopressors is associated with high mortality rates. However, after the pandemic that occurred in 2009 by H1N1 influenza, the number of cases being admitted to ICUs with viral infections is on the rise. Patients in whom an etiology would not have been identified in the past are currently being tested with more sensitive viral molecular diagnostic tools, and patients being admitted to ICUs have more preexisting medical conditions that can predispose to viral infections. Viral infections can trigger the dysregulation of the immune system by inducing a massive cytokine response. This cytokine storm can cause endothelial damage and dysfunction, deregulation of coagulation, and, consequently, alteration of microvascular permeability, tissue edema, and shock. In severe influenza, this vascular hyperpermeability can lead to acute lung injury, multiorgan failure, and encephalopathy. In immunocompetent patients, the most common viral infections are respiratory, and influenza should be considered in patients with severe respiratory failure being admitted to ICU. Seasonality and coinfection are two important features when considering influenza as a pathogen in critically ill patients. Herpesviridae (HSV, CMV, and EBV) may reactivate in ICU patients, and their reactivation is associated with morbidity/mortality. However, whether a specific treatment may impact on outcome remains to be determined.
Collapse
Affiliation(s)
- Ben Cantan
- Multidisciplinary Intensive Care Research Organization, St James's Hospital, Dublin, Ireland
| | - Charles-Edouard Luyt
- Médecine Intensive Réanimation, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne University (Paris 6), Paris, France.,INSERM, UMRS 1166-iCAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Ignacio Martin-Loeches
- Multidisciplinary Intensive Care Research Organization, St James's Hospital, Dublin, Ireland.,Department of Pulmonology, Hospital Clínic de Barcelona, Universitat de Barcelona and IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER), University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Bui CHT, Chan RWY, Ng MMT, Cheung MC, Ng KC, Chan MPK, Chan LLY, Fong JHM, Nicholls JM, Peiris JSM, Chan MCW. Tropism of influenza B viruses in human respiratory tract explants and airway organoids. Eur Respir J 2019; 54:13993003.00008-2019. [PMID: 31097520 DOI: 10.1183/13993003.00008-2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/07/2019] [Indexed: 11/05/2022]
Abstract
Despite causing regular seasonal epidemics with substantial morbidity, mortality and socioeconomic burden, there is still a lack of research into influenza B viruses (IBVs). In this study, we provide for the first time a systematic investigation on the tropism, replication kinetics and pathogenesis of IBVs in the human respiratory tract.Physiologically relevant ex vivo explant cultures of human bronchus and lung, human airway organoids, and in vitro cultures of differentiated primary human bronchial epithelial cells and type-I-like alveolar epithelial cells were used to study the cellular and tissue tropism, replication competence and induced innate immune response of 16 IBV strains isolated from 1940 to 2012 in comparison with human seasonal influenza A viruses (IAVs), H1N1 and H3N2. IBVs from the diverged Yamagata- and Victoria-like lineages and the earlier undiverged period were included.The majority of IBVs replicated productively in human bronchus and lung with similar competence to seasonal IAVs. IBVs infected a variety of cell types, including ciliated cells, club cells, goblet cells and basal cells, in human airway organoids. Like seasonal IAVs, IBVs are low inducers of pro-inflammatory cytokines and chemokines. Most results suggested a higher preference for the conducting airway than the lower lung and strain-specific rather than lineage-specific pathogenicity of IBVs.Our results highlighted the non-negligible virulence of IBVs which require more attention and further investigation to alleviate the disease burden, especially when treatment options are limited.
Collapse
Affiliation(s)
- Christine H T Bui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Joint first authors
| | - Renee W Y Chan
- Dept of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Joint first authors
| | - Mandy M T Ng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - M-C Cheung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ka-Chun Ng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Megan P K Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Louisa L Y Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Dept of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joanne H M Fong
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - J M Nicholls
- Dept of Pathology, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
22
|
Complete Genomic Sequences of Highly Pathogenic H5N1 Avian Influenza Viruses Obtained Directly from Human Autopsy Specimens. Microbiol Resour Announc 2018; 7:MRA01498-18. [PMID: 30533850 PMCID: PMC6284082 DOI: 10.1128/mra.01498-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 11/24/2022] Open
Abstract
The complete genomic sequences of H5N1 highly pathogenic avian influenza (HPAI) viruses were directly obtained from lung, trachea, and colon tissues and an intestinal fecal sample of a patient by using the next-generation sequencing technique. This is the first report on complete H5N1 viral genomes from human autopsy specimens. The complete genomic sequences of H5N1 highly pathogenic avian influenza (HPAI) viruses were directly obtained from lung, trachea, and colon tissues and an intestinal fecal sample of a patient by using the next-generation sequencing technique. This is the first report on complete H5N1 viral genomes from human autopsy specimens.
Collapse
|
23
|
Iwatsuki-Horimoto K, Nakajima N, Kiso M, Takahashi K, Ito M, Inoue T, Horiuchi M, Okahara N, Sasaki E, Hasegawa H, Kawaoka Y. The Marmoset as an Animal Model of Influenza: Infection With A(H1N1)pdm09 and Highly Pathogenic A(H5N1) Viruses via the Conventional or Tracheal Spray Route. Front Microbiol 2018; 9:844. [PMID: 29867791 PMCID: PMC5954801 DOI: 10.3389/fmicb.2018.00844] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/12/2018] [Indexed: 12/29/2022] Open
Abstract
To control infectious diseases in humans, it is important to understand the pathogenicity of the infecting organism(s). Although non-human primates, such as cynomolgus and rhesus macaques, have been used for influenza virus infection models, their size can limit their use in confined animal facilities. In this study, we investigated the susceptibility of marmosets to influenza viruses to assess the possibility of using these animals as a non-human primate model for influenza research. We first used an influenza A (H1N1)pdm09 virus to compare two inoculation routes: the conventional route, via a combination of the intratracheal, intranasal, ocular, and oral routes; and the tracheal spray route. In marmosets inoculated via the tracheal spray route, we found inflammation throughout the lungs and trachea. In contrast, in marmosets inoculated via the conventional route, the inflammation was confined to roughly the center of the lung. These data suggest that the tracheal spray route may be more suitable than the conventional route to inoculate marmosets with influenza viruses. We also tested an influenza A(H5N1) highly pathogenic avian influenza (HPAI) virus and found that some marmosets inoculated with this virus via the tracheal spray route showed weight loss, decreased body temperature, and loss of appetite and activity. The replication of this H5N1 virus in respiratory organs was confirmed. These results indicate the potential of marmosets as an animal model for infection with seasonal or HPAI viruses.
