1
|
Stegeman SK, Kourko O, Amsden H, Pellizzari Delano IE, Mamatis JE, Roth M, Colpitts CC, Gee K. RNA Viruses, Toll-Like Receptors, and Cytokines: The Perfect Storm? J Innate Immun 2025; 17:126-153. [PMID: 39820070 PMCID: PMC11845175 DOI: 10.1159/000543608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs. BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs.
Collapse
Affiliation(s)
- Sophia K Stegeman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Heather Amsden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Madison Roth
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
2
|
Gil A, Hoag GE, Salerno JP, Hornig M, Klimas N, Selin LK. Identification of CD8 T-cell dysfunction associated with symptoms in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and Long COVID and treatment with a nebulized antioxidant/anti-pathogen agent in a retrospective case series. Brain Behav Immun Health 2024; 36:100720. [PMID: 38327880 PMCID: PMC10847863 DOI: 10.1016/j.bbih.2023.100720] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 02/09/2024] Open
Abstract
Background Patients with post-acute sequelae of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection (PASC, i.e., Long COVID) have a symptom complex highly analogous to many features of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), suggesting they may share some aspects of pathogenesis in these similar disorders. ME/CFS is a complex disease affecting numerous organ systems and biological processes and is often preceded by an infection-like episode. It is postulated that the chronic manifestations of illness may result from an altered host response to infection or inability to resolve inflammation, as is being reported in Long COVID. The immunopathogenesis of both disorders is still poorly understood. Here, we show data that suggest Long COVID and ME/CFS may be due to an aberrant response to an immunological trigger-like infection, resulting in a dysregulated immune system with CD8 T-cell dysfunction reminiscent of some aspects of T-cell clonal exhaustion, a phenomenon associated with oxidative stress. As there is an urgent need for diagnostic tools and treatment strategies for these two related disabling disorders, here, in a retrospective case series, we have also identified a potential nebulized antioxidant/anti-pathogen treatment that has evidence of a good safety profile. This nebulized agent is comprised of five ingredients previously reported individually to relieve oxidative stress, attenuate NF-κB signaling, and/or to act directly to inhibit pathogens, including viruses. Administration of this treatment by nebulizer results in rapid access of small doses of well-studied antioxidants and agents with anti-pathogen potential to the lungs; components of this nebulized agent are also likely to be distributed systemically, with potential to enter the central nervous system. Methods and Findings: We conducted an analysis of CD8 T-cell function and severity of symptoms by self-report questionnaires in ME/CFS, Long COVID and healthy controls. We developed a CD8 T-cell functional assay, assessing CD8 T-cell dysfunction by intracellular cytokine staining (ICS) in a group of ME/CFS (n = 12) and Long COVID patients (n = 8), comparing to healthy controls (HC) with similar age and sex (n = 10). Magnet-enriched fresh CD8 T-cells in both patient groups had a significantly diminished capacity to produce both cytokines, IFNγ or TNFα, after PMA stimulation when compared to HC. The symptom severity questionnaire showed similar symptom profiles for the two disorders. Fortuitously, through a retrospective case series, we were able to examine the ICS and questionnaire data of 4 ME/CFS and 4 Long COVID patients in conjunction with their treatment (3-15 months). In parallel with the treatment pursued electively by participants in this retrospective case series, there was an increase in CD8 T-cell IFNγ and TNFα production and a decrease in overall self-reported symptom severity score by 54%. No serious treatment-associated side effects or laboratory anomalies were noted in these patients. Conclusions Here, in this small study, we present two observations that appear potentially fundamental to the pathogenesis and treatment of Long COVID and ME/CFS. The first is that both disorders appear to be characterized by dysfunctional CD8 T-cells with severe deficiencies in their abilities to produce IFNγ and TNFα. The second is that in a small retrospective Long COVID and ME/CFS case series, this immune dysfunction and patient health improved in parallel with treatment with an immunomodulatory, antioxidant pharmacological treatment with anticipated anti-pathogen activity. This work provides evidence of the potential utility of a biomarker, CD8 T-cell dysfunction, and suggests the potential for benefit from a new nebulized antioxidant/anti-pathogen treatment. These immune biomarker data may help build capacity for improved diagnosis and tracking of treatment outcomes during clinical trials for both Long COVID and ME/CFS while providing clues to new treatment avenues that suggest potential efficacy for both conditions.
Collapse
Affiliation(s)
- Anna Gil
- University of Massachusetts Chan Medical School, Department of Pathology, Worcester, MA, USA
| | | | - John P. Salerno
- Inspiritol, Inc., Fairfield, CT, USA
- The Salerno Center for Complementary Medicine, New York, USA
| | - Mady Hornig
- Columbia University Mailman School of Public Health, New York, USA
| | - Nancy Klimas
- Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Liisa K. Selin
- University of Massachusetts Chan Medical School, Department of Pathology, Worcester, MA, USA
| |
Collapse
|
3
|
The Degree of Hydroxylation of Phenolic Rings Determines the Ability of Flavonoids and Stilbenes to Inhibit Calcium-Mediated Membrane Fusion. Nutrients 2023; 15:nu15051121. [PMID: 36904120 PMCID: PMC10005302 DOI: 10.3390/nu15051121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/11/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
This paper discusses the possibility of using plant polyphenols as viral fusion inhibitors with a lipid-mediated mechanism of action. The studied agents are promising candidates for the role of antiviral compounds due to their high lipophilicity, low toxicity, bioavailability, and relative cheapness. Fluorimetry of calcein release at the calcium-mediated fusion of liposomes, composed of a ternary mixture of dioleoyl phosphatidylcholine, dioleoyl phosphatidylglycerol, and cholesterol, in the presence of 4'-hydroxychalcone, cardamonin, isoliquiritigenin, phloretin, resveratrol, piceatannol, daidzein, biochanin A, genistein, genistin, liquiritigenin, naringenin, catechin, taxifolin, and honokiol, was performed. It was found that piceatannol significantly inhibited the calcium-induced fusion of negatively charged vesicles, while taxifolin and catechin showed medium and low antifusogenic activity, respectively. As a rule, polyphenols containing at least two OH-groups in both phenolic rings were able to inhibit the calcium-mediated fusion of liposomes. In addition, there was a correlation between the ability of the tested compounds to inhibit vesicle fusions and to perturb lipid packing. We suggest that the antifusogenic action of polyphenols was determined by the depth of immersion and the orientation of the molecules in the membrane.
Collapse
|
4
|
Modelling within-host macrophage dynamics in influenza virus infection. J Theor Biol 2020; 508:110492. [PMID: 32966828 DOI: 10.1016/j.jtbi.2020.110492] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022]
Abstract
Human respiratory disease associated with influenza virus infection is of significant public health concern. Macrophages, as part of the front line of host innate cellular defence, have been shown to play an important role in controlling viral replication. However, fatal outcomes of infection, as evidenced in patients infected with highly pathogenic viral strains, are often associated with prompt activation and excessive accumulation of macrophages. Activated macrophages can produce a large amount of pro-inflammatory cytokines, which leads to severe symptoms and at times death. However, the mechanism for rapid activation and excessive accumulation of macrophages during infection remains unclear. It has been suggested that the phenomena may arise from complex interactions between macrophages and influenza virus. In this work, we develop a novel mathematical model to study the relationship between the level of macrophage activation and the level of viral load in influenza infection. Our model combines a dynamic model of viral infection, a dynamic model of macrophages and the essential interactions between the virus and macrophages. Our model predicts that the level of macrophage activation can be negatively correlated with the level of viral load when viral infectivity is sufficiently high. We further identify that temporary depletion of resting macrophages in response to viral infection is a major driver in our model for the negative relationship between the level of macrophage activation and viral load, providing new insight into the mechanisms that regulate macrophage activation. Our model serves as a framework to study the complex dynamics of virus-macrophage interactions and provides a mechanistic explanation for existing experimental observations, contributing to an enhanced understanding of the role of macrophages in influenza viral infection.
Collapse
|
5
|
LeMessurier KS, Tiwary M, Morin NP, Samarasinghe AE. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front Immunol 2020; 11:3. [PMID: 32117216 PMCID: PMC7011736 DOI: 10.3389/fimmu.2020.00003] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022] Open
Abstract
The primary function of the respiratory system of gas exchange renders it vulnerable to environmental pathogens that circulate in the air. Physical and cellular barriers of the respiratory tract mucosal surface utilize a variety of strategies to obstruct microbe entry. Physical barrier defenses including the surface fluid replete with antimicrobials, neutralizing immunoglobulins, mucus, and the epithelial cell layer with rapidly beating cilia form a near impenetrable wall that separates the external environment from the internal soft tissue of the host. Resident leukocytes, primarily of the innate immune branch, also maintain airway integrity by constant surveillance and the maintenance of homeostasis through the release of cytokines and growth factors. Unfortunately, pathogens such as influenza virus and Streptococcus pneumoniae require hosts for their replication and dissemination, and prey on the respiratory tract as an ideal environment causing severe damage to the host during their invasion. In this review, we outline the host-pathogen interactions during influenza and post-influenza bacterial pneumonia with a focus on inter- and intra-cellular crosstalk important in pulmonary immune responses.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Meenakshi Tiwary
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Nicholas P Morin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Critical Care Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amali E Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| |
Collapse
|
6
|
Downey J, Pernet E, Coulombe F, Divangahi M. Dissecting host cell death programs in the pathogenesis of influenza. Microbes Infect 2018; 20:560-569. [PMID: 29679740 PMCID: PMC7110448 DOI: 10.1016/j.micinf.2018.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Abstract
Influenza A virus (IAV) is a pulmonary pathogen, responsible for significant yearly morbidity and mortality. Due to the absence of highly effective antiviral therapies and vaccine, as well as the constant threat of an emerging pandemic strain, there is considerable need to better understand the host-pathogen interactions and the factors that dictate a protective versus detrimental immune response to IAV. Even though evidence of IAV-induced cell death in human pulmonary epithelial and immune cells has been observed for almost a century, very little is known about the consequences of cell death on viral pathogenesis. Recent study indicates that both the type of cell death program and its kinetics have major implications on host defense and survival. In this review, we discuss advances in our understanding of cell death programs during influenza virus infection, in hopes of fostering new areas of investigation for targeted clinical intervention.
