1
|
Williams SL, Qi L, Sheng ZM, Xiao Y, Freeman A, Matthews L, Legaspi SF, Fodor E, Taubenberger JK. Effect of pandemic influenza A virus PB1 genes of avian origin on viral RNA polymerase activity and pathogenicity. SCIENCE ADVANCES 2024; 10:eads5735. [PMID: 39671482 PMCID: PMC11641000 DOI: 10.1126/sciadv.ads5735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Zoonotic influenza A virus (IAV) infections pose a substantial threat to global health. The influenza RNA-dependent RNA polymerase (RdRp) comprises the PB2, PB1, and PA proteins. Of the last four pandemic IAVs, three featured avian-origin PB1 genes. Prior research linked these avian PB1 genes to increased viral fitness when reassorted with human IAV genes. This study evaluated chimeric RdRps with PB1 genes from the 1918, 1957, and 1968 pandemic IAVs in a low pathogenic avian influenza (LPAI) virus background to assess polymerase activity and pathogenicity. Substituting in the pandemic PB1 genes reduced polymerase activity, virulence, and altered lung pathology, while the native LPAI PB1 showed the highest pathogenicity and polymerase activity. The native LPAI PB1 virus caused severe pneumonia and high early viral RNA levels, correlating with elevated host cytokine signaling. Increased genetic distance from the LPAI PB1 sequence correlated with reduced polymerase activity, IFN-β expression, viral replication, and pathogenicity.
Collapse
Affiliation(s)
- Stephanie L. Williams
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Li Qi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Ashley Freeman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Lex Matthews
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Sharon Fong Legaspi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Ervin Fodor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| |
Collapse
|
2
|
Narciso AR, Dookie R, Nannapaneni P, Normark S, Henriques-Normark B. Streptococcus pneumoniae epidemiology, pathogenesis and control. Nat Rev Microbiol 2024:10.1038/s41579-024-01116-z. [PMID: 39506137 DOI: 10.1038/s41579-024-01116-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Infections caused by Streptococcus pneumoniae (also known as pneumococci) pose a threat to human health. Pneumococcal infections are the most common cause of milder respiratory tract infections, such as otitis and sinusitis, and of more severe diseases, including pneumonia (with or without septicaemia) and meningitis. The introduction of pneumococcal conjugate vaccines in the childhood vaccination programme in many countries has led to a notable decrease of severe invasive pneumococcal disease in vaccinated children. However, infections caused by non-vaccine types have concurrently increased, causing invasive pneumococcal disease in unvaccinated populations (such as older adults), which has hampered the effect of these vaccines. Moreover, emerging antibiotic resistance is threatening effective therapy. Thus, new approaches are needed for the treatment and prevention of pneumococcal infections, and recent advances in the field may pave the way for new strategies. Recently, several important findings have been gained regarding pneumococcal epidemiology, genomics and the effect of the introduction of pneumococcal conjugate vaccines and of the COVID-19 pandemic. Moreover, elucidative pathogenesis studies have shown that the interactions between pneumococcal virulence factors and host receptors may be exploited for new therapies, and new vaccine candidates have been suggested. In this Review, we summarize some recent findings from clinical disease to basic pathogenesis studies that may be of importance for future control strategies.