Collapse
Affiliation(s)
- Kiyoko Iwatsuki-Horimoto
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Noriko Nakajima
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Maki Kiso
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kenta Takahashi
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mutsumi Ito
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takashi Inoue
- Marmoset Research Department, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Machiko Horiuchi
- BioSciences Group, Summit Pharmaceuticals International Corporation, Tokyo, Japan
| | - Norio Okahara
- Marmoset Research Department, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Erika Sasaki
- Marmoset Research Department, Central Institute for Experimental Animals, Kawasaki, Japan.,Keio Advanced Research Center, Keio University, Tokyo, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
Sriwilaijaroen N, Nakakita SI, Kondo S, Yagi H, Kato K, Murata T, Hiramatsu H, Kawahara T, Watanabe Y, Kanai Y, Ono T, Hirabayashi J, Matsumoto K, Suzuki Y. N-glycan structures of human alveoli provide insight into influenza A virus infection and pathogenesis. FEBS J 2018. [PMID: 29542865 DOI: 10.1111/febs.14431] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The rapidly evolvable influenza A virus has caused pandemics linked to millions of deaths in the past century. Influenza A viruses are categorized by H (hemagglutinin; HA) and N (neuraminidase; NA) proteins expressed on the viral envelope surface. Analyses of past pandemics suggest that the HA gene segment comes from a nonhuman virus, which is then introduced into an immunologically naïve human population with potentially devastating consequences. As a prerequisite for infection, the nonhuman HA molecules of H1-H16 viruses must be able to bind to specific sialyl receptors on the host cell surface along the human respiratory tract. Thus, additional insight into the structures of host cell glycans and how different HAs interact with different glycans might provide new insight into the mechanisms underlying sustained infection and transmission in humans. In this work, we identified the sialyl N-glycans found in normal human alveoli and characterized the influenza viruses that preferentially bound to these different structures. We also determined the amino acid changes in HA that were linked to a switch of receptor-binding preference from nonhuman to pandemic, as well as pandemic to seasonal. Our data provide insight into why seasonal viruses are associated with reduced alveolar infection and damage and suggest new considerations for designing anti-HA vaccines and drugs. The results provide a better understanding of viral tropism and pathogenesis in humans that will be important for prediction and surveillance of zoonotic, pandemic, and epidemic influenza outbreaks. DATABASE The novel hemagglutinin nucleotide sequences reported here were deposited in GISAID under the accession numbers of EPI685738 for A/Yamaguchi/20/2006(H1N1) and EPI685740 for A/Kitakyushu/10/2006(H1N1).
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, Thailand.,College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Shin-Ichi Nakakita
- Department of Functional Glycomics, Life Science Research Center, Kagawa University, Japan
| | - Sachiko Kondo
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | - Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan.,Institute for Molecular Science and Okazaki Institute for Integrative Biosciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Takeomi Murata
- Department of Applied Biological Chemistry, Faculty of Agriculture, Shizuoka University, Japan
| | | | | | - Yohei Watanabe
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, Japan
| | - Yasushi Kanai
- The Institute of Scientific and Industrial Research, Osaka University, Japan
| | - Takao Ono
- The Institute of Scientific and Industrial Research, Osaka University, Japan
| | - Jun Hirabayashi
- Department of Functional Glycomics, Life Science Research Center, Kagawa University, Japan
| | - Kazuhiko Matsumoto
- The Institute of Scientific and Industrial Research, Osaka University, Japan
| | - Yasuo Suzuki
- College of Life and Health Sciences, Chubu University, Aichi, Japan
| |
Collapse
|
25
|
pH Optimum of Hemagglutinin-Mediated Membrane Fusion Determines Sensitivity of Influenza A Viruses to the Interferon-Induced Antiviral State and IFITMs. J Virol 2017; 91:JVI.00246-17. [PMID: 28356532 DOI: 10.1128/jvi.00246-17] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022] Open
Abstract
The replication and pathogenicity of influenza A viruses (IAVs) critically depend on their ability to tolerate the antiviral interferon (IFN) response. To determine a potential role for the IAV hemagglutinin (HA) in viral sensitivity to IFN, we studied the restriction of IAV infection in IFN-β-treated human epithelial cells by using 2:6 recombinant IAVs that shared six gene segments of A/Puerto Rico/8/1934 virus (PR8) and contained HAs and neuraminidases of representative avian, human, and zoonotic H5N1 and H7N9 viruses. In A549 and Calu-3 cells, viruses displaying a higher pH optimum of HA-mediated membrane fusion, H5N1-PR8 and H7N9-PR8, were less sensitive to the IFN-induced antiviral state than their counterparts with HAs from duck and human viruses, which fused at a lower pH. The association between a high pH optimum of fusion and reduced IFN sensitivity was confirmed by using HA point mutants of A/Hong Kong/1/1968-PR8 that differed solely by their fusion properties. Furthermore, similar effects of the viral fusion pH on IFN sensitivity were observed in experiments with (i) primary human type II alveolar epithelial cells and differentiated cultures of human airway epithelial cells, (ii) nonrecombinant zoonotic and pandemic IAVs, and (iii) preparations of IFN-α and IFN-λ1. A higher pH of membrane fusion and reduced sensitivity to IFN correlated with lower restriction of the viruses in MDCK cells stably expressing the IFN-inducible transmembrane proteins IFITM2 and IFITM3, which are known to inhibit viral fusion. Our results reveal that the pH optimum of HA-driven membrane fusion of IAVs is a determinant of their sensitivity to IFN and IFITM proteins.IMPORTANCE The IFN system constitutes an important innate defense against viral infection. Substantial information is available on how IAVs avoid detection by sensors of the IFN system and disable IFN signaling pathways. Much less is known about the ability of IAVs to tolerate the antiviral activity of IFN-induced cellular proteins. The IFN-induced proteins of the IFITM family block IAV entry into target cells and can restrict viral spread and pathogenicity. Here we show for the first time that the sensitivity of IAVs to the IFN-induced antiviral state and IFITM2 and IFITM3 proteins depends on the pH value at which the viral HA undergoes a conformational transition and mediates membrane fusion. Our data imply that the high pH optimum of membrane fusion typical of zoonotic IAVs of gallinaceous poultry, such as H5N1 and H7N9, may contribute to their enhanced virulence in humans.