Collapse
Affiliation(s)
- Jeffrey Downey
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Erwan Pernet
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - François Coulombe
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Maziar Divangahi
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| |
Collapse
|
7
|
Petersen H, Mostafa A, Tantawy MA, Iqbal AA, Hoffmann D, Tallam A, Selvakumar B, Pessler F, Beer M, Rautenschlein S, Pleschka S. NS Segment of a 1918 Influenza A Virus-Descendent Enhances Replication of H1N1pdm09 and Virus-Induced Cellular Immune Response in Mammalian and Avian Systems. Front Microbiol 2018; 9:526. [PMID: 29623073 PMCID: PMC5874506 DOI: 10.3389/fmicb.2018.00526] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/08/2018] [Indexed: 12/14/2022] Open
Abstract
The 2009 pandemic influenza A virus (IAV) H1N1 strain (H1N1pdm09) has widely spread and is circulating in humans and swine together with other human and avian IAVs. This fact raises the concern that reassortment between H1N1pdm09 and co-circulating viruses might lead to an increase of H1N1pdm09 pathogenicity in different susceptible host species. Herein, we explored the potential of different NS segments to enhance the replication dynamics, pathogenicity and host range of H1N1pdm09 strain A/Giessen/06/09 (Gi-wt). The NS segments were derived from (i) human H1N1- and H3N2 IAVs, (ii) highly pathogenic- (H5- or H7-subtypes) or (iii) low pathogenic avian influenza viruses (H7- or H9-subtypes). A significant increase of growth kinetics in A549 (human lung epithelia) and NPTr (porcine tracheal epithelia) cells was only noticed in vitro for the reassortant Gi-NS-PR8 carrying the NS segment of the 1918-descendent A/Puerto Rico/8/34 (PR8-wt, H1N1), whereas all other reassortants showed either reduced or comparable replication efficiencies. Analysis using ex vivo tracheal organ cultures of turkeys (TOC-Tu), a species susceptible to IAV H1N1 infection, demonstrated increased replication of Gi-NS-PR8 compared to Gi-wt. Also, Gi-NS-PR8 induced a markedly higher expression of immunoregulatory and pro-inflammatory cytokines, chemokines and interferon-stimulated genes in A549 cells, THP-1-derived macrophages (dHTP) and TOC-Tu. In vivo, Gi-NS-PR8 induced an earlier onset of mortality than Gi-wt in mice, whereas, 6-week-old chickens were found to be resistant to both viruses. These data suggest that the specific characteristics of the PR8 NS segments can impact on replication, virus induced cellular immune responses and pathogenicity of the H1N1pdm09 in different avian and mammalian host species.
Collapse
Affiliation(s)
- Henning Petersen
- Clinic for Poultry, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Ahmed Mostafa
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany.,Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC), Cairo, Egypt
| | - Mohamed A Tantawy
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Hanover, Germany.,Department of Hormones, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Azeem A Iqbal
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Hanover, Germany.,Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Aravind Tallam
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Hanover, Germany
| | - Balachandar Selvakumar
- Max-Planck Laboratory for Heart and Lung Research, Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Frank Pessler
- Institute for Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Hanover, Germany.,Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Stephan Pleschka
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
8
|
Critical Role of HAX-1 in Promoting Avian Influenza Virus Replication in Lung Epithelial Cells. Mediators Inflamm 2018; 2018:3586132. [PMID: 29576744 PMCID: PMC5822872 DOI: 10.1155/2018/3586132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/21/2017] [Accepted: 09/17/2017] [Indexed: 02/04/2023] Open
Abstract
The PB1-F2 protein of influenza A virus has been considered a virulence factor, but its function in inducing apoptosis may be of disadvantage to viral replication. Host mechanisms to regulate PB1-F2-induced apoptosis remain unknown. We generated a PB1-F2-deficient avian influenza virus (AIV) H9N2 and found that the mutant virus replicated less efficiently in human lung epithelial cells. The PB1-F2-deficient virus produced less apoptotic cells, indicating that PB1-F2 of the H9N2 virus promotes apoptosis, occurring at the early stage of infection, in the lung epithelial cells. To understand how host cells regulate PB1-F2-induced apoptosis, we explored to identify cellular proteins interacting with PB1-F2 and found that HCLS1-associated protein X-1 (HAX-1), located mainly in the mitochondria as an apoptotic inhibitor, interacted with PB1-F2. Increased procaspase-9 activations, induced by PB1-F2, could be suppressed by HAX-1. In HAX-1 knockdown A549 cells, the replication of AIV H9N2 was suppressed in parallel to the activation of caspase-3 activation, which increased at the early stage of infection. We hypothesize that HAX-1 promotes AIV replication by interacting with PB1-F2, resulting in the suppression of apoptosis, prolonged cell survival, and enhancement of viral replication. Our data suggest that HAX-1 may be a promoting factor for AIV H9N2 replication through desensitizing PB1-F2 from its apoptotic induction in human lung epithelial cells.
Collapse
|
9
|
Cline TD, Beck D, Bianchini E. Influenza virus replication in macrophages: balancing protection and pathogenesis. J Gen Virol 2017; 98:2401-2412. [PMID: 28884667 DOI: 10.1099/jgv.0.000922] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Macrophages are essential for protection against influenza A virus infection, but are also implicated in the morbidity and mortality associated with severe influenza disease, particularly during infection with highly pathogenic avian influenza (HPAI) H5N1 virus. While influenza virus infection of macrophages was once thought to be abortive, it is now clear that certain virus strains can replicate productively in macrophages. This may have important consequences for the antiviral functions of macrophages, the course of disease and the outcome of infection for the host. In this article, we review findings related to influenza virus replication in macrophages and the impact of productive replication on macrophage antiviral functions. A clear understanding of the interactions between influenza viruses and macrophages may lead to new antiviral therapies to relieve the burden of severe disease associated with influenza viruses.
Collapse
Affiliation(s)
- Troy D Cline
- Department of Biological Sciences, California State University, Chico, California, USA
| | - Donald Beck
- Department of Biological Sciences, California State University, Chico, California, USA
| | - Elizabeth Bianchini
- Department of Biological Sciences, California State University, Chico, California, USA
| |
Collapse
|
10
|
Xu Y, Liu L. Curcumin alleviates macrophage activation and lung inflammation induced by influenza virus infection through inhibiting the NF-κB signaling pathway. Influenza Other Respir Viruses 2017. [PMID: 28646616 PMCID: PMC5596526 DOI: 10.1111/irv.12459] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Influenza A viruses (IAV) result in severe public health problems with worldwide each year. Overresponse of immune system to IAV infection leads to complications, and ultimately causing morbidity and mortality. OBJECTIVE Curcumin has been reported to have anti-inflammatory ability. However, its molecular mechanism in immune responses remains unclear. METHODS We detected the pro-inflammatory cytokine secretion and nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB)-related protein expression in human macrophages or mice infected by IAV with or without curcumin treatment. RESULTS We found that the IAV infection caused a dramatic enhancement of pro-inflammatory cytokine productions of human macrophages and mice immune cells. However, curcumin treatment after IAV infection downregulated these cytokines production in a dose-dependent manner. Moreover, the NF-κB has been activated in human macrophages after IAV infection, while administration of curcumin inhibited NF-κB signaling pathway via promoting the expression of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα), and inhibiting the translocation of p65 from cytoplasm to nucleus. CONCLUSIONS In summary, IAV infection could result in the inflammatory responses of immune cells, especially macrophages. Curcumin has the therapeutic potentials to relieve these inflammatory responses through inhibiting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yiming Xu
- Department of Respiration Medicine, The Affiliated Wuxi Second People's Hospital of Nanjing Medical University, Wuxi, China
| | - Ling Liu
- Department of Respiration Medicine, The Affiliated Wuxi Second People's Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
11
|
Contribution of innate immune cells to pathogenesis of severe influenza virus infection. Clin Sci (Lond) 2017; 131:269-283. [PMID: 28108632 DOI: 10.1042/cs20160484] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/19/2016] [Accepted: 11/25/2016] [Indexed: 12/12/2022]
Abstract
Influenza A viruses (IAVs) cause respiratory illness of varying severity based on the virus strains, host predisposition and pre-existing immunity. Ultimately, outcome and recovery from infection rely on an effective immune response comprising both innate and adaptive components. The innate immune response provides the first line of defence and is crucial to the outcome of infection. Airway epithelial cells are the first cell type to encounter the virus in the lungs, providing antiviral and chemotactic molecules that shape the ensuing immune response by rapidly recruiting innate effector cells such as NK cells, monocytes and neutrophils. Each cell type has unique mechanisms to combat virus-infected cells and limit viral replication, however their actions may also lead to pathology. This review focuses how innate cells contribute to protection and pathology, and provides evidence for their involvement in immune pathology in IAV infections.
Collapse
|
12
|
Wang Y, Zhou B, Lu J, Chen Q, Ti H, Huang W, Li J, Yang Z, Jiang Z, Wang X. Inhibition of influenza virus via a sesquiterpene fraction isolated from Laggera pterodonta by targeting the NF-κB and p38 pathways. Altern Ther Health Med 2017; 17:25. [PMID: 28061784 PMCID: PMC5217203 DOI: 10.1186/s12906-016-1528-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/08/2016] [Indexed: 11/14/2022]
Abstract
Background Influenza virus poses serious threats to human health, especially human infection with avian influenza virus. Laggera pterodonta (DC.) Benth is a medicinal plant that is widely used in Traditional Chinese Medicine, especially in Yunnan province, and has been used to treat influenza, pharyngolaryngitis, and bronchitis. However, the compound(s) responsible for the activity and their mechanisms of action against the influenza virus remain to be elucidated. Methods L. pterodonta extract was fractionated, and the active fraction was identified as Fraction 14 (Fr 14). Fr 14 was further analysed and characterized by ultra-high-performance liquid chromatography hyphenated with quadrupole-time of flight mass spectrometry (UHPLC/Q-TOF-MS). The inhibitory effect against influenza virus was evaluated using a cytotoxicity assay. Then, cytokines and chemokines were detected by qRT-PCR and a bio-plex assay. Signalling pathways that inhibited the influenza virus were identified using a western blotting assay. Results The active fr 14 showed a wide spectrum of anti-influenza virus activity. The pharmacological mechanisms showed that Fr 14 acts on the early stage of virus replication (0–6 h). It inhibited the p38/MAPK pathway and then inhibited the NF-κB pathway and COX-2. Fr 14 also prevented the increased expression of cytokines and chemokines. Conclusion This study demonstrated the preliminary mechanisms of fr 14 against the influenza virus. Fr 14 possessed antiviral and anti-inflammatory effects. L. pterodonta can be used to develop innovative antiviral drugs, and further studies will be performed to illustrate the detailed mechanisms.