Collapse
Affiliation(s)
- Ana Rita Narciso
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Dookie
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Priyanka Nannapaneni
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Normark
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
3
|
Liu X, Ke S, Wang X, Li Y, Lyu J, Liu Y, Geng Z. Interpretation of the anti-influenza active ingredients and potential mechanisms of Ge Gen Decoction based on spectrum-effect relationships and network analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117290. [PMID: 37806538 DOI: 10.1016/j.jep.2023.117290] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/16/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ge Gen Decoction (GGD) is a classic traditional Chinese medicine (TCM) prescription that originated in the ancient Chinese medical book "Treatise on Febrile Diseases". The prescription consists of 7 herbs: Pueraria lobata (Willd.) Ohwi, Ephedra sinica Stapf, Cinnamomum cassia (L.) J.Presl, Paeonia lactiflora Pall., Glycyrrhiza uralensis Fisch., Zingiber officinale Rosc., and Ziziphus jujuba Mill. It can alleviate high fever and soreness in the neck and shoulders caused by exogenous wind chill and is widely used in both China and Japan. Currently, GGD is primarily utilized for treating flu and the common cold. GGD has been reported to show significant anti-influenza A virus (IAV) activity both in vitro and in vivo. However, the active ingredients responsible for its anti-influenza properties have not been elucidated, and the mechanisms underlying its anti-influenza effects require further research. AIM OF THE STUDY This study aims to investigate the active ingredients and molecular mechanisms of GGD in treating influenza. MATERIALS AND METHODS HPLC chromatograms were established for GGD water and different polar extracts. The effect of different GGD extracts on pulmonary virus titers and TNFα expression was assessed through RT-PCR analysis. Spectrum-effect relationships between chromatographic peaks of GGD and its virus inhibition rate and TNFα inhibition rate were investigated using partial least squares regression (PLSR) analysis. HPLC-Q-TOF-MS was utilized to identify the constituents absorbed into the blood after oral administration of GGD. Network analysis of the absorbed forms of active ingredients was conducted to predict the potential mechanisms of GGD. Subsequently, total SOD activity, CAT and HO-1 expression and Nrf2 nuclear translocation were then analyzed. Finally, the impact of interfering with HO-1 expression on the anti-IAV activity of GGD was examined. RESULTS The study identified 11 anti-influenza active ingredients in GGD, which are daidzein, ononin, genistin, daidzin, 3'-methoxypuerarin, puerarin, pseudoephedrine, paeoniflorin, pormononetin-7-xylosyl-glucoside, penistein-7-O-apiosyl-glucoside, and ephedrine. Network analysis revealed various biological activities of GGD, including responses to ROS and oxidative stress. GGD also involves multiple antiviral pathways, such as hepatitis B, IAV, and Toll-like receptor pathways. Experimental assays demonstrated that GGD possesses independent antioxidant activity both in vitro and in vivo. In vitro, GGD inhibits the increase in intracellular ROS induced by IAV. In vivo, it reduces MDA levels and increases total pulmonary SOD activity. Applying siRNA and flow cytometry analysis revealed that GGD alleviates IAV-induced oxidative burst by promoting the expression of HO-1 and CAT. Western blot analysis revealed that GGD effectively promotes Nrf2 nuclear translocation and enhances Nrf2 expression. Furthermore, this study found that the enhancement of HO-1 expression by GGD contributed to its anti-IAV activity. CONCLUSIONS The study identified the active ingredients of GGD against influenza and demonstrated the beneficial role of GGD's antioxidant activity in treating flu. The antioxidant activity of GGD is associated with the promotion of Nrf2 nuclear translocation and the upregulation of antioxidant enzymes such as SOD, HO-1, and CAT. Overall, this study provides evidence supporting the use of GGD as an adjunctive or complementary therapy for influenza.
Collapse
Affiliation(s)
- Xiyu Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Siyuan Ke
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiuyi Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Yaqun Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiantao Lyu
- Pharmacy Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, China.