Collapse
|
26
|
Wonderlich ER, Swan ZD, Bissel SJ, Hartman AL, Carney JP, O'Malley KJ, Obadan AO, Santos J, Walker R, Sturgeon TJ, Frye LJ, Maiello P, Scanga CA, Bowling JD, Bouwer AL, Duangkhae PA, Wiley CA, Flynn JL, Wang J, Cole KS, Perez DR, Reed DS, Barratt-Boyes SM. Widespread Virus Replication in Alveoli Drives Acute Respiratory Distress Syndrome in Aerosolized H5N1 Influenza Infection of Macaques. THE JOURNAL OF IMMUNOLOGY 2017; 198:1616-1626. [PMID: 28062701 DOI: 10.4049/jimmunol.1601770] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/09/2016] [Indexed: 01/01/2023]
Abstract
Human infections with highly pathogenic avian influenza A (H5N1) virus are frequently fatal but the mechanisms of disease remain ill-defined. H5N1 infection is associated with intense production of proinflammatory cytokines, but whether this cytokine storm is the main cause of fatality or is a consequence of extensive virus replication that itself drives disease remains controversial. Conventional intratracheal inoculation of a liquid suspension of H5N1 influenza virus in nonhuman primates likely results in efficient clearance of virus within the upper respiratory tract and rarely produces severe disease. We reasoned that small particle aerosols of virus would penetrate the lower respiratory tract and blanket alveoli where target cells reside. We show that inhalation of aerosolized H5N1 influenza virus in cynomolgus macaques results in fulminant pneumonia that rapidly progresses to acute respiratory distress syndrome with a fatal outcome reminiscent of human disease. Molecular imaging revealed intense lung inflammation coincident with massive increases in proinflammatory proteins and IFN-α in distal airways. Aerosolized H5N1 exposure decimated alveolar macrophages, which were widely infected and caused marked influx of interstitial macrophages and neutrophils. Extensive infection of alveolar epithelial cells caused apoptosis and leakage of albumin into airways, reflecting loss of epithelial barrier function. These data establish inhalation of aerosolized virus as a critical source of exposure for fatal human infection and reveal that direct viral effects in alveoli mediate H5N1 disease. This new nonhuman primate model will advance vaccine and therapeutic approaches to prevent and treat human disease caused by highly pathogenic avian influenza viruses.
Collapse
Affiliation(s)
- Elizabeth R Wonderlich
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261
| | - Zachary D Swan
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261
| | - Stephanie J Bissel
- Division of Neuropathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Amy L Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261
| | - Jonathan P Carney
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Katherine J O'Malley
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Adebimpe O Obadan
- Department of Population Health, University of Georgia, Athens, GA 30602
| | - Jefferson Santos
- Department of Population Health, University of Georgia, Athens, GA 30602
| | - Reagan Walker
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, PA 15260
| | - Timothy J Sturgeon
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - Lonnie J Frye
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219; and
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219; and
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219; and
| | - Jennifer D Bowling
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261
| | - Anthea L Bouwer
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261
| | - Parichat A Duangkhae
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261
| | - Clayton A Wiley
- Division of Neuropathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219; and
| | - Jieru Wang
- Division of Pulmonary Medicine, Allergy, and Immunology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Kelly S Cole
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Daniel R Perez
- Department of Population Health, University of Georgia, Athens, GA 30602
| | - Douglas S Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Simon M Barratt-Boyes
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261; .,Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
27
|
Hui KPY, Li HS, Cheung MC, Chan RWY, Yuen KM, Mok CKP, Nicholls JM, Peiris JSM, Chan MCW. Highly pathogenic avian influenza H5N1 virus delays apoptotic responses via activation of STAT3. Sci Rep 2016; 6:28593. [PMID: 27344974 PMCID: PMC4921847 DOI: 10.1038/srep28593] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/06/2016] [Indexed: 12/25/2022] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 virus continues to pose pandemic threat, but there is a lack of understanding of its pathogenesis. We compared the apoptotic responses triggered by HPAI H5N1 and low pathogenic H1N1 viruses using physiologically relevant respiratory epithelial cells. We demonstrated that H5N1 viruses delayed apoptosis in primary human bronchial and alveolar epithelial cells (AECs) compared to H1N1 virus. Both caspase-8 and -9 were activated by H5N1 and H1N1 viruses in AECs, while H5N1 differentially up-regulated TRAIL. H5N1-induced apoptosis was reduced by TRAIL receptor silencing. More importantly, STAT3 knock-down increased apoptosis by H5N1 infection suggesting that H5N1 virus delays apoptosis through activation of STAT3. Taken together, we demonstrate that STAT3 is involved in H5N1-delayed apoptosis compared to H1N1. Since delay in apoptosis prolongs the duration of virus replication and production of pro-inflammatory cytokines and TRAIL from H5N1-infected cells, which contribute to orchestrate cytokine storm and tissue damage, our results suggest that STAT3 may play a previously unsuspected role in H5N1 pathogenesis.
Collapse
Affiliation(s)
- Kenrie P. Y. Hui
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung Sing Li
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Man Chun Cheung
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Renee W. Y. Chan
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kit M. Yuen
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chris K. P. Mok
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The HKU-Pasteur Research Pole, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - John M. Nicholls
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - J. S. Malik Peiris
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michael C. W. Chan
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
28
|
New England harbor seal H3N8 influenza virus retains avian-like receptor specificity. Sci Rep 2016; 6:21428. [PMID: 26888262 PMCID: PMC4757820 DOI: 10.1038/srep21428] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 01/25/2016] [Indexed: 11/30/2022] Open
Abstract
An influenza H3N8 virus, carrying mammalian adaptation mutations, was isolated from New England harbor seals in 2011. We sought to assess the risk of its human transmissibility using two complementary approaches. First, we tested the binding of recombinant hemagglutinin (HA) proteins of seal H3N8 and human-adapted H3N2 viruses to respiratory tissues of humans and ferrets. For human tissues, we observed strong tendency of the seal H3 to bind to lung alveoli, which was in direct contrast to the human-adapted H3 that bound mainly to the trachea. This staining pattern was also consistent in ferrets, the primary animal model for human influenza pathogenesis. Second, we compared the binding of the recombinant HAs to a library of 610 glycans. In contrast to the human H3, which bound almost exclusively to α-2,6 sialylated glycans, the seal H3 bound preferentially to α-2,3 sialylated glycans. Additionally, the seal H3N8 virus replicated in human lung carcinoma cells. Our data suggest that the seal H3N8 virus has retained its avian-like receptor binding specificity, but could potentially establish infection in human lungs.
Collapse
|
29
|
Predicting Disease Severity and Viral Spread of H5N1 Influenza Virus in Ferrets in the Context of Natural Exposure Routes. J Virol 2015; 90:1888-97. [PMID: 26656692 DOI: 10.1128/jvi.01878-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/24/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Although avian H5N1 influenza virus has yet to develop the capacity for human-to-human spread, the severity of the rare cases of human infection has warranted intensive follow-up of potentially exposed individuals that may require antiviral prophylaxis. For countries where antiviral drugs are limited, the World Health Organization (WHO) has developed a risk categorization for different levels of exposure to environmental, poultry, or human sources of infection. While these take into account the infection source, they do not account for the likely mode of virus entry that the individual may have experienced from that source and how this could affect the disease outcome. Knowledge of the kinetics and spread of virus after natural routes of exposure may further inform the risk of infection, as well as the likely disease severity. Using the ferret model of H5N1 infection, we compared the commonly used but artificial inoculation method that saturates the total respiratory tract (TRT) with virus to upper respiratory tract (URT) and oral routes of delivery, those likely to be encountered by humans in nature. We show that there was no statistically significant difference in survival rate with the different routes of infection, but the disease characteristics were somewhat different. Following URT infection, viral spread to systemic organs was comparatively delayed and more focal than after TRT infection. By both routes, severe disease was associated with early viremia and central nervous system infection. After oral exposure to the virus, mild infections were common suggesting consumption of virus-contaminated liquids may be associated with seroconversion in the absence of severe disease. IMPORTANCE Risks for human H5N1 infection include direct contact with infected birds and frequenting contaminated environments. We used H5N1 ferret infection models to show that breathing in the virus was more likely to produce clinical infection than swallowing contaminated liquid. We also showed that virus could spread from the respiratory tract to the brain, which was associated with end-stage disease, and very early viremia provided a marker for this. With upper respiratory tract exposure, infection of the brain was common but hard to detect, suggesting that human neurological infections might be typically undetected at autopsy. However, viral spread to systemic sites was slower after exposure to virus by this route than when virus was additionally delivered to the lungs, providing a better therapeutic window. In addition to exposure history, early parameters of infection, such as viremia, could help prioritize antiviral treatments for patients most at risk of succumbing to infection.