Collapse
|
13
|
G45R on nonstructural protein 1 of influenza A virus contributes to virulence by increasing the expression of proinflammatory cytokines in mice. Arch Virol 2016; 162:45-55. [PMID: 27664027 DOI: 10.1007/s00705-016-3072-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/15/2016] [Indexed: 01/15/2023]
Abstract
Nonstructural protein 1 (NS1) is a multifunctional protein that is a viral replication enhancer and virulence factor. In this study, we investigated the effect of the amino acid substitution G45R on the NS1 of A/Puerto Rico/8/1934 (H1N1) (G45R/NS1) on viral virulence and host gene expression in a mouse model and the human lung cell line A549. The G45R/NS1 virus had increased virulence by inducing an earlier and robust proinflammatory cytokine response in mice. Mice infected with the G45R/NS1 virus lost more body weight and had lower survival rates than mice infected with the wild type (WT/NS1) virus. Replication of the G45R/NS1 virus was higher than that of the WT/NS1 virus in vitro, but the replication of both viruses was similar in mouse lungs. In A549 cells, the majority of G45R/NS1 protein was localized in the cytoplasm whereas the majority of WT/NS1 protein was localized in the nucleus. Microarray analysis revealed that A549 cells infected with the G45R/NS1 virus had higher expression of genes encoding proteins associated with the innate immune response and cytokine activity than cells infected with the WT/NS1 virus. These data agree with cytokine production observed in mouse lungs. Our findings suggest that G45R on NS1 protein contributes to viral virulence by increasing the expression of inflammatory cytokines early in infection.
Collapse
|
14
|
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
|
15
|
Rose JJ, Voora D, Cyr DD, Lucas JE, Zaas AK, Woods CW, Newby LK, Kraus WE, Ginsburg GS. Gene Expression Profiles Link Respiratory Viral Infection, Platelet Response to Aspirin, and Acute Myocardial Infarction. PLoS One 2015; 10:e0132259. [PMID: 26193668 PMCID: PMC4507878 DOI: 10.1371/journal.pone.0132259] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/12/2015] [Indexed: 01/09/2023] Open
Abstract
Background Influenza infection is associated with myocardial infarction (MI), suggesting that respiratory viral infection may induce biologic pathways that contribute to MI. We tested the hypotheses that 1) a validated blood gene expression signature of respiratory viral infection (viral GES) was associated with MI and 2) respiratory viral exposure changes levels of a validated platelet gene expression signature (platelet GES) of platelet function in response to aspirin that is associated with MI. Methods A previously defined viral GES was projected into blood RNA data from 594 patients undergoing elective cardiac catheterization and used to classify patients as having evidence of viral infection or not and tested for association with acute MI using logistic regression. A previously defined platelet GES was projected into blood RNA data from 81 healthy subjects before and after exposure to four respiratory viruses: Respiratory Syncytial Virus (RSV) (n=20), Human Rhinovirus (HRV) (n=20), Influenza A virus subtype H1N1 (H1N1) (n=24), Influenza A Virus subtype H3N2 (H3N2) (n=17). We tested for the change in platelet GES with viral exposure using linear mixed-effects regression and by symptom status. Results In the catheterization cohort, 32 patients had evidence of viral infection based upon the viral GES, of which 25% (8/32) had MI versus 12.2% (69/567) among those without evidence of viral infection (OR 2.3; CI [1.03-5.5], p=0.04). In the infection cohorts, only H1N1 exposure increased platelet GES over time (time course p-value = 1e-04). Conclusions A viral GES of non-specific, respiratory viral infection was associated with acute MI; 18% of the top 49 genes in the viral GES are involved with hemostasis and/or platelet aggregation. Separately, H1N1 exposure, but not exposure to other respiratory viruses, increased a platelet GES previously shown to be associated with MI. Together, these results highlight specific genes and pathways that link viral infection, platelet activation, and MI especially in the case of H1N1 influenza infection.
Collapse
Affiliation(s)
- Jason J. Rose
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Deepak Voora
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Derek D. Cyr
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Joseph E. Lucas
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Aimee K. Zaas
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Christopher W. Woods
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - L. Kristin Newby
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - William E. Kraus
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Geoffrey S. Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
16
|
Nasal priming with immunobiotic Lactobacillus rhamnosus modulates inflammation-coagulation interactions and reduces influenza virus-associated pulmonary damage. Inflamm Res 2015; 64:589-602. [PMID: 26072063 DOI: 10.1007/s00011-015-0837-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 05/29/2015] [Accepted: 05/29/2015] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE To evaluate the effect of the nasal administration of live and heat-killed Lactobacillus rhamnosus CRL1505 (Lr1505) on immune-coagulative response during influenza virus (IFV) infection to improve survival and reduce lung injury. METHODS Six-week-old BALB/c mice were treated with live or heat-killed Lr1505 by the nasal route during two consecutive days. Treated and untreated control mice were then nasally challenged with IFV. RESULTS Both viable and non-viable Lr1505 protected infected mice by reducing pulmonary injury and lung viral loads trough several mechanisms: (a) Inflammatory cytokines were efficiently regulated allowing higher clearance of virus and reduction of inflammatory lung tissue damage, associated to higher levels of the regulatory cytokine IL-10. (b) The antiviral immune response was enhanced with improved levels of type I interferons, CD4(+)IFN-γ(+) lymphocytes, and lung CD11c(+)CD11b(low)CD103(+) and CD11c(+)CD11b(high)CD103(-) dendritic cells. (c) The procoagulant state was reversed mainly by down-regulating tissue factor expression and restoring thrombomodulin levels in lung. The capacity of Lr1505 to improve the outcome of IFV infection would be related to its ability to beneficially modulate lung TLR3-triggered immune response. CONCLUSIONS Our work is the first to demonstrate the ability of an immunobiotic strain to beneficially modulate inflammation-coagulation interactions during IFV infection. Interestingly, non-viable L. rhamnosus CRL1505 was as effective as the viable strain to beneficially modulate respiratory antiviral immune response.
Collapse
|
17
|
Zarubaev VV, Garshinina AV, Tretiak TS, Fedorova VA, Shtro AA, Sokolova AS, Yarovaya OI, Salakhutdinov NF. Broad range of inhibiting action of novel camphor-based compound with anti-hemagglutinin activity against influenza viruses in vitro and in vivo. Antiviral Res 2015; 120:126-33. [PMID: 26072310 DOI: 10.1016/j.antiviral.2015.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 10/23/2022]
Abstract
Influenza virus continues to remain one of the leading human respiratory pathogens causing significant morbidity and mortality around the globe. Due to short-term life cycle and high rate of mutations influenza virus is able to rapidly develop resistance to clinically available antivirals. This makes necessary the search and development of new drugs with different targets and mechanisms of activity. Here we report anti-influenza activity of camphor derivative 1,7,7-trimethylbicyclo[2.2.1]heptan-2-ylidene-aminoethanol (camphecene). In in vitro experiments it inhibited influenza viruses A(H1, H1pdm09, H3 and H5 subtypes) and B with EC50's lying in micromolar range. Due to low cytotoxicity it resulted in high selectivity indices (74-661 depending on the virus). This effect did not depend on susceptibility or resistance of the viruses to adamantane derivatives amantadine and rimantadine. The compound appeared the most effective when added at the early stages of viral life cycle (0-2h p.i.). In direct hemagglutinin inhibition tests camphecene was shown to decrease the activity of HA's of influenza viruses A and B. The activity of camphecene was further confirmed in experiments with influenza virus-infected mice, in which, being used orally by therapeutic schedule (once a day, days 1-5 p.i.) it decreased specific mortality of animals infected with both influenza A and B viruses (highest indices of protection 66.7% and 88.9%, respectively). Taken together, these results are encouraging for further development of camphecene-based drug(s) and for exploration of camphor derivatives as highly prospective group of potential antivirals.
Collapse
Affiliation(s)
- V V Zarubaev
- Influenza Research Institute, Prof. Popova 15/17, 197376 St. Petersburg, Russia.
| | - A V Garshinina
- Influenza Research Institute, Prof. Popova 15/17, 197376 St. Petersburg, Russia
| | - T S Tretiak
- Influenza Research Institute, Prof. Popova 15/17, 197376 St. Petersburg, Russia
| | - V A Fedorova
- Influenza Research Institute, Prof. Popova 15/17, 197376 St. Petersburg, Russia
| | - A A Shtro
- Influenza Research Institute, Prof. Popova 15/17, 197376 St. Petersburg, Russia
| | - A S Sokolova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentjev Avenue 9, 630090 Novosibirsk, Russia; Novosibirsk State University, Pirogova St. 2, 630090 Novosibirsk, Russia
| | - O I Yarovaya
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentjev Avenue 9, 630090 Novosibirsk, Russia; Novosibirsk State University, Pirogova St. 2, 630090 Novosibirsk, Russia
| | - N F Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentjev Avenue 9, 630090 Novosibirsk, Russia; Novosibirsk State University, Pirogova St. 2, 630090 Novosibirsk, Russia
| |
Collapse
|
18
|
James SJ, Jiao H, Teh HY, Takahashi H, Png CW, Phoon MC, Suzuki Y, Sawasaki T, Xiao H, Chow VTK, Yamamoto N, Reynolds JM, Flavell RA, Dong C, Zhang Y. MAPK Phosphatase 5 Expression Induced by Influenza and Other RNA Virus Infection Negatively Regulates IRF3 Activation and Type I Interferon Response. Cell Rep 2015; 10:1722-1734. [PMID: 25772359 DOI: 10.1016/j.celrep.2015.02.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/20/2015] [Accepted: 02/08/2015] [Indexed: 11/17/2022] Open
Abstract
The type I interferon system is essential for antiviral immune response and is a primary target of viral immune evasion strategies. Here, we show that virus infection induces the expression of MAPK phosphatase 5 (MKP5), a dual-specificity phosphatase (DUSP), in host cells. Mice deficient in MKP5 were resistant to H1N1 influenza infection, which is associated with increased IRF3 activation and type I interferon expression in comparison with WT mice. Increased type I interferon responses were also observed in MKP5-deficient cells and animals upon other RNA virus infection, including vesicular stomatitis virus and sendai virus. These observations were attributed to the ability of MKP5 to interact with and dephosphorylate IRF3. Our study reveals a critical function of a DUSP in negative regulation of IRF3 activity and demonstrates a mechanism by which influenza and other RNA viruses inhibit type I interferon response in the host through MKP5.