| | - Yu Liu
- Respiratory Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| | - Zikai Geng
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
4
|
Shirey KA, Lai W, Sunday ME, Cuttitta F, Blanco JCG, Vogel SN. Novel neuroendocrine role of γ-aminobutyric acid and gastrin-releasing peptide in the host response to influenza infection. Mucosal Immunol 2023; 16:302-311. [PMID: 36965691 PMCID: PMC10330014 DOI: 10.1016/j.mucimm.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023]
Abstract
Gastrin-releasing peptide (GRP), an evolutionarily conserved neuropeptide, significantly contributes to influenza-induced lethality and inflammation in rodent models. Because GRP is produced by pulmonary neuroendocrine cells (PNECs) in response to γ-aminobutyric acid (GABA), we hypothesized that influenza infection promotes GABA release from PNECs that activate GABAB receptors on PNECs to secrete GRP. Oxidative stress was increased in the lungs of influenza A/PR/8/34 (PR8)-infected mice, as well as serum glutamate decarboxylase 1, the enzyme that converts L-glutamic acid into GABA. The therapeutic administration of saclofen, a GABAB receptor antagonist, protected PR8-infected mice, reduced lung proinflammatory gene expression of C-C chemokine receptor type 2 (Ccr2), cluster of differentiation 68 (Cd68), and Toll like receptor 4 (Tlr4) and decreased the levels of GRP and high-mobility group box 1 (HMGB1) in sera. Conversely, baclofen, a GABAB receptor agonist, significantly increased the lethality and inflammatory responses. The GRP antagonist, NSC77427, as well as the GABAB antagonist, saclofen, blunted the PR8-induced monocyte infiltration into the lung. Together, these data provide the first report of neuroregulatory control of influenza-induced disease.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| | - Wendy Lai
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Mary E Sunday
- Duke University Medical Center, Durham, North Carolina, USA
| | - Frank Cuttitta
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | | | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Park J, Legaspi SLF, Schwartzman LM, Gygli SM, Sheng ZM, Freeman AD, Matthews LM, Xiao Y, Ramuta MD, Batchenkova NA, Qi L, Rosas LA, Williams SL, Scherler K, Gouzoulis M, Bellayr I, Morens DM, Walters KA, Memoli MJ, Kash JC, Taubenberger JK. An inactivated multivalent influenza A virus vaccine is broadly protective in mice and ferrets. Sci Transl Med 2022; 14:eabo2167. [PMID: 35857640 PMCID: PMC11022527 DOI: 10.1126/scitranslmed.abo2167] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Influenza A viruses (IAVs) present major public health threats from annual seasonal epidemics and pandemics and from viruses adapted to a variety of animals including poultry, pigs, and horses. Vaccines that broadly protect against all such IAVs, so-called "universal" influenza vaccines, do not currently exist but are urgently needed. Here, we demonstrated that an inactivated, multivalent whole-virus vaccine, delivered intramuscularly or intranasally, was broadly protective against challenges with multiple IAV hemagglutinin and neuraminidase subtypes in both mice and ferrets. The vaccine is composed of four β-propiolactone-inactivated low-pathogenicity avian IAV subtypes of H1N9, H3N8, H5N1, and H7N3. Vaccinated mice and ferrets demonstrated substantial protection against a variety of IAVs, including the 1918 H1N1 strain, the highly pathogenic avian H5N8 strain, and H7N9. We also observed protection against challenge with antigenically variable and heterosubtypic avian, swine, and human viruses. Compared to control animals, vaccinated mice and ferrets demonstrated marked reductions in viral titers, lung pathology, and host inflammatory responses. This vaccine approach indicates the feasibility of eliciting broad, heterosubtypic IAV protection and identifies a promising candidate for influenza vaccine clinical development.
Collapse
Affiliation(s)
- Jaekeun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharon L. Fong Legaspi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis M. Schwartzman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sebastian M. Gygli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ashley D. Freeman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lex M. Matthews
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mitchell D. Ramuta
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Natalia A. Batchenkova
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Qi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephanie L. Williams
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Monica Gouzoulis
- Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ian Bellayr
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - David M. Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Matthew J. Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John C. Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
D’Agnillo F, Walters KA, Xiao Y, Sheng ZM, Scherler K, Park J, Gygli S, Rosas LA, Sadtler K, Kalish H, Blatti CA, Zhu R, Gatzke L, Bushell C, Memoli MJ, O’Day SJ, Fischer TD, Hammond TC, Lee RC, Cash JC, Powers ME, O’Keefe GE, Butnor KJ, Rapkiewicz AV, Travis WD, Layne SP, Kash JC, Taubenberger JK. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med 2021; 13:eabj7790. [PMID: 34648357 PMCID: PMC11000440 DOI: 10.1126/scitranslmed.abj7790] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Felice D’Agnillo