Collapse
|
30
|
Fusco FM, Scappaticci L, Schilling S, De Iaco G, Brouqui P, Maltezou HC, Brodt HR, Bannister B, Ippolito G, Puro V. A 2009 cross-sectional survey of procedures for post-mortem management of highly infectious disease patients in 48 isolation facilities in 16 countries: data from EuroNHID. Infection 2015; 44:57-64. [PMID: 26267332 PMCID: PMC7099275 DOI: 10.1007/s15010-015-0831-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022]
Abstract
Purpose The handling of human remains may pose a risk for transmission of highly infectious agents. The use of appropriate biosafety measures is very important in case of management of patients deceased from highly infectious diseases (HIDs), such as Ebola virus disease. This paper presents the capabilities and resources in this field in 16 European countries, and suggests indications for the safe post-mortem management of HID patients. Methods The European Network for Highly Infectious Diseases conducted in 2009 a survey in 48 isolation facilities in 16 European countries. A set of standardized checklists, filled during on-site visits, have been used for data collection. Results Thirty-nine facilities (81.2 %) reported to have written procedures for the management of human remains, and 27 (56.2 %) for the performance of autopsies in HID patients. A Biosafety Level 3 autopsy room was available in eight (16.6 %) facilities, other technical devices for safe autopsies were available in nine (18.7 %). Overall, four facilities (8.3 %) reported to have all features explored for the safe management of human remains. Conversely, in five (10.4 %) none of these features were available. Conclusions The level of preparedness of surveyed isolation facilities in the field of post-mortem management in case of HIDs was not satisfactory, and improvements are needed.
Collapse
Affiliation(s)
- Francesco M Fusco
- National Institute for Infectious Diseases (INMI) "L. Spallanzani", Via Portuense 292, 00149, Rome, Italy.
| | - Lucia Scappaticci
- National Institute for Infectious Diseases (INMI) "L. Spallanzani", Via Portuense 292, 00149, Rome, Italy.
| | - Stefan Schilling
- Department of Infectious Diseases, J W Goethe University, Frankfurt, Germany.
| | - Giuseppina De Iaco
- National Institute for Infectious Diseases (INMI) "L. Spallanzani", Via Portuense 292, 00149, Rome, Italy.
| | - Philippe Brouqui
- Department of Infectious Diseases and Tropical Medicine, CHU Nord and URMITE IRDCNRS UMR 6236, Marseille, France.
| | | | - Hans-Reinhard Brodt
- Department of Infectious Diseases, J W Goethe University, Frankfurt, Germany.
| | | | - Giuseppe Ippolito
- National Institute for Infectious Diseases (INMI) "L. Spallanzani", Via Portuense 292, 00149, Rome, Italy.
| | - Vincenzo Puro
- National Institute for Infectious Diseases (INMI) "L. Spallanzani", Via Portuense 292, 00149, Rome, Italy.
| | | |
Collapse
|
31
|
Gambaryan AS, Matrosovich MN. What adaptive changes in hemagglutinin and neuraminidase are necessary for emergence of pandemic influenza virus from its avian precursor? BIOCHEMISTRY (MOSCOW) 2015; 80:872-880. [DOI: 10.1134/s000629791507007x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/13/2015] [Indexed: 08/30/2023]
|
32
|
Abstract
BACKGROUND The clinical features of avian-origin influenza virus A (H7N9) virus infection have been extensively characterized, but viral RNA detection in extra-pulmonary samples has seldom been studied. OBJECTIVES To study shedding of viral RNA in extra pulmonary samples in patients with avian influenza H7N9 infections. STUDY DESIGN A retrospective study of throat swabs, urine, fecal samples and sera collected sequentially from 18 hospitalized patients with H7N9 infections in Shanghai, China, between April and July in 2013 was conducted. RESULTS Viral RNA could be detected in urine samples from 17 patients, in fecal samples from 15 and in sera from 14 with a real-time reverse transcription polymerase chain reaction. The median duration of shedding of viral RNA was 19.7 days in throat swabs, 22 days in feces, 21.1 days in urines and 16.2 days in sera, indicating prolonged shedding of viral RNA in feces and urine compared with that in throat swabs. Prolonged duration of viral RNA detection in throat swabs and urine samples was observed in more severe patients. Moreover, in previously reported oseltamivir resistant patients, the NA gene with a 292K mutation could also be detected in their extra-pulmonary as well as in their respiratory samples. CONCLUSION Our data indicated a high frequency of viral RNA detection in feces, urine and sera in H7N9-infected patients and pointed out the potential risk of transmission.
Collapse
|
33
|
Abstract
Each year, influenza causes substantial mortality and morbidity worldwide. It is important to understand influenza in the tropics because of the significant burden in the region and its relevance to global influenza circulation. In this review, influenza burden, transmission dynamics, and their determinants in the tropics are discussed. Environmental, cultural, and social conditions in the tropics are very diverse and often differ from those of temperate regions. Theories that account for and predict influenza dynamics in temperate regions do not fully explain influenza epidemic patterns observed in the tropics. Routine surveillance and household studies have been useful in understanding influenza dynamics in the tropics, but these studies have been limited to only some regions; there is still a lack of information regarding influenza burden and transmission dynamics in many tropical countries. Further studies in the tropics will provide useful insight on many questions that remain.