Collapse
Affiliation(s)
- Sharmy J James
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore; Immunology Progamme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Huipeng Jiao
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore; Immunology Progamme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Hong-Ying Teh
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore; Immunology Progamme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Hirotaka Takahashi
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore; Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Chin Wen Png
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore; Immunology Progamme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Meng Chee Phoon
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore
| | - Youichi Suzuki
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore
| | - Tatsuy Sawasaki
- Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Hui Xiao
- Unit of Immune Regulation and Signaling, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Vincent T K Chow
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore
| | - Naoki Yamamoto
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore
| | - Joseph M Reynolds
- Department of Microbiology & Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Chicago, IL 60064, USA
| | - Richard A Flavell
- Department of Immunology, Howard Hughes Medical Institute, Yale University, New Haven, CT 06520, USA
| | - Chen Dong
- Tsinghua University, Beijing 100084, China
| | - Yongliang Zhang
- Department of Microbiology, Yong Loo Lin School of Medicine, Singapore 117597, Singapore; Immunology Progamme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
19
|
Abstract
Influenza viruses pose a substantial threat to human and animal health worldwide. Recent studies in mouse models have revealed an indispensable role for the innate immune system in defense against influenza virus. Recognition of the virus by innate immune receptors in a multitude of cell types activates intricate signaling networks, functioning to restrict viral replication. Downstream effector mechanisms include activation of innate immune cells and, induction and regulation of adaptive immunity. However, uncontrolled innate responses are associated with exaggerated disease, especially in pandemic influenza virus infection. Despite advances in the understanding of innate response to influenza in the mouse model, there is a large knowledge gap in humans, particularly in immunocompromised groups such as infants and the elderly. We propose here, the need for further studies in humans to decipher the role of innate immunity to influenza virus, particularly at the site of infection. These studies will complement the existing work in mice and facilitate the quest to design improved vaccines and therapeutic strategies against influenza.
Collapse
Affiliation(s)
- Michael B. A. Oldstone
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California USA
| | - Richard W. Compans
- IDepartment of Microbiology and Immunology, Emory University, Atlanta, Georgia USA
| |
Collapse
|
20
|
Jain S, Gitter A, Bar-Joseph Z. Multitask learning of signaling and regulatory networks with application to studying human response to flu. PLoS Comput Biol 2014; 10:e1003943. [PMID: 25522349 PMCID: PMC4270428 DOI: 10.1371/journal.pcbi.1003943] [Citation(s) in RCA: 11] [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: 06/03/2014] [Accepted: 09/28/2014] [Indexed: 01/04/2023] Open
Abstract
Reconstructing regulatory and signaling response networks is one of the major goals of systems biology. While several successful methods have been suggested for this task, some integrating large and diverse datasets, these methods have so far been applied to reconstruct a single response network at a time, even when studying and modeling related conditions. To improve network reconstruction we developed MT-SDREM, a multi-task learning method which jointly models networks for several related conditions. In MT-SDREM, parameters are jointly constrained across the networks while still allowing for condition-specific pathways and regulation. We formulate the multi-task learning problem and discuss methods for optimizing the joint target function. We applied MT-SDREM to reconstruct dynamic human response networks for three flu strains: H1N1, H5N1 and H3N2. Our multi-task learning method was able to identify known and novel factors and genes, improving upon prior methods that model each condition independently. The MT-SDREM networks were also better at identifying proteins whose removal affects viral load indicating that joint learning can still lead to accurate, condition-specific, networks. Supporting website with MT-SDREM implementation: http://sb.cs.cmu.edu/mtsdrem To understand why some flu strains are more virulent than others, researchers attempt to profile and model the molecular human response to these strains and identify similarities and differences between the resulting models. So far, the modeling and analysis part has been done independently for each strain and the results contrasted in a post-processing step. Here we present a new method, termed MT-SDREM, that simultaneously models the response to all strains allowing us to identify both, the core response elements that are shared among the strains, and factors that are uniquely activated or repressed by individual strains. We applied this method to study the human response to three flu strains: H1N1, H3N2 and H5N1. As we show, the method was able to correctly identify several common and known factors regulating immune response to such strains and also identified unique factors for each of the strains. The models reconstructed by the simultaneous analysis method improved upon those generated by methods that model each strain response separately. Our joint models can be used to identify strain specific treatments as well as treatments that are likely to be effective against all three strains.
Collapse
Affiliation(s)
- Siddhartha Jain
- Computer Science Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Anthony Gitter
- Microsoft Research, Cambridge, Massachusetts, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Ziv Bar-Joseph
- Lane Center for Computational Biology and Machine Learning Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
21
|
Doerr HW, Berger A. Vaccination against infectious diseases: what is promising? Med Microbiol Immunol 2014; 203:365-71. [PMID: 25064610 DOI: 10.1007/s00430-014-0346-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/10/2014] [Indexed: 12/17/2022]
Abstract
Vaccination has proven to be one of the best weapons protecting the mankind against infectious diseases. Along with the huge progress in microbiology, numerous highly efficacious and safe vaccines have been produced by conventional technology (cultivation), by the use of molecular biology (genetic modification), or by synthetic chemistry. Sterilising prevention is achieved by the stimulation of antibody production, while the stimulation of cell-mediated immune responses may prevent the outbreak of disease in consequence of an acute or reactivated infection. From several examples, two rules are deduced to evaluate the perspectives of future vaccine developments: They are promising, if (1) the natural infectious disease induces immunity or (2) passive immunisation (transfer of antibodies, adoptive transfer of lymphocytes) is successful in preventing infection.
Collapse
Affiliation(s)
- Hans Wilhelm Doerr
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University, Paul-Ehrlich-Str. 40, 60596, Frankfurt/M., Germany,
| | | |
Collapse
|
22
|
Heldens J, Hulskotte E, Voeten T, Breedveld B, Verweij P, van Duijnhoven W, Rudenko L, van Damme P, van den Bosch H. Safety and immunogenicity in man of a cell culture derived trivalent live attenuated seasonal influenza vaccine: a Phase I dose escalating study in healthy volunteers. Vaccine 2014; 32:5118-24. [PMID: 24858566 DOI: 10.1016/j.vaccine.2014.05.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 05/06/2014] [Accepted: 05/09/2014] [Indexed: 11/17/2022]
Abstract
Live attenuated influenza vaccine (LAIV) offers the promise of inducing a variety of immune responses thereby conferring protection to circulating field strains. LAIVs are based on cold adapted and temperature sensitive phenotypes of master donor viruses (MDVs) containing the surface glycoprotein genes of seasonal influenza strains. Two types of MDV lineages have been described, the Ann Arbor lineages and the A/Leningrad/17 and B/USSR/60 lineages. Here the safety and immunogenicity of a Madin Darby Canine Kidney - cell culture based, intranasal LAIV derived from A/Leningrad/17 and B/USSR, was evaluated in healthy influenza non-naive volunteers 18-50 years of age. In a double-blind, randomized, placebo-controlled design, single escalating doses of 1×10(5), 1×10(6), or 1×10(7) tissue culture infectious dose 50% (TCID50) of vaccine containing each of the three influenza virus re-assortants recommended by the World Health Organization for the 2008-2009 season were administered intranasally. A statistically significant geometric mean increase in hemagglutination inhibition titer was reached for influenza strain A/H3N2 after immunization with all doses of LAIV. For the A/H1N1 and B strains, the GMI in HI titer did not increase for any of the doses. Virus neutralization antibody titers showed a similar response pattern. A dose-response effect could not be demonstrated for any of the strains, neither for the HI antibody nor for the VN antibody responses. No influenza like symptoms, no nasal congestions, no rhinorrhea, or other influenza related upper respiratory tract symptoms were observed. In addition, no difference in the incidence or nature of adverse events was found between vaccine and placebo treated subjects. Overall, the results indicated that the LAIV for nasal administration is immunogenic (i.e. able to provoke an immune response) and safe both from the perspective of the attenuated virus and the MDCK cell line from which it was derived, and it warrants further development.
Collapse
Affiliation(s)
- Jacco Heldens
- Nobilon International BV, Wim de Körverstraat 35, 5831 AN Boxmeer, The Netherlands.
| | - Ellen Hulskotte
- Merck Sharpe and Dohme Oss BV, Moleneind 110, 5342 CC Oss, The Netherlands
| | - Theo Voeten
- Nobilon International BV, Wim de Körverstraat 35, 5831 AN Boxmeer, The Netherlands
| | - Belinda Breedveld
- Merck Sharpe and Dohme Oss BV, Moleneind 110, 5342 CC Oss, The Netherlands
| | - Pierre Verweij
- Merck Sharpe and Dohme Oss BV, Moleneind 110, 5342 CC Oss, The Netherlands
| | | | - Larissa Rudenko
- Institute of Experimental Medicine, Russian Academy of Medical Sciences, 12 Acad. Pavlov Street, St. Petersburg, 197376, Russia
| | - Pierre van Damme
- Vaccine & Infectious Diseases Institute (VAXINFECTIO), University of Antwerp, Universiteitsplein 1, B2610 Antwerp, Belgium
| | - Han van den Bosch
- Nobilon International BV, Wim de Körverstraat 35, 5831 AN Boxmeer, The Netherlands
| |
Collapse
|
23
|
Cannon G, Callahan MA, Gronemus JQ, Lowy RJ. Early activation of MAP kinases by influenza A virus X-31 in murine macrophage cell lines. PLoS One 2014; 9:e105385. [PMID: 25166426 PMCID: PMC4148262 DOI: 10.1371/journal.pone.0105385] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/23/2014] [Indexed: 12/17/2022] Open
Abstract
Early molecular responses to Influenza A (FLUA) virus strain A/X-31 H3N2 in macrophages were explored using J774.A1 and RAW 264.7 murine cell lines. NF-kappa B (NFκB) was reported to be central to FLUA host-response in other cell types. Our data showed that FLUA activation of the classical NFκB dependent pathway in these macrophages was minimal. Regulator proteins, IkappaB-alpha and -beta (IκBα, IκBβ), showed limited degradation peaking at 2 h post FLUA exposure and p65 was not observed to translocate from the cytoplasm to the nucleus. Additionally, the non-canonical NFκB pathway was not activated in response to FLUA. The cells did display early increases in TNFα and other inflammatory cytokine and chemokine production. Mitogen activated phosphokinase (MAPK) signaling pathways are also reported to control production of inflammatory cytokines in response to FLUA. The activation of the MAPKs, cJun kinases 1 and 2 (JNK 1/2), extracellular regulated kinases 1 and 2 (ERK 1/2), and p38 were investigated in both cell lines between 0.25 and 3 h post-infection. Each of these kinases showed increased phosphorylation post FLUA exposure. JNK phosphorylation occurred early while p38 phosphorylation appeared later. Phosphorylation of ERK 1/2 occurred earlier in J774.A1 cells compared to RAW 264.7 cells. Inhibition of MAPK activation resulted in decreased production of most FLUA responsive cytokines and chemokines in these cells. The results suggest that in these monocytic cells the MAPK pathways are important in the early response to FLUA.