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | | | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Jaekeun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Gygli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kalish
- Bioengineering and Physical Sciences Shared Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Charles A. Blatti
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ruoqing Zhu
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Lisa Gatzke
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Colleen Bushell
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Raymond C. Lee
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - J. Christian Cash
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Matthew E. Powers
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Grant E. O’Keefe
- Department of Surgery, University of Washington, Harborview Medical Center, Seattle, WA, USA
| | - Kelly J. Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Amy V. Rapkiewicz
- Department of Pathology, New York University Long Island School of Medicine, Mineola, NY, USA
| | - William D. Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - John C. Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Hsieh IN, White M, Hoeksema M, Deluna X, Hartshorn K. Histone H4 potentiates neutrophil inflammatory responses to influenza A virus: Down-modulation by H4 binding to C-reactive protein and Surfactant protein D. PLoS One 2021; 16:e0247605. [PMID: 33635872 PMCID: PMC7909658 DOI: 10.1371/journal.pone.0247605] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/10/2021] [Indexed: 01/05/2023] Open
Abstract
Neutrophils participate in the early phase of the innate response to uncomplicated influenza A virus (IAV) infection but also are a major component in later stages of severe IAV or COVID 19 infection where neutrophil extracellular traps (NETs) and associated cell free histones are highly pro-inflammatory. It is likely that IAV interacts with histones during infection. We show that histone H4 binds to IAV and aggregates viral particles. In addition, histone H4 markedly potentiates IAV induced neutrophil respiratory burst responses. Prior studies have shown reactive oxidants to be detrimental during severe IAV infection. C reactive protein (CRP) and surfactant protein D (SP-D) rise during IAV infection. We now show that both of these innate immune proteins bind to histone H4 and significantly down regulate respiratory burst and other responses to histone H4. Isolated constructs composed only of the neck and carbohydrate recognition domain of SP-D also bind to histone H4 and partially limit neutrophil responses to it. These studies indicate that complexes formed of histones and IAV are a potent neutrophil activating stimulus. This finding could account for excess inflammation during IAV or other severe viral infections. The ability of CRP and SP-D to bind to histone H4 may be part of a protective response against excessive inflammation in vivo.
Collapse
Affiliation(s)
- I-Ni Hsieh
- Department of Medicine, Section of Hematology Oncology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Mitchell White
- Department of Medicine, Section of Hematology Oncology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | | | - Xavier Deluna
- Department of Medicine, Section of Hematology Oncology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Kevan Hartshorn
- Department of Medicine, Section of Hematology Oncology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
8
|
Capillary leakage provides nutrients and antioxidants for rapid pneumococcal proliferation in influenza-infected lower airways. Proc Natl Acad Sci U S A 2020; 117:31386-31397. [PMID: 33229573 PMCID: PMC7733805 DOI: 10.1073/pnas.2012265117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Mechanisms for why influenza A virus (IAV) infections sensitize for pneumococcal infections are not clear. Here, we show that IAV-induced capillary leakage results in influx of nutrients and antioxidants to the lungs, thereby promoting pneumococcal growth in the lower respiratory tract. The evoked inflammation leads to redox imbalances that require bacterial adaptation to the oxidized environment, including induction of the pneumococcal chaperone/protease HtrA that protects the bacteria from clearance by the immune system. The results give us insight into the delicate interplay between the bacteria and the host environment during coinfections that needs to be explored in order to find novel therapeutic approaches. Influenza A virus (IAV)-related mortality is often due to secondary bacterial infections, primarily by pneumococci. Here, we study how IAV-modulated changes in the lungs affect bacterial replication in the lower respiratory tract (LRT). Bronchoalveolar lavages (BALs) from coinfected mice showed rapid bacterial proliferation 4 to 6 h after pneumococcal challenge. Metabolomic and quantitative proteomic analyses demonstrated capillary leakage with efflux of nutrients and antioxidants into the alveolar space. Pneumococcal adaptation to IAV-induced inflammation and redox imbalance increased the expression of the pneumococcal chaperone/protease HtrA. Presence of HtrA resulted in bacterial growth advantage in the IAV-infected LRT and protection from complement-mediated opsonophagocytosis due to capsular production. Absence of HtrA led to growth arrest in vitro that was partially restored by antioxidants. Pneumococcal ability to grow in the IAV-infected LRT depends on the nutrient-rich milieu with increased levels of antioxidants such as ascorbic acid and its ability to adapt to and cope with oxidative damage and immune clearance.