Collapse
Affiliation(s)
- Sophia Ng
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109 USA
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109 USA
| |
Collapse
|
34
|
Hofer CC, Woods PS, Davis IC. Infection of mice with influenza A/WSN/33 (H1N1) virus alters alveolar type II cell phenotype. Am J Physiol Lung Cell Mol Physiol 2015; 308:L628-38. [PMID: 25595651 DOI: 10.1152/ajplung.00373.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/12/2015] [Indexed: 11/22/2022] Open
Abstract
Influenza viruses cause acute respiratory disease of great importance to public health. Alveolar type II (ATII) respiratory epithelial cells are central to normal lung function and are a site of influenza A virus replication in the distal lung. However, the consequences of infection for ATII cell function are poorly understood. To determine the impact of influenza infection on ATII cells we used C57BL/6-congenic SP-C(GFP) mice that express green fluorescent protein (GFP) under the control of the surfactant protein-C (SP-C) promoter, which is only active in ATII cells. Most cells isolated from the lungs of uninfected SP-C(GFP) mice were GFP(+) but did not express the alveolar type I (ATI) antigen podoplanin (PODO). ATII cells were also EpCAM(+) and α2,3-linked sialosaccharide(+). Infection with influenza A/WSN/33 virus caused severe hypoxemia and pulmonary edema. This was accompanied by loss of whole lung GFP fluorescence, reduced ATII cell yields, increased ATII cell apoptosis, reduced SP-C gene and protein expression in ATII cell lysates, and increased PODO gene and protein levels. Flow cytometry indicated that infection decreased GFP(+)/PODO(-) cells and increased GFP(-)/PODO(+) and GFP(-)/PODO(-) cells. Very few GFP(+)/PODO(+) cells were detectable. Finally, infection resulted in a significant decline in EpCAM expression by PODO(+) cells, but had limited effects on α2,3-linked sialosaccharides. Our findings indicate that influenza infection results in a progressive differentiation of ATII cells into ATI-like cells, possibly via an SP-C(-)/PODO(-) intermediate, to replace dying or dead ATI cells. However, impaired SP-C synthesis is likely to contribute significantly to reduced lung compliance in infected mice.
Collapse
Affiliation(s)
- Christian C Hofer
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, Ohio; and
| | - Parker S Woods
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Ian C Davis
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| |
Collapse
|
35
|
Feng Y, Hu L, Lu S, Chen Q, Zheng Y, Zeng D, Zhang J, Zhang A, Chen L, Hu Y, Zhang Z. Molecular pathology analyses of two fatal human infections of avian influenza A(H7N9) virus. J Clin Pathol 2014; 68:57-63. [PMID: 25378539 DOI: 10.1136/jclinpath-2014-202441] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS To investigate the histopathological manifestations of two fatal cases of H7N9 influenza A virus infection. METHODS Pulmonary and hepatic specimens from two fatal cases of H7N9 influenza virus infection were examined using H&E staining. Additionally, in situ hybridisation was performed with probes (ViewRNA) targeting H7N9 RNA and IP-10, interleukin (IL)-6 mRNA. The distribution of surfactant protein A (SP-A), surfactant protein B (SP-B), CD3, CD4, CD8, CD68 and C4d were determined with immunohistochemistry. RESULTS Apart from the typical diffuse alveolar damage and hyaline membrane observed in severe influenza infection, we detected H7N9 RNA and massive intrapulmonary production of IP-10 and IL-6 mRNA using in situ hybridisation. Hyperplasia of type II pneumocytes was observed by H&E staining and immunohistochemistry. Proliferating macrophages and clustered neutrophils in the infected lungs were observed, whereas T lymphocytes, especially CD4T helper cells, were markedly depleted. No obvious complement deposition was found in lung tissues. CONCLUSIONS Our findings suggest that H7N9 influenza virus induced an immunological response towards overt pulmonary inflammation and systemic lymphopenia which led to intense alveolar damage and respiratory failure.
Collapse
Affiliation(s)
- Yanling Feng
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lvyin Hu
- Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shuihua Lu
- Department of Respiratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qingguo Chen
- Department of Respiratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ye Zheng
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Dong Zeng
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jun Zhang
- Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Anli Zhang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Liang Chen
- Department of Viral Hepatitis, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yunwen Hu
- Department of Pathogen Diagnosis and Biosafety, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhiyong Zhang
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Disease severity is associated with differential gene expression at the early and late phases of infection in nonhuman primates infected with different H5N1 highly pathogenic avian influenza viruses. J Virol 2014; 88:8981-97. [PMID: 24899188 DOI: 10.1128/jvi.00907-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Occasional transmission of highly pathogenic avian H5N1 influenza viruses to humans causes severe pneumonia with high mortality. To better understand the mechanisms via which H5N1 viruses induce severe disease in humans, we infected cynomolgus macaques with six different H5N1 strains isolated from human patients and compared their pathogenicity and the global host responses to the virus infection. Although all H5N1 viruses replicated in the respiratory tract, there was substantial heterogeneity in their replicative ability and in the disease severity induced, which ranged from asymptomatic to fatal. A comparison of global gene expression between severe and mild disease cases indicated that interferon-induced upregulation of genes related to innate immunity, apoptosis, and antigen processing/presentation in the early phase of infection was limited in severe disease cases, although interferon expression was upregulated in both severe and mild cases. Furthermore, coexpression analysis of microarray data, which reveals the dynamics of host responses during the infection, demonstrated that the limited expression of these genes early in infection led to a failure to suppress virus replication and to the hyperinduction of genes related to immunity, inflammation, coagulation, and homeostasis in the late phase of infection, resulting in a more severe disease. Our data suggest that the attenuated interferon-induced activation of innate immunity, apoptosis, and antigen presentation in the early phase of H5N1 virus infection leads to subsequent severe disease outcome. IMPORTANCE Highly pathogenic avian H5N1 influenza viruses sometimes transmit to humans and cause severe pneumonia with ca. 60% lethality. The continued circulation of these viruses poses a pandemic threat; however, their pathogenesis in mammals is not fully understood. We, therefore, investigated the pathogenicity of six H5N1 viruses and compared the host responses of cynomolgus macaques to the virus infection. We identified differences in the viral replicative ability of and in disease severity caused by these H5N1 viruses. A comparison of global host responses between severe and mild disease cases identified the limited upregulation of interferon-stimulated genes early in infection in severe cases. The dynamics of the host responses indicated that the limited response early in infection failed to suppress virus replication and led to hyperinduction of pathological condition-related genes late in infection. These findings provide insight into the pathogenesis of H5N1 viruses in mammals.
Collapse
|
37
|
Leung YHC, Nicholls JM, Ho CK, Sia SF, Mok CKP, Valkenburg SA, Cheung P, Hui KPY, Chan RWY, Guan Y, Akira S, Peiris JSM. Highly pathogenic avian influenza A H5N1 and pandemic H1N1 virus infections have different phenotypes in Toll-like receptor 3 knockout mice. J Gen Virol 2014; 95:1870-1879. [PMID: 24878639 DOI: 10.1099/vir.0.066258-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Toll-like receptors (TLRs) play an important role in innate immunity to virus infections. We investigated the role of TLR3 in the pathogenesis of H5N1 and pandemic H1N1 (pH1N1) influenza virus infections in mice. Wild-type mice and those defective in TLR3 were infected with influenza A/HK/486/97 (H5N1) or A/HK/415742/09 (pH1N1) virus. For comparison, mice defective in the gene for myeloid differential factor 88 (MyD88) were also infected with the viruses, because MyD88 signals through a TLR pathway different from TLR3. Survival and body weight loss were monitored for 14 days, and lung pathology, the lung immune-cell profile, viral load and cytokine responses were studied. H5N1-infected TLR3(-/-) mice had better survival than H5N1-infected WT mice, evident by significantly faster regain of body weight, lower viral titre in the lung and fewer pathological changes in the lung. However, this improved survival was not seen upon pH1N1 infection of TLR3(-/-) mice. In contrast, MyD88(-/-) mice had an increased viral titre and decreased leukocyte infiltration in the lungs after infection with H5N1 virus and poorer survival after pH1N1 infection. In conclusion, TLR3 worsens the pathogenesis of H5N1 infection but not of pH1N1 infection, highlighting the differences in the pathogenesis of these two viruses and the different roles of TLR3 in their pathogenesis.