Collapse
Affiliation(s)
- Georgetta Cannon
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Michelle A. Callahan
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Jenny Q. Gronemus
- Central Accessing Unit, American Type Culture Collection, Manassas, Virginia, United States of America
| | - R. Joel Lowy
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
24
|
Michaelis M, Sithisarn P, Cinatl J. Effects of flavonoid-induced oxidative stress on anti-H5N1 influenza a virus activity exerted by baicalein and biochanin A. BMC Res Notes 2014; 7:384. [PMID: 24958200 PMCID: PMC4080993 DOI: 10.1186/1756-0500-7-384] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/13/2014] [Indexed: 12/26/2022] Open
Abstract
Background Different flavonoids are known to interfere with influenza A virus replication. Recently, we showed that the structurally similar flavonoids baicalein and biochanin A inhibit highly pathogenic avian H5N1 influenza A virus replication by different mechanisms in A549 lung cells. Here, we investigated the effects of both compounds on H5N1-induced reactive oxygen species (ROS) formation and the role of ROS formation during H5N1 replication. Findings Baicalein and biochanin A enhanced H5N1-induced ROS formation in A549 cells and primary human monocyte-derived macrophages. Suppression of ROS formation induced by baicalein and biochanin A using the antioxidant N-acetyl-L-cysteine strongly increased the anti-H5N1 activity of both compounds in A549 cells but not in macrophages. Conclusions These findings emphasise that flavonoids induce complex pharmacological actions some of which may interfere with H5N1 replication while others may support H5N1 replication. A more detailed understanding of these actions and the underlying structure-activity relationships is needed to design agents with optimised anti-H5N1 activity.
Collapse
Affiliation(s)
| | | | - Jindrich Cinatl
- Institute for Medical Virology, Clinics of the Goethe-University, Paul Ehrlich-Str, 40, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
25
|
Attenuation of pathogenic immune responses during infection with human and simian immunodeficiency virus (HIV/SIV) by the tetracycline derivative minocycline. PLoS One 2014; 9:e94375. [PMID: 24732038 PMCID: PMC3986096 DOI: 10.1371/journal.pone.0094375] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 03/15/2014] [Indexed: 01/16/2023] Open
Abstract
HIV immune pathogenesis is postulated to involve two major mechanisms: 1) chronic innate immune responses that drive T cell activation and apoptosis and 2) induction of immune regulators that suppress T cell function and proliferation. Both arms are elevated chronically in lymphoid tissues of non-natural hosts, which ultimately develop AIDS. However, these mechanisms are not elevated chronically in natural hosts of SIV infection that avert immune pathogenesis despite similarly high viral loads. In this study we investigated whether minocycline could modulate these pathogenic antiviral responses in non-natural hosts of HIV and SIV. We found that minocycline attenuated in vitro induction of type I interferon (IFN) and the IFN-stimulated genes indoleamine 2,3-dioxygenase (IDO1) and TNF-related apoptosis inducing ligand (TRAIL) in human plasmacytoid dendritic cells and PBMCs exposed to aldrithiol-2 inactivated HIV or infectious influenza virus. Activation-induced TRAIL and expression of cytotoxic T-lymphocyte antigen 4 (CTLA-4) in isolated CD4+ T cells were also reduced by minocycline. Translation of these in vitro findings to in vivo effects, however, were mixed as minocycline significantly reduced markers of activation and activation-induced cell death (CD25, Fas, caspase-3) but did not affect expression of IFNβ or the IFN-stimulated genes IDO1, FasL, or Mx in the spleens of chronically SIV-infected pigtailed macaques. TRAIL expression, reflecting the mixed effects of minocycline on activation and type I IFN stimuli, was reduced by half, but this change was not significant. These results show that minocycline administered after infection may protect against aspects of activation-induced cell death during HIV/SIV immune disease, but that in vitro effects of minocycline on type I IFN responses are not recapitulated in a rapid progressor model in vivo.
Collapse
|
26
|
Lv J, Hua Y, Wang D, Liu A, An J, Li A, Wang Y, Wang X, Jia N, Jiang Q. Kinetics of pulmonary immune cells, antibody responses and their correlations with the viral clearance of influenza A fatal infection in mice. Virol J 2014; 11:57. [PMID: 24666970 PMCID: PMC3986938 DOI: 10.1186/1743-422x-11-57] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/13/2014] [Indexed: 12/03/2022] Open
Abstract
Fatal influenza A virus infection is a major threat to public health throughout the world. Lung macrophages and neutrophils have critical roles for both the pathogenesis and viral clearance of fatal viral infections. These are complicated by the interaction of innate immunity and adaptive immunity against viral infection. In this study, we investigated the overall kinetics of lung macrophages, neutrophils, CD4+T cells, CD8+T cells, CD38+ cells, and CD138+ cells, the levels of antibody and cytokine responses, both in the early and late phases of fatal infection with A/PR/8/34 (H1N1) virus in mice. The changes in lung viral load were also evaluated. We found that pulmonary macrophages and neutrophils both accumulated in the early and late phases of fatal infections and they positively correlated with the lung and serum antibody titers, and negatively correlated with the viral load locally. The secretion of IL-6 might relate to high numbers of macrophages and neutrophils in the early infection. The work implies that pulmonary macrophages, neutrophils and the antibody response all have an essential role in virus elimination of fatal influenza A viral infection. These findings may have implications for the development of prophylactic and therapeutic strategies in fatal influenza A viral infection. Further evaluation of the cooperation among macrophages, neutrophils and antibody responses in eliminating the virus with fatal infection is needed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiliang Wang
- The Second Artillery General Hospital, PLA, 16 Xinjiekouwai Street, Xicheng District Beijing 100088, China.
| | | | | |
Collapse
|
27
|
Abstract
Influenza has been recognized as a respiratory disease in swine since its first appearance concurrent with the 1918 "Spanish flu" human pandemic. All influenza viruses of significance in swine are type A, subtype H1N1, H1N2, or H3N2 viruses. Influenza viruses infect epithelial cells lining the surface of the respiratory tract, inducing prominent necrotizing bronchitis and bronchiolitis and variable interstitial pneumonia. Cell death is due to direct virus infection and to insult directed by leukocytes and cytokines of the innate immune system. The most virulent viruses consistently express the following characteristics of infection: (1) higher or more prolonged virus replication, (2) excessive cytokine induction, and (3) replication in the lower respiratory tract. Nearly all the viral proteins contribute to virulence. Pigs are susceptible to infection with both human and avian viruses, which often results in gene reassortment between these viruses and endemic swine viruses. The receptors on the epithelial cells lining the respiratory tract are major determinants of infection by influenza viruses from other hosts. The polymerases, especially PB2, also influence cross-species infection. Methods of diagnosis and characterization of influenza viruses that infect swine have improved over the years, driven both by the availability of new technologies and by the necessity of keeping up with changes in the virus. Testing of oral fluids from pigs for virus and antibody is a recent development that allows efficient sampling of large numbers of animals.
Collapse
Affiliation(s)
- B H Janke
- DVM, PhD, Veterinary Diagnostic Laboratory, Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
28
|
Yang SQ, Qu JX, Wang C, Yu XM, Liu YM, Cao B. Influenza pneumonia among adolescents and adults: a concurrent comparison between influenza A (H1N1) pdm09 and A (H3N2) in the post-pandemic period. CLINICAL RESPIRATORY JOURNAL 2013; 8:185-91. [PMID: 24106842 PMCID: PMC4204512 DOI: 10.1111/crj.12056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 06/29/2013] [Accepted: 09/10/2013] [Indexed: 12/25/2022]
Abstract
Introduction Comparisons of the characteristics between the influenza A (H1N1) pdm09 and common seasonal influenza are important for both clinical management and epidemiological studies. However, the differences between pandemic and seasonal influenza during the post-pandemic period are poorly understood. Objectives The aim of our research was to investigate clinical and immune response differences between patients with influenza A (H1N1) pdm09 pneumonia and seasonal influenza A (H3N2) pneumonia in the post-pandemic period. Methods During the first flu season in post-pandemic period, patients from Beijing Network for Adult Community-Acquired Pneumonia present A (H1N1) pdm09 or A (H3N2) influenza were compared concurrently in the aspects of clinical characteristics and inflammatory profile in acute phase. Result Patients with A (H1N1) pdm09 influenza pneumonia showed a close mean age to A (H3N2) pneumonia (51 ± 20 vs 53 ± 16, mean ± standard deviation, years) but tended to have more underlying diseases (32.8% vs 10%, P = 0.036). Although clinical characteristics were similar, no statistical difference were found in pneumonia severity index (PSI) score or intensive care unit admission rate or mortality, patients in A (H1N1) pdm09 cohort present higher levels of aspartate aminotransferase, lactase dehydrogenase (P = 0.006, 0.018, respectively) in blood and also longer duration of fever than A (H3N2) cohort. Levels of interleukin (IL)-10 and IL-12 (p70) were higher in A (H1N1) pdm09 cohort (P = 0.031, 0.047, respectively). Conclusios During the first post-pandemic flu season, patients with the A (H1N1) pdm09 pneumonia showed similar clinical characteristics but slightly higher disease severity and stronger systemic inflammatory response than A (H3N2) pneumonia.