Collapse
|
9
|
Hartshorn KL. Innate Immunity and Influenza A Virus Pathogenesis: Lessons for COVID-19. Front Cell Infect Microbiol 2020; 10:563850. [PMID: 33194802 PMCID: PMC7642997 DOI: 10.3389/fcimb.2020.563850] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022] Open
Abstract
There is abundant evidence that the innate immune response to influenza A virus (IAV) is highly complex and plays a key role in protection against IAV induced infection and illness. Unfortunately it also clear that aspects of innate immunity can lead to severe morbidity or mortality from IAV, including inflammatory lung injury, bacterial superinfection, and exacerbation of reactive airways disease. We review broadly the virus and host factors that result in adverse outcomes from IAV and show evidence that inflammatory responses can become damaging even apart from changes in viral replication per se, with special focus on the positive and adverse effects of neutrophils and monocytes. We then evaluate in detail the role of soluble innate inhibitors including surfactant protein D and antimicrobial peptides that have a potential dual capacity for down-regulating viral replication and also inhibiting excessive inflammatory responses and how these innate host factors could possibly be harnessed to treat IAV infection. Where appropriate we draw comparisons and contrasts the SARS-CoV viruses and IAV in an effort to point out where the extensive knowledge existing regarding severe IAV infection could help guide research into severe COVID 19 illness or vice versa.
Collapse
Affiliation(s)
- Kevan L Hartshorn
- Section of Hematology Oncology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
10
|
Sauter JL, Baine MK, Butnor KJ, Buonocore DJ, Chang JC, Jungbluth AA, Szabolcs MJ, Morjaria S, Mount SL, Rekhtman N, Selbs E, Sheng ZM, Xiao Y, Kleiner DE, Pittaluga S, Taubenberger JK, Rapkiewicz AV, Travis WD. Insights into pathogenesis of fatal COVID-19 pneumonia from histopathology with immunohistochemical and viral RNA studies. Histopathology 2020; 77:915-925. [PMID: 32614086 PMCID: PMC7361244 DOI: 10.1111/his.14201] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/27/2020] [Indexed: 01/08/2023]
Abstract
Introduction We describe post‐mortem pulmonary histopathologic findings of COVID‐19 pneumonia in patients with a spectrum of disease course, from rapid demise to prolonged hospitalisation. Methods and results Histopathologic findings in post‐mortem lung tissue from eight patients who died from COVID‐19 pneumonia were reviewed. Immunohistochemistry (IHC) and next‐generation sequencing (NGS) were performed to detect virus. Diffuse alveolar damage (DAD) was seen in all cases with a spectrum of acute phase and/or organising phase. IHC with monoclonal antibodies against SARS‐CoV‐2 viral nucleoprotein and spike protein detected virus in areas of acute but not organising DAD, with intracellular viral antigen and RNA expression seen predominantly in patients with duration of illness less than 10 days. Major vascular findings included thrombi in medium‐ and large‐calibre vessels, platelet microthrombi detected by CD61 IHC and fibrin microthrombi. Conclusions Presence of SARS‐CoV‐2 viral RNA by NGS early in the disease course and expression of viral antigen by IHC exclusively in the acute, but not in the organising phase of DAD, suggests that the virus may play a major role in initiating the acute lung injury of DAD, but when DAD progresses to the organising phase the virus may have been cleared from the lung by the patient's immune response. These findings suggest the possibility of a major change during the disease course of COVID‐19 pneumonia that may have therapeutic implications. Frequent thrombi and microthrombi may also present potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer L Sauter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marina K Baine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelly J Butnor
- Department of Laboratory Medicine and Pathology, University of Vermont Medical Center, Burlington, VT, USA
| | - Darren J Buonocore
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthias J Szabolcs
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Sejal Morjaria
- Infectious Disease, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Sharon L Mount
- Department of Laboratory Medicine and Pathology, University of Vermont Medical Center, Burlington, VT, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elena Selbs
- Department of Pathology, New York University Long Island School of Medicine, Mineola, NY, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David E Kleiner
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stefania Pittaluga
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amy V Rapkiewicz
- Department of Pathology, New York University Long Island School of Medicine, Mineola, NY, USA
| | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
11
|
Taubenberger JK, Morens DM. The 1918 Influenza Pandemic and Its Legacy. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a038695. [PMID: 31871232 DOI: 10.1101/cshperspect.a038695] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Just over a century ago in 1918-1919, the "Spanish" influenza pandemic appeared nearly simultaneously around the world and caused extraordinary mortality-estimated at 50-100 million fatalities-associated with unexpected clinical and epidemiological features. The pandemic's sudden appearance and high fatality rate were unprecedented, and 100 years later still serve as a stark reminder of the continual threat influenza poses. Sequencing and reconstruction of the 1918 virus have allowed scientists to answer many questions about its origin and pathogenicity, although many questions remain. Several of the unusual features of the 1918-1919 pandemic, including age-specific mortality patterns and the high frequency of severe pneumonias, are still not fully understood. The 1918 pandemic virus initiated a pandemic era still ongoing. The descendants of the 1918 virus remain today as annually circulating and evolving influenza viruses causing significant mortality each year. This review summarizes key findings and unanswered questions about this deadliest of human events.