Collapse
Affiliation(s)
- Y H Connie Leung
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - John M Nicholls
- Department of Pathology, University of Hong Kong, Hong Kong, PR China
| | - Chuk Kwan Ho
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Sin Fun Sia
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Chris K P Mok
- HKU-Pasteur Research Pole, University of Hong Kong, Hong Kong, PR China.,Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Sophie A Valkenburg
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Peter Cheung
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Kenrie P Y Hui
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Renee W Y Chan
- Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - Y Guan
- State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Hong Kong, PR China.,Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| | - S Akira
- Department of Host Defense, Immunology Frontier Research Center, Osaka University, Japan
| | - J S Malik Peiris
- State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Hong Kong, PR China.,HKU-Pasteur Research Pole, University of Hong Kong, Hong Kong, PR China.,Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
38
|
Abstract
Despite heroic efforts to prevent the emergence of an influenza pandemic, avian influenza A virus has prevailed by crossing the species barriers to infect humans worldwide, occasionally with morbidity and mortality at unprecedented levels, and the virus later usually continues circulation in humans as a seasonal influenza virus, resulting in health-social-economic problems each year. Here, we review current knowledge of influenza viruses, their life cycle, interspecies transmission, and past pandemics and discuss the molecular basis of pandemic acquisition, notably of hemagglutinin (lectin) acting as a key contributor to change in host specificity in viral infection.
Collapse
Affiliation(s)
- Jun Hirabayashi
- National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | | |
Collapse
|
39
|
Mak TM, Hanson BJ, Tan YJ. Chimerization and characterization of a monoclonal antibody with potent neutralizing activity across multiple influenza A H5N1 clades. Antiviral Res 2014; 107:76-83. [PMID: 24797696 DOI: 10.1016/j.antiviral.2014.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 01/06/2023]
Abstract
The persistent evolution and circulation of highly pathogenic avian influenza H5N1 viruses pose a serious threat to global heath and hamper pandemic preparedness through conventional vaccine strategies. Combination passive immunotherapy using non-competing neutralizing antibodies has been proposed as a viable alternative to provide broad protection against drift variants. This necessitates the pre-pandemic production and characterization of potently neutralizing monoclonal antibodies (MAbs). One such antibody, MAb 9F4 was shown to provide heterologous protection against multiple H5N1 clade viruses, including one of the recently designated subclades, namely 2.3.4, through binding to a novel epitope, warranting its further development and characterization as a therapeutic candidate. In this study, the conversion of MAb 9F4 from mouse IgG2b to mouse-human chimeric (xi) IgG1 and IgA1 was achieved. These chimeric MAb versions were found to retain high degrees of binding and neutralizing activity against H5N1. The demonstration that xi-IgA1-9F4 retains a fairly high level of neutralizing activity, which is ∼10-fold lower than the corresponding xi-IgG1 isotype, suggests that this MAb could be further developed and engineered for intranasal administration.
Collapse
Affiliation(s)
- Tze-Minn Mak
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Brendon J Hanson
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore
| | - Yee-Joo Tan
- Infrastructure, Technology and Translational Division, Institute of Molecular and Cell Biology, A∗STAR, Singapore; Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.
| |
Collapse
|
40
|
van den Brand JMA, Haagmans BL, van Riel D, Osterhaus ADME, Kuiken T. The pathology and pathogenesis of experimental severe acute respiratory syndrome and influenza in animal models. J Comp Pathol 2014; 151:83-112. [PMID: 24581932 PMCID: PMC7094469 DOI: 10.1016/j.jcpa.2014.01.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/04/2013] [Accepted: 01/06/2014] [Indexed: 02/08/2023]
Abstract
Respiratory viruses that emerge in the human population may cause high morbidity and mortality, as well as concern about pandemic spread. Examples are severe acute respiratory syndrome coronavirus (SARS-CoV) and novel variants of influenza A virus, such as H5N1 and pandemic H1N1. Different animal models are used to develop therapeutic and preventive measures against such viruses, but it is not clear which are most suitable. Therefore, this review compares animal models of SARS and influenza, with an emphasis on non-human primates, ferrets and cats. Firstly, the pathology and pathogenesis of SARS and influenza are compared. Both diseases are similar in that they affect mainly the respiratory tract and cause inflammation and necrosis centred on the pulmonary alveoli and bronchioles. Important differences are the presence of multinucleated giant cells and intra-alveolar fibrosis in SARS and more fulminant necrotizing and haemorrhagic pneumonia in H5N1 influenza. Secondly, the pathology and pathogenesis of SARS and influenza in man and experimental animals are compared. Host species, host age, route of inoculation, location of sampling and timing of sampling are important to design an animal model that most closely mimics human disease. The design of appropriate animal models requires an accurate pathological description of human cases, as well as a good understanding of the effect of experimental variables on disease outcome.
Collapse
Affiliation(s)
- J M A van den Brand
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - B L Haagmans
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - D van Riel
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - A D M E Osterhaus
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - T Kuiken
- Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands.
| |
Collapse
|
41
|
Abstract
Although influenza A and B viruses are primarily known as respiratory viruses and mainly infected only the upper respiratory tract in humans, patients with influenza often develop signs and symptoms that are not due to the respiratory system. Frequently individuals with influenza develop headaches, meningismus, and even seizures in addition to their typical respiratory symptoms. In the past decades, influenza viruses have also been associated with serious non-respiratory signs. The famous 1918 strain of influenza was associated with von Economo's encephalitis lethargica and postencephalitic parkinsonism. In the 1960s influenza virus infections in children were associated with Reye's syndrome characterized often by fatty non-inflammatory hepatic disease and an encephalopathy with marked non-inflammatory cerebral edema. Intermittently children with influenza develop focal myalgia and myositis. Guillain–Barré syndrome was epidemiologically associated with the 1978 killed influenza vaccine but not subsequent vaccines. Although occasional children with influenza have developed encephalopathy, from 2000 through 2004 there was an increase in the number of serious cases of acute necrotizing encephalopathy accompanying infection with the influenza A 2009 strain. The current H5N1 strain of bird influenza occasionally infects humans with a high mortality rate and some appear to have central nervous signs. This chapter explores what is known about these influenza neurologic associations.