Collapse
Affiliation(s)
- Shu Qiao Yang
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chao-Yang Hospital, Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
29
|
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: 407] [Impact Index Per Article: 33.9] [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
|
30
|
Börgeling Y, Schmolke M, Viemann D, Nordhoff C, Roth J, Ludwig S. Inhibition of p38 mitogen-activated protein kinase impairs influenza virus-induced primary and secondary host gene responses and protects mice from lethal H5N1 infection. J Biol Chem 2013; 289:13-27. [PMID: 24189062 PMCID: PMC3879537 DOI: 10.1074/jbc.m113.469239] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Highly pathogenic avian influenza viruses (HPAIV) induce severe inflammation in poultry and men. One characteristic of HPAIV infections is the induction of a cytokine burst that strongly contributes to viral pathogenicity. This cell-intrinsic hypercytokinemia seems to involve hyperinduction of p38 mitogen-activated protein kinase. Here we investigate the role of p38 MAPK signaling in the antiviral response against HPAIV in mice as well as in human endothelial cells, the latter being a primary source of cytokines during systemic infections. Global gene expression profiling of HPAIV-infected endothelial cells in the presence of the p38-specific inhibitor SB 202190 revealed that inhibition of p38 MAPK leads to reduced expression of IFNβ and other cytokines after H5N1 and H7N7 infection. More than 90% of all virus-induced genes were either partially or fully dependent on p38 signaling. Moreover, promoter analysis confirmed a direct impact of p38 on the IFNβ promoter activity. Furthermore, upon treatment with IFN or conditioned media from HPAIV-infected cells, p38 controls interferon-stimulated gene expression by coregulating STAT1 by phosphorylation at serine 727. In vivo inhibition of p38 MAPK greatly diminishes virus-induced cytokine expression concomitant with reduced viral titers, thereby protecting mice from lethal infection. These observations show that p38 MAPK acts on two levels of the antiviral IFN response. Initially the kinase regulates IFN induction and, at a later stage, p38 controls IFN signaling and thereby expression of IFN-stimulated genes. Thus, inhibition of MAP kinase p38 may be an antiviral strategy that protects mice from lethal influenza by suppressing excessive cytokine expression.
Collapse
Affiliation(s)
- Yvonne Börgeling
- From the Institute of Molecular Virology, Center for Molecular Biology of Inflammation
| | | | | | | | | | | |
Collapse
|
31
|
Matsuoka Y, Matsumae H, Katoh M, Eisfeld AJ, Neumann G, Hase T, Ghosh S, Shoemaker JE, Lopes TJS, Watanabe T, Watanabe S, Fukuyama S, Kitano H, Kawaoka Y. A comprehensive map of the influenza A virus replication cycle. BMC SYSTEMS BIOLOGY 2013; 7:97. [PMID: 24088197 PMCID: PMC3819658 DOI: 10.1186/1752-0509-7-97] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/24/2013] [Indexed: 02/05/2023]
Abstract
Background Influenza is a common infectious disease caused by influenza viruses. Annual epidemics cause severe illnesses, deaths, and economic loss around the world. To better defend against influenza viral infection, it is essential to understand its mechanisms and associated host responses. Many studies have been conducted to elucidate these mechanisms, however, the overall picture remains incompletely understood. A systematic understanding of influenza viral infection in host cells is needed to facilitate the identification of influential host response mechanisms and potential drug targets. Description We constructed a comprehensive map of the influenza A virus (‘IAV’) life cycle (‘FluMap’) by undertaking a literature-based, manual curation approach. Based on information obtained from publicly available pathway databases, updated with literature-based information and input from expert virologists and immunologists, FluMap is currently composed of 960 factors (i.e., proteins, mRNAs etc.) and 456 reactions, and is annotated with ~500 papers and curation comments. In addition to detailing the type of molecular interactions, isolate/strain specific data are also available. The FluMap was built with the pathway editor CellDesigner in standard SBML (Systems Biology Markup Language) format and visualized as an SBGN (Systems Biology Graphical Notation) diagram. It is also available as a web service (online map) based on the iPathways+ system to enable community discussion by influenza researchers. We also demonstrate computational network analyses to identify targets using the FluMap. Conclusion The FluMap is a comprehensive pathway map that can serve as a graphically presented knowledge-base and as a platform to analyze functional interactions between IAV and host factors. Publicly available webtools will allow continuous updating to ensure the most reliable representation of the host-virus interaction network. The FluMap is available at http://www.influenza-x.org/flumap/.
Collapse
Affiliation(s)
- Yukiko Matsuoka
- JST ERATO Kawaoka infection-induced host responses project, Minato-ku, Tokyo 108-8639, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bordon JM, Uriarte S, Arnold FW, Fernandez-Botran R, Rane M, Peyrani P, Cavallazzi R, Saad M, Ramirez J. Cytokines and neutrophils responses in influenza pneumonia. Infection 2013; 41:1021-4. [PMID: 23589279 DOI: 10.1007/s15010-013-0461-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 04/04/2013] [Indexed: 01/26/2023]
Abstract
This case report shows a striking correlation of remarkable brief high levels of pro- and anti-inflammatory cytokines coupled with increased neutrophil activation, followed by a sharp decrease in cytokine levels and increased neutrophil apoptosis associated with the favorable clinical outcomes of a patient with severe influenza infection. The host response examined in our case is not complete, given it did not assess the full spectrum of host response. The brief neutrophil and cytokine response seen in our case in the absence of antiviral therapy and in the presence of methotrexate immunosuppressive therapy rise the question as to whether the latter optimally modulated the macrophage function, resulting in a favorable outcome of severe influenza viral infection.
Collapse
Affiliation(s)
- J M Bordon
- Section of Infectious Diseases, Providence Hospital, 1150 Varnum Street, NE, Washington, DC, 20017, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Michael P, Brabant D, Bleiblo F, Ramana CV, Rutherford M, Khurana S, Tai T, Kumar A, Kumar A. Influenza A induced cellular signal transduction pathways. J Thorac Dis 2013; 5 Suppl 2:S132-41. [PMID: 23977434 PMCID: PMC3747532 DOI: 10.3978/j.issn.2072-1439.2013.07.42] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 07/25/2013] [Indexed: 12/28/2022]
Abstract
Influenza A is a negative sense single stranded RNA virus that belongs to the Orthomyxoviridae Family. This enveloped virus contains 8 segments of viral RNA which encodes 11 viral proteins. Influenza A infects humans and is the causative agent of the flu. Annually it infects approximately 5% to 15% of the population world wide and results in an estimated 250,000 to 500,000 deaths a year. The nature of influenza A replication results in a high mutation rate which results in the need for seasonal vaccinations. In addition the zoonotic nature of the influenza virus allows for recombination of viral segments from different strains creating new variants that have not been encountered before. This type of mutation is the method by which pandemic strains of the flu arises. Infection with influenza results in a respiratory illness that for most individuals is self limiting. However in susceptible populations which include individuals with pre-existing pulmonary or cardiac conditions, the very young and the elderly fatal complications may arise. The most serious of these is the development of viral pneumonia which may be accompanied by secondary bacterial infections. Progression of pneumonia leads to the development of acute respiratory distress syndrome (ARDS), acute lung injury (ALI) and potentially respiratory failure. This progression is a combined effect of the host immune system response to influenza infection and the viral infection itself. This review will focus on molecular aspects of viral replication in alveolar cells and their response to infection. The response of select innate immune cells and their contribution to viral clearance and lung epithelial damage will also be discussed. Molecular aspects of antiviral response in the cells in particular the protein kinase RNA dependent response, and the oligoadenylate synthetase RNAse L system in relation to influenza infection.
Collapse
Affiliation(s)
- Paul Michael
- Department of Chemistry and Biochemistry and the Biomolecular Sciences Program, Laurentian University, Sudbury, P3E 2C6, ON, Canada
| | - Danielle Brabant
- Department of Chemistry and Biochemistry and the Biomolecular Sciences Program, Laurentian University, Sudbury, P3E 2C6, ON, Canada
| | - Farag Bleiblo
- Department of Chemistry and Biochemistry and the Biomolecular Sciences Program, Laurentian University, Sudbury, P3E 2C6, ON, Canada
- Department of Biology, University of Benghazi, Benghazi, Libya
| | | | - Michael Rutherford
- Department of Pathology, Health Sciences North, Sudbury, P3E 5J1, ON, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, P3E 2C6, ON, Canada
| | - Sandhya Khurana
- Department of Chemistry and Biochemistry and the Biomolecular Sciences Program, Laurentian University, Sudbury, P3E 2C6, ON, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, P3E 2C6, ON, Canada
| | - T.C. Tai
- Department of Chemistry and Biochemistry and the Biomolecular Sciences Program, Laurentian University, Sudbury, P3E 2C6, ON, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, P3E 2C6, ON, Canada
| | - Anand Kumar
- Section of Critical Care Medicine, University of Manitoba, Winnipeg, R3A 1R9, MB, Canada
| | - Aseem Kumar
- Department of Chemistry and Biochemistry and the Biomolecular Sciences Program, Laurentian University, Sudbury, P3E 2C6, ON, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, P3E 2C6, ON, Canada
| |
Collapse
|
34
|
Chan RWY, Chan MCW, Nicholls JM, Malik Peiris JS. Use of ex vivo and in vitro cultures of the human respiratory tract to study the tropism and host responses of highly pathogenic avian influenza A (H5N1) and other influenza viruses. Virus Res 2013; 178:133-45. [PMID: 23684848 DOI: 10.1016/j.virusres.2013.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 02/24/2013] [Accepted: 03/05/2013] [Indexed: 12/27/2022]
Abstract
The tropism of influenza viruses for the human respiratory tract is a key determinant of host-range, and consequently, of pathogenesis and transmission. Insights can be obtained from clinical and autopsy studies of human disease and relevant animal models. Ex vivo cultures of the human respiratory tract and in vitro cultures of primary human cells can provide complementary information provided they are physiologically comparable in relevant characteristics to human tissues in vivo, e.g. virus receptor distribution, state of differentiation. We review different experimental models for their physiological relevance and summarize available data using these cultures in relation to highly pathogenic avian influenza H5N1, in comparison where relevant, with other influenza viruses. Transformed continuous cell-lines often differ in important ways to the corresponding tissues in vivo. The state of differentiation of primary human cells (respiratory epithelium, macrophages) can markedly affect virus tropism and host responses. Ex vivo cultures of human respiratory tissues provide a close resemblance to tissues in vivo and may be used to risk assess animal viruses for pandemic threat. Physiological factors (age, inflammation) can markedly affect virus receptor expression and virus tropism. Taken together with data from clinical studies on infected humans and relevant animal models, data from ex vivo and in vitro cultures of human tissues and cells can provide insights into virus transmission and pathogenesis and may provide understanding that leads to novel therapeutic interventions.