Collapse
Affiliation(s)
- Larry E Davis
- Neurology Service, New Mexico VA Health Care System and Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| | - Fredrick Koster
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | | |
Collapse
|
42
|
Böttcher-Friebertshäuser E, Garten W, Matrosovich M, Klenk HD. The hemagglutinin: a determinant of pathogenicity. Curr Top Microbiol Immunol 2014; 385:3-34. [PMID: 25031010 DOI: 10.1007/82_2014_384] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The hemagglutinin (HA) is a prime determinant of the pathogenicity of influenza A viruses. It initiates infection by binding to cell surface receptors and by inducing membrane fusion. The fusion capacity of HA depends on cleavage activation by host proteases, and it has long been known that highly pathogenic avian influenza viruses displaying a multibasic cleavage site differ in protease sensitivity from low pathogenic avian and mammalian influenza viruses with a monobasic cleavage site. Evidence is increasing that there are also variations in proteolytic activation among the viruses with a monobasic cleavage site, and several proteases have been identified recently that activate these viruses in a natural setting. Differences in protease sensitivity of HA and in tissue specificity of the enzymes are important determinants for virus tropism in the respiratory tract and for systemic spread of infection. Protease inhibitors that interfere with cleavage activation have the potential to be used for antiviral therapy and attenuated viruses have been generated by mutation of the cleavage site that can be used for the development of inactivated and live vaccines. It has long been known that human and avian influenza viruses differ in their specificity for sialic acid-containing cell receptors, and it is now clear that human tissues contain also receptors for avian viruses. Differences in receptor-binding specificity of seasonal and zoonotic viruses and differential expression of receptors for these viruses in the human respiratory tract account, at least partially, for the severity of disease. Receptor binding and fusion activation are modulated by HA glycosylation, and interaction of the glycans of HA with cellular lectins also affects virus infectivity. Interestingly, some of the mechanisms underlying pathogenicity are determinants of host range and transmissibility, as well.
Collapse
|
43
|
H1N1, but not H3N2, influenza A virus infection protects ferrets from H5N1 encephalitis. J Virol 2013; 88:3077-91. [PMID: 24371072 DOI: 10.1128/jvi.01840-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Seasonal influenza causes substantial morbidity and mortality because of efficient human-to-human spread. Rarely, zoonotic strains of influenza virus spread to humans, where they have the potential to mediate new pandemics with high mortality. We studied systemic viral spread after intranasal infection with highly pathogenic avian influenza virus (H5N1 [A/Viet Nam/1203/2004]) in ferrets with or without prior pandemic H1N1pdm09 (A/Mexico/4108/2009) or H3N2 (A/Victoria/361/2011) infection. After intranasal challenge with H5N1 influenza virus, naive ferrets rapidly succumbed to systemic infection. Animals challenged with H5N1 influenza virus greater than 3 months after recovering from an initial H1N1pdm09 infection survived H5N1 virus challenge and cleared virus from the respiratory tract 4 days after infection. However, a prolonged low-level infection of hematopoietic elements in the small bowel lamina propria, liver, and spleen was present for greater than 2 weeks postinfection, raising the potential for reassortment of influenza genes in a host infected with multiple strains of influenza. Animals previously infected with an H3N2 influenza virus succumbed to systemic disease and encephalitis after H5N1 virus challenge. These results indicate prior infection with different seasonal influenza strains leads to radically different protection from H5N1 challenge and fatal encephalitis. IMPORTANCE Seasonal influenza is efficiently transmitted from human to human, causing substantial morbidity and mortality. Rarely, zoonotic strains of influenza virus spread to humans, where they have the potential to mediate new pandemics with high mortality. Infection of naive ferrets with H5N1 avian influenza virus causes a rapid and lethal systemic disease. We studied systemic H5N1 viral spread after infection of ferrets with or without prior exposure to either of two seasonal influenza virus strains, H1N1 and H3N2. Ferrets previously infected with H1N1 survive H5N1 challenge while those previously infected with H3N2 die of encephalitis. However ferrets protected from lethal H5N1 infection develop persistent low-level infection of the small intestine, liver, or spleen, providing a nidus for future viral strain recombination. The mechanism by which prior infection with specific strains of seasonal influenza virus protect from lethal H5N1 challenge needs to be elucidated in order to design effective immunization and treatments.
Collapse
|
44
|
Immunomorphologic manifestations in mice liver infected with influenza A/H5N1, A/goose/Krasnoozerskoye/627/05 strain. Clin Dev Immunol 2013; 2013:342686. [PMID: 24454472 PMCID: PMC3886489 DOI: 10.1155/2013/342686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/01/2013] [Indexed: 11/24/2022]
Abstract
Highly pathogenic avian influenza H5N1 (HPAI H5N1) viruses can infect mammals, including humans, causing severe systemic disease with the inhibition of the immune system and a high mortality rate. In conditions of lymphoid tissue depletion, the liver plays an important role in host defence against viruses. The changes in mice liver infected with HPAI H5N1 virus A/goose/Krasnoozerskoye/627/05 have been studied. It has been shown that the virus persistence in the liver leads to the expression of proinflammatory cytokines (TNF-α, IL-6) and intracellular proteases (lysozyme, cathepsin D, and myeloperoxidase) by Kupffer cells. Defective antiviral response exacerbates destructive processes in the liver accelerating the development of liver failure.
Collapse
|
45
|
Receptor binding and transmission studies of H5N1 influenza virus in mammals. Emerg Microbes Infect 2013; 2:e85. [PMID: 26038448 PMCID: PMC3880874 DOI: 10.1038/emi.2013.89] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 11/10/2013] [Accepted: 11/18/2013] [Indexed: 02/04/2023]
Abstract
The H5N1 influenza A virus that is currently circulating in Asia, Africa and Europe has resulted in persistent outbreaks in poultry with sporadic transmission to humans. Thus far, it is believed that H5N1 does not possess sufficient ability for human-to-human transmission and subsequent pandemic infection. Both receptor binding specificity and virus infectivity are key factors in determining whether influenza A virus becomes pandemic. The use of human viral isolates in various studies has helped to illustrate the changes in receptor binding specificity and virulence as a result of adaptation in humans. In this review, we highlight the important amino acids and domains of viral proteins related to receptor binding specificity that have been reported for humans and avians using mammalian models. Thus, this review will consolidate findings from studies that have shed light on the receptor binding and transmission characteristics of the H5N1 influenza virus, with the goal of improving our ability to predict the transmission efficiency or pandemic potential of new viral strains.
Collapse
|
46
|
Tropism of avian influenza A (H5N1) virus to mesenchymal stem cells and CD34+ hematopoietic stem cells. PLoS One 2013; 8:e81805. [PMID: 24339969 PMCID: PMC3858287 DOI: 10.1371/journal.pone.0081805] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 10/16/2013] [Indexed: 01/09/2023] Open
Abstract
The presence of abnormal hematologic findings such as lymphopenia, thrombocytopenia, and pancytopenia were diagnosed in severe cases of avian influenza A H5N1. Whether direct viral dissemination to bone marrow (BM) cells causes this phenomenon remains elusive. We explore the susceptibility of the two stem cell types; hematopoietic stem cells (HSCs) and mesenchymal stromal cells (MSCs) isolated from human BM cells or cord blood, to infection with avian H5N1 viruses. For the first time, we demonstrated that the H5N1 virus could productively infect and induce cell death in both human stem cell types. In contrast, these activities were not observed upon human influenza virus infection. We also determined whether infection affects the immunomodulatory function of MSCs. We noted a consequent dysregulation of MSC-mediated immune modulation as observed by high cytokine and chemokine production in H5N1 infected MSCs and monocytes cocultures. These findings provide a better understanding of H5N1 pathogenesis in terms of broad tissue tropism and systemic spread.