Collapse
Affiliation(s)
- Renee W Y Chan
- Department of Pathology, The University of Hong Kong, Hong Kong Special Administrative Region; Centre of Influenza Research, The University of Hong Kong, Hong Kong Special Administrative Region.
| | | | | | | |
Collapse
|
35
|
Abstract
Avian influenza virus infections in the human population are rare due to their inefficient direct human-to-human transmission. However, when humans are infected, a strong inflammatory response is usually induced, characterized by elevated levels of cytokines and chemokines in serum, believed to be important in the severe pathogenesis that develops in a high proportion of these patients. Extensive research has been performed to understand the molecular viral mechanisms involved in the H5N1 pathogenesis in humans, providing interesting insights about the virus-host interaction and the regulation of the innate immune response by these highly pathogenic viruses. In this review we summarize and discuss the most important findings in this field, focusing mainly on H5N1 virulence factors and their impact on the modulation of the innate immunity in humans.
Collapse
Affiliation(s)
- Irene Ramos
- Authors to whom correspondence should be addressed; (A.F.S.); (I.R.); Tel. +1-212-241-5182 (A.F.S.); +1-212 241-0994 (I.R.); Fax: +1-212-534-1684 (A.F.S.); +1-212-534-1684 (I.R.)
| | - Ana Fernandez-Sesma
- Authors to whom correspondence should be addressed; (A.F.S.); (I.R.); Tel. +1-212-241-5182 (A.F.S.); +1-212 241-0994 (I.R.); Fax: +1-212-534-1684 (A.F.S.); +1-212-534-1684 (I.R.)
| |
Collapse
|
36
|
Matthaei M, Budt M, Wolff T. Highly pathogenic H5N1 influenza A virus strains provoke heterogeneous IFN-α/β responses that distinctively affect viral propagation in human cells. PLoS One 2013; 8:e56659. [PMID: 23451066 PMCID: PMC3581526 DOI: 10.1371/journal.pone.0056659] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 01/14/2013] [Indexed: 12/24/2022] Open
Abstract
The fatal transmissions of highly pathogenic avian influenza A viruses (IAV) of the H5N1 subtype to humans and high titer replication in the respiratory tract indicate that these pathogens can overcome the bird-to-human species barrier. While type I interferons (IFN-α/β) are well described to contribute to the species barrier of many zoonotic viruses, current data to the role of these antiviral cytokines during human H5N1 IAV infections is limited and contradictory. We hypothesized an important role for the IFN system in limiting productive infection of avian H5N1 strains in human cells. Hence, we examined IFN-α/β gene activation by different avian and human H5N1 isolates, if the IFN-α/β response restricts H5N1 growth and whether the different strains were equally capable to regulate the IFN-α/β system via their IFN-antagonistic NS1 proteins. Two human H5N1 isolates and a seasonal H3N2 strain propagated efficiently in human respiratory cells and induced little IFN-β, whereas three purely avian H5N1 strains were attenuated for replication and provoked higher IFN secretion. Replication of avian viruses was significantly enhanced on interferon-deficient cells, and exogenous IFN potently limited the growth of all strains in human cells. Moreover, IFN-α/β activation by all strains depended on retinoic acid-inducible gene I excluding principal differences in receptor activation between the different viruses. Interestingly, all H5N1 NS1 proteins suppressed IFN-α/β induction comparably well to the NS1 of seasonal IAV. Thus, our study shows that H5N1 strains are heterogeneous in their capacity to activate human cells in an NS1-independent manner. Our findings also suggest that H5N1 viruses need to acquire adaptive changes to circumvent strong IFN-α/β activation in human host cells. Since no single amino acid polymorphism could be associated with a respective high- or low induction phenotype we propose that the necessary adaptations to overcome the human IFN-α/β barrier involve mutations in multiple H5N1 genes.
Collapse
Affiliation(s)
- Markus Matthaei
- Division of Influenza/Respiratory Viruses, Robert Koch-Institut, Berlin, Germany
| | - Matthias Budt
- Division of Influenza/Respiratory Viruses, Robert Koch-Institut, Berlin, Germany
| | - Thorsten Wolff
- Division of Influenza/Respiratory Viruses, Robert Koch-Institut, Berlin, Germany
- * E-mail:
| |
Collapse
|
37
|
Sithisarn P, Michaelis M, Schubert-Zsilavecz M, Cinatl J. Differential antiviral and anti-inflammatory mechanisms of the flavonoids biochanin A and baicalein in H5N1 influenza A virus-infected cells. Antiviral Res 2012; 97:41-8. [PMID: 23098745 DOI: 10.1016/j.antiviral.2012.10.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 10/04/2012] [Accepted: 10/15/2012] [Indexed: 12/22/2022]
Abstract
From a panel of 22 flavonoids, we identified six compounds (apigenin, baicalein, biochanin A, kaempferol, luteolin, naringenin) that inhibited influenza A nucleoprotein production in human lung epithelial (A549) cells infected with the highly pathogenic avian influenza H5N1 virus strain A/Thailand/Kan-1/04 in non-toxic concentrations. Baicalein (IC(50): 18.79±1.17μM, selectivity index 5.82) and biochanin A (IC(50) 8.92±1.87μM, selectivity index 5.60) were selected for further experiments. Both compounds reduced H5N1 infectious titres (baicalein 40μM: 29-fold reduction, biochanin A 40μM: 55-fold reduction after infection at MOI 0.01), virus-induced caspase 3 cleavage, nuclear export of viral RNP complexes, and enhanced the effects of the neuraminidase inhibitor zanamivir. Biochanin A and baicalein also inhibited the replication of the H5N1 strain A/Vietnam/1203/04. Time of addition experiments indicated that both compounds interfere with H5N1 replication after the adsorption period. Further mechanistic investigations revealed clear differences between these two flavonoids. Only baicalein interfered with the viral neuraminidase activity (39±7% inhibition at 100μM, the maximum concentration tested). In contrast to baicalein, biochanin A affected cellular signalling pathways resulting in reduced virus-induced activation of AKT, ERK 1/2, and NF-kB. Moreover, biochanin A inhibited the virus-induced production of IL-6, IL-8, and IP-10 while baicalein inhibited IL-6 and IL-8 production without affecting IP-10 levels. In primary human monocyte-derived macrophages, only baicalein but not biochanin A impaired H5N1 virus replication. Both flavonoids interfered with the H5N1-induced production of IL-6, IP-10, and TNF-α but not of IL-8 in macrophages. These findings indicate that closely related flavonoids can exert anti-H5N1 effects by different molecular mechanisms.
Collapse
Affiliation(s)
- Patchima Sithisarn
- Institute for Medical Virology, Clinics of the Goethe-University, Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
38
|
Liu KC, Fang JM, Jan JT, Cheng TJR, Wang SY, Yang ST, Cheng YSE, Wong CH. Enhanced anti-influenza agents conjugated with anti-inflammatory activity. J Med Chem 2012; 55:8493-501. [PMID: 22963087 DOI: 10.1021/jm3009844] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Influenza therapy with a single targeted compound is often limited in efficacy due to the rapidly developed drug resistance. Moreover, the uncontrolled virus-induced cytokines could cause the high mortality of human infected by H5N1 avian influenza virus. In this study, we explored the novel dual-targeted bifunctional anti-influenza drugs formed by conjugation with anti-inflammatory agents. In particular, the caffeic acid (CA)-bearing zanamivir (ZA) conjugates ZA-7-CA (1) and ZA-7-CA-amide (7) showed simultaneous inhibition of influenza virus neuraminidase and suppression of pro-inflammatory cytokines. These ZA conjugates provided remarkable protection of cells and mice against influenza infections. Intranasal administration of low dosage (<1.2 μmol/kg/day) of ZA conjugates exhibited much greater effect than the combination therapy with ZA and the anti-inflammatory agents in protection of the lethally infected mice by H1N1 or H5N1 influenza viruses.
Collapse
Affiliation(s)
- Kung-Cheng Liu
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Lee SMY, Dutry I, Peiris JSM. Editorial: macrophage heterogeneity and responses to influenza virus infection. J Leukoc Biol 2012; 92:1-4. [PMID: 22745457 DOI: 10.1189/jlb.0312130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
40
|
Doerr HW, Cinatl J. Recent publications in medical microbiology and immunology: a retrospective. Med Microbiol Immunol 2012; 201:1-5. [PMID: 22033658 DOI: 10.1007/s00430-011-0219-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Indexed: 01/15/2023]
Abstract
A look back is done to some clinical and basic research activities recently published in medical microbiology and immunology. The review covers clinical experiences and in vitro experiments to understand the emergency, pathogenicity, epidemic spread, and vaccine-based prevention of avian and swine-origin flu. Some new developments and concepts in diagnosis, (molecular) epidemiology, and therapy of AIDS, viral hepatitis C, and herpesvirus-associated diseases are outlined. Regulation of immune system has been discussed in a special issue 2010 including some aspects of CNS affections (measles). Mycobacterial infection and its prevention by modern recombinant vaccines have reached new interest, as well as new concepts of vaccination and prophylaxis against several other bacteria. Adaptation to host niches enables immune escape (example brucella) and determines virulence (example N. meningitidis). Chlamydia pneumoniae, previously considered to trigger atherosclerosis, is hypothetically associated to Alzheimer disease, while CMV, another putative trigger of atherosclerosis, gains evidence of oncomodulation in CNS tumor diseases. In terms of globalization, exotic virus infections are increasingly imported from southern countries.
Collapse
Affiliation(s)
- H W Doerr
- Institute of Medical Virology, University Hospital of Frankfurt/M., Frankfurt/Main, Germany.
| | | |
Collapse
|
41
|
Lam WY, Yeung ACM, Chan PKS. Apoptosis, cytokine and chemokine induction by non-structural 1 (NS1) proteins encoded by different influenza subtypes. Virol J 2011; 8:554. [PMID: 22185562 PMCID: PMC3274490 DOI: 10.1186/1743-422x-8-554] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 12/21/2011] [Indexed: 12/23/2022] Open
Abstract
Background Influenza pandemic remains a serious threat to human health. Viruses of avian origin, H5N1, H7N7 and H9N2, have repeatedly crossed the species barrier to infect humans. Recently, a novel strain originated from swine has evolved to a pandemic. This study aims at improving our understanding on the pathogenic mechanism of influenza viruses, in particular the role of non-structural (NS1) protein in inducing pro-inflammatory and apoptotic responses. Methods Human lung epithelial cells (NCI-H292) was used as an in-vitro model to study cytokine/chemokine production and apoptosis induced by transfection of NS1 mRNA encoded by seven infleunza subtypes (seasonal and pandemic H1, H2, H3, H5, H7, and H9), respectively. Results The results showed that CXCL-10/IP10 was most prominently induced (> 1000 folds) and IL-6 was slightly induced (< 10 folds) by all subtypes. A subtype-dependent pattern was observed for CCL-2/MCP-1, CCL3/MIP-1α, CCL-5/RANTES and CXCL-9/MIG; where induction by H5N1 was much higher than all other subtypes examined. All subtypes induced a similar temporal profile of apoptosis following transfection. The level of apoptosis induced by H5N1 was remarkably higher than all others. The cytokine/chemokine and apoptosis inducing ability of the 2009 pandemic H1N1 was similar to previous seasonal strains. Conclusions In conclusion, the NS1 protein encoded by H5N1 carries a remarkably different property as compared to other avian and human subtypes, and is one of the keys to its high pathogenicity. NCI-H292 cells system proves to be a good in-vitro model to delineate the property of NS1 proteins.
Collapse
Affiliation(s)
- W Y Lam
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong Special Administration Region, People's Republic of China
| | | | | |
Collapse
|
42
|
Ling MT, Tu W, Han Y, Mao H, Chong WP, Guan J, Liu M, Lam KT, Law HKW, Peiris JSM, Takahashi K, Lau YL. Mannose-binding lectin contributes to deleterious inflammatory response in pandemic H1N1 and avian H9N2 infection. J Infect Dis 2011; 205:44-53. [PMID: 22080095 PMCID: PMC3242741 DOI: 10.1093/infdis/jir691] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mannose-binding lectin (MBL) is a pattern-recognition molecule, which functions as a first line of host defense. Pandemic H1N1 (pdmH1N1) influenza A virus caused massive infection in 2009 and currently circulates worldwide. Avian influenza A H9N2 (H9N2/G1) virus has infected humans and has the potential to be the next pandemic virus. Antiviral function and immunomodulatory role of MBL in pdmH1N1 and H9N2/G1 virus infection have not been investigated. METHODS In this study, MBL wild-type (WT) and MBL knockout (KO) murine models were used to examine the role of MBL in pdmH1N1 and H9N2/G1 virus infection. RESULTS Our study demonstrated that in vitro, MBL binds to pdmH1N1 and H9N2/G1 viruses, likely via the carbohydrate recognition domain of MBL. Wild-type mice developed more severe disease, as evidenced by a greater weight loss than MBL KO mice during influenza virus infection. Furthermore, MBL WT mice had enhanced production of proinflammatory cytokines and chemokines compared with MBL KO mice, suggesting that MBL could upregulate inflammatory responses that may potentially worsen pdmH1N1 and H9N2/G1 virus infections. CONCLUSIONS Our study provided the first in vivo evidence that MBL may be a risk factor during pdmH1N1 and H9N2/G1 infection by upregulating proinflammatory response.
Collapse
Affiliation(s)
- Man To Ling
- Department of Paediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lee N, Wong CK, Chan PKS, Chan MCW, Wong RYK, Lun SWM, Ngai KLK, Lui GCY, Wong BCK, Lee SKW, Choi KW, Hui DSC. Cytokine response patterns in severe pandemic 2009 H1N1 and seasonal influenza among hospitalized adults. PLoS One 2011; 6:e26050. [PMID: 22022504 PMCID: PMC3192778 DOI: 10.1371/journal.pone.0026050] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 09/16/2011] [Indexed: 12/16/2022] Open
Abstract
Background Studying cytokine/chemokine responses in severe influenza infections caused by different virus subtypes may improve understanding on pathogenesis. Methods Adults hospitalized for laboratory-confirmed seasonal and pandemic 2009 A/H1N1 (pH1N1) influenza were studied. Plasma concentrations of 13 cytokines/chemokines were measured at presentation and then serially, using cytometric-bead-array with flow-cytometry and ELISA. PBMCs from influenza patients were studied for cytokine/chemokine expression using ex-vivo culture (Whole Blood Assay,±PHA/LPS stimulation). Clinical variables were prospectively recorded and analyzed. Results 63 pH1N1 and 53 seasonal influenza patients were studied. pH1N1 patients were younger (mean±S.D. 42.8±19.2 vs 70.5±16.7 years), and fewer had comorbidities. Respiratory/cardiovascular complications were common in both groups (71.4% vs 81.1%), although severe pneumonia with hypoxemia (54.0% vs 28.3%) and ICU admissions (25.4% vs 1.9%) were more frequent with pH1N1. Hyperactivation of the proinflammatory cytokines IL-6, CXCL8/IL-8, CCL2/MCP-1 and sTNFR-1 was found in pH1N1 pneumonia (2–15 times normal) and in complicated seasonal influenza, but not in milder pH1N1 infections. The adaptive-immunity (Th1/Th17)-related CXCL10/IP-10, CXCL9/MIG and IL-17A however, were markedly suppressed in severe pH1N1 pneumonia (2–27 times lower than seasonal influenza; P−values<0.01). This pattern was further confirmed with serial measurements. Hypercytokinemia tended to be sustained in pH1N1 pneumonia, associated with a slower viral clearance [PCR-negativity: day 3–4, 55% vs 85%; day 6–7, 67% vs 100%]. Elevated proinflammatory cytokines, particularly IL-6, predicted ICU admission (adjusted OR 12.6, 95%CI 2.6–61.5, per log10unit increase; P = 0.002), and correlated with fever, tachypnoea, deoxygenation, and length-of-stay (Spearman's rho, P-values<0.01) in influenza infections. PBMCs in seasonal influenza patients were activated and expressed cytokines ex vivo (e.g. IL-6, CXCL8/IL-8, CCL2/MCP-1, CXCL10/IP-10, CXCL9/MIG); their ‘responsiveness’ to stimuli was shown to change dynamically during the illness course. Conclusions A hyperactivated proinflammatory, but suppressed adaptive-immunity (Th1/Th17)-related cytokine response pattern was found in severe pH1N1 pneumonia, different from seasonal influenza. Cytokine/immune-dysregulation may be important in its pathogenesis.
Collapse
Affiliation(s)
- Nelson Lee
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Oslund KL, Baumgarth N. Influenza-induced innate immunity: regulators of viral replication, respiratory tract pathology & adaptive immunity. Future Virol 2011; 6:951-962. [PMID: 21909336 DOI: 10.2217/fvl.11.63] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Influenza virus infections usually cause mild to moderately severe respiratory disease, however some infections, like those involving the avian H5N1 virus, can cause massive viral pneumonia, systemic disease and death. The innate immune response of respiratory tract resident cells is the first line of defense and limits virus replication. Enhanced cytokine and chemokine production following infection, however, appears to underlie much of the pathology that develops after infection with highly pathogenic strains. A so-called `cytokine storm' can damage the lung tissue and cause systemic disease, despite the control of viral replication. By summarizing current knowledge of the innate responses mounted to influenza infection, this review highlights the importance of the respiratory tract epithelial cells as regulators of innate and adaptive immunity to influenza virus.
Collapse
Affiliation(s)
- Karen L Oslund
- Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | | |
Collapse
|
45
|
Wiesener N, Zimmer C, Jarasch-Althof N, Wutzler P, Henke A. Therapy of experimental influenza virus infection with pyrrolidine dithiocarbamate. Med Microbiol Immunol 2010; 200:115-26. [PMID: 21174118 DOI: 10.1007/s00430-010-0182-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Indexed: 12/27/2022]
Abstract
The search for new antiviral strategies to treat influenza A virus (IAV) infections is one major international health care activity. Hereby, the IAV-caused misuse of cellular nuclear factor kappa B (NF-κB) signaling pathways in infected cells represents one target for antiviral therapy. In the present study, pyrrolidine dithiocarbamate (PDTC), which is known as an antioxidant and as an inhibitor of IAV-induced NF-κB activation, was studied in vivo. After the antiviral activity of PDTC was confirmed in MDCK cells, mice-infected with the mouse-adapted strain of IAV A/PR/8/34 (H1N1)-were treated intraperitoneally simultaneously with PDTC (75, 150, 200 mg/kg body weight). The influence of PDTC administrations was evaluated on viral replication and inflammatory reactions in lung tissue up to 14 days postinfection (p. i.). This therapy increased survival up to 80% and reduced IAV-caused weight loss and viral replication in lung tissue in a dose-dependent manner. Protective effects were less pronounced, if the therapy started later on during an ongoing IAV infection. In addition, simultaneous PDTC treatment also limited IAV-caused infiltration of immune cells as well as local interferon-γ expression in lung tissue. These results imply that PDTC decreases IAV-caused disease in mice significantly. Therefore, the development of drugs like PDTC that interfere with NF-κB signaling may represent a modern focus of anti-IAV therapy.
Collapse
Affiliation(s)
- Nadine Wiesener
- Department of Virology and Antiviral Therapy, Jena University Hospital, Friedrich Schiller University, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | | | | | | | | |
Collapse
|