Collapse
|
47
|
Short KR, Kroeze EJBV, Fouchier RAM, Kuiken T. Pathogenesis of influenza-induced acute respiratory distress syndrome. THE LANCET. INFECTIOUS DISEASES 2013; 14:57-69. [PMID: 24239327 DOI: 10.1016/s1473-3099(13)70286-x] [Citation(s) in RCA: 402] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a fatal complication of influenza infection. In this Review we provide an integrated model for its pathogenesis. ARDS involves damage to the epithelial-endothelial barrier, fluid leakage into the alveolar lumen, and respiratory insufficiency. The most important part of the epithelial-endothelial barrier is the alveolar epithelium, strengthened by tight junctions. Influenza virus targets these epithelial cells, reducing sodium pump activity, damaging tight junctions, and killing infected cells. Infected epithelial cells produce cytokines that attract leucocytes--neutrophils and macrophages--and activate adjacent endothelial cells. Activated endothelial cells and infiltrated leucocytes stimulate further infiltration, and leucocytes induce production of reactive oxygen species and nitric oxide that damage the barrier. Activated macrophages also cause direct apoptosis of epithelial cells. This model for influenza-induced ARDS differs from the classic model, which is centred on endothelial damage, and provides a rationale for therapeutic intervention to moderate host response in influenza-induced ARDS.
Collapse
Affiliation(s)
- Kirsty R Short
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Ron A M Fouchier
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Thijs Kuiken
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
48
|
Surveillance of the first case of human avian influenza A (H7N9) virus in Beijing, China. Infection 2013; 42:127-33. [PMID: 24129555 PMCID: PMC7102393 DOI: 10.1007/s15010-013-0533-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 09/10/2013] [Indexed: 11/25/2022]
Abstract
Purpose Human infections with avian influenza A (H7N9) virus manifested in China in March 2013. The first case infected with H7N9 virus in Beijing involved a family member of a chicken dealer and was reported in April 2013. The clinical and epidemiological characteristics of this case and her parents were examined to illustrate some key traits regarding this novel H7N9 virus. Methods The index case was subjected to intensive clinical examination in order to observed the clinical process. Real-time PCR was used to confirm cases infected with H7N9 virus. The index case was administered oseltamivir (45 mg, twice daily) at the early stage of the infection. Sera were collected from the index case and her parents from the onset of illness onwards. The subjects were followed for 4 weeks. Results
The sera were confirmed by neutralizing antibody tests. The index case’s clinical manifestation progressed quickly. The pharyngeal swab tested positive for influenza A based on the detection of influenza A antigen (rapid influenza diagnostic test) 15 h after the onset of fever and was positive for H7N9 virus. The patient’s temperature dropped to 36.2 °C 18 h after treatment by oseltamivir (32 h after fever). Cough and other symptoms alleviated rapidly. A number of specimens from the environment of this cluster and from the feces specimens tested positive for viral RNA of the H7N9 virus on the fourth day following onset of the index case’s illness. Pharyngeal swabs of the mother tested positive for H7N9 virus twice, but she showed no clinical symptoms. Four weeks after disease onset, the family did not present any clinical symptoms, and the results of the physical examination and blood tests were normal. The mother and the case’s sera had a fourfold increased neutralizing antibody titer. Conclusion Early diagnosis and early initiation of the treatment of confirmed infections is the most effective strategy for managing H7N9 virus infection. Human beings exposed to H7N9 virus may develop asymptomatic infection.
Collapse
|
49
|
Liu Q, Liu DY, Yang ZQ. Characteristics of human infection with avian influenza viruses and development of new antiviral agents. Acta Pharmacol Sin 2013; 34:1257-69. [PMID: 24096642 PMCID: PMC3791557 DOI: 10.1038/aps.2013.121] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/01/2013] [Indexed: 12/21/2022]
Abstract
Since 1997, several epizootic avian influenza viruses (AIVs) have been transmitted to humans, causing diseases and even deaths. The recent emergence of severe human infections with AIV (H7N9) in China has raised concerns about efficient interpersonal viral transmission, polygenic traits in viral pathogenicity and the management of newly emerging strains. The symptoms associated with viral infection are different in various AI strains: H5N1 and newly emerged H7N9 induce severe pneumonia and related complications in patients, while some H7 and H9 subtypes cause only conjunctivitis or mild respiratory symptoms. The virulence and tissue tropism of viruses as well as the host responses contribute to the pathogenesis of human AIV infection. Several preventive and therapeutic approaches have been proposed to combat AIV infection, including antiviral drugs such as M2 inhibitors, neuraminidase inhibitors, RNA polymerase inhibitors, attachment inhibitors and signal-transduction inhibitors etc. In this article, we summarize the recent progress in researches on the epidemiology, clinical features, pathogenicity determinants, and available or potential antivirals of AIV.
Collapse
Affiliation(s)
- Qiang Liu
- State Key Laboratory of Virology/Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan 430071, China
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang 443000, China
| | - Dong-ying Liu
- State Key Laboratory of Virology/Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan 430071, China
- Department of Microbiology, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Zhan-qiu Yang
- State Key Laboratory of Virology/Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan 430071, China
| |
Collapse
|
50
|
Decrease of virus receptors during highly pathogenic H5N1 virus infection in humans and other mammals. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1382-1389. [PMID: 23993779 DOI: 10.1016/j.ajpath.2013.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 06/18/2013] [Accepted: 07/12/2013] [Indexed: 01/28/2023]
Abstract
Highly pathogenic avian influenza H5N1 virus causes a severe, often fatal, pneumonia in humans. The tropism and pathogenesis of highly pathogenic avian influenza H5N1 virus can partly be explained by the presence of H5N1 virus receptors in the human alveoli, which are the site of inflammation during pneumonia. Although studies on the distribution of influenza virus receptors in normal respiratory tract tissues have provided significant insights into the cell tropism and pathogenesis of influenza viruses, the distribution of influenza virus receptors have not been studied during influenza virus infection. Therefore, we studied the distribution of H5N1 virus receptors, by virus and lectin histochemistry, during highly pathogenic avian influenza H5N1 virus infection in alveolar tissues of humans, macaques, ferrets, and cats. In all species, we observed a decrease of H5N1 virus receptors in influenza virus-infected and neighboring cells. The observed decrease of H5N1 virus receptors was associated with the presence of MxA, a known marker for interferon activity. Taken together, our data suggest that the decrease of H5N1 virus receptors might be part of a defense mechanism that limits viral replication in the lower respiratory tract.
Collapse
|