Collapse
Affiliation(s)
- Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - David M Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
12
|
Pursuing the Elixir of Life: In Vivo Antioxidative Effects of Manganosalen Complexes. Antioxidants (Basel) 2020; 9:antiox9080727. [PMID: 32785017 PMCID: PMC7465912 DOI: 10.3390/antiox9080727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
Manganosalen complexes are coordination compounds that possess a chelating salen-type ligand, a class of bis-Schiff bases obtained by condensation of salicylaldehyde and a diamine. They may act as catalytic antioxidants mimicking both the structure and the reactivity of the native antioxidant enzymes active site. Thus, manganosalen complexes have been shown to exhibit superoxide dismutase, catalase, and glutathione peroxidase activities, and they could potentially facilitate the scavenging of excess reactive oxygen species (ROS), thereby restoring the redox balance in damaged cells and organs. Initial catalytic studies compared the potency of these compounds as antioxidants in terms of rate constants of the chemical reactivity against ROS, giving catalytic values approaching and even exceeding that of the native antioxidative enzymes. Although most of these catalytic studies lack of biological relevance, subsequent in vitro studies have confirmed the efficiency of many manganosalen complexes in oxidative stress models. These synthetic catalytic scavengers, cheaper than natural antioxidants, have accordingly attracted intensive attention for the therapy of ROS-mediated injuries. The aim of this review is to focus on in vivo studies performed on manganosalen complexes and their activity on the treatment of several pathological disorders associated with oxidative damage. These disorders, ranging from the prevention of fetal malformations to the extension of lifespan, include neurodegenerative, inflammatory, and cardiovascular diseases; tissue injury; and other damages related to the liver, kidney, or lungs.
Collapse
|
13
|
Abstract
The year 2018 marked the 100th anniversary of the deadliest event in human history. In 1918-1919, pandemic influenza spread globally and caused an estimated 50-100 million deaths associated with unexpected clinical and epidemiological features. The descendants of the 1918 virus continue to circulate as annual epidemic viruses causing significant mortality each year. The 1918 influenza pandemic serves as a benchmark for the development of universal influenza vaccines. Challenges to producing a truly universal influenza vaccine include eliciting broad protection against antigenically different influenza viruses that can prevent or significantly downregulate viral replication and reduce morbidity by preventing development of viral and secondary bacterial pneumonia. Perhaps the most important goal of such vaccines is not to prevent influenza, but to prevent influenza deaths.
Collapse
Affiliation(s)
- David M Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
14
|
Rouco L, Liberato A, Fernández-Trujillo MJ, Máñez A, Basallote MG, Alvariño R, Alfonso A, Botana LM, Maneiro M. Salen‑manganese complexes for controlling ROS damage: Neuroprotective effects, antioxidant activity and kinetic studies. J Inorg Biochem 2019; 203:110918. [PMID: 31759263 DOI: 10.1016/j.jinorgbio.2019.110918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/23/2022]
Abstract
A new manganese(III) complex [MnL1(DCA)(H2O)](H2O),1 [H2L1 is the chelating ligand N,N'-bis(2-hydroxy-3-methoxybenzylidene)-1,2-diaminopropane, and DCA is dicyanamide], has been prepared and characterized by different analytical and spectroscopic techniques. The tetragonally elongated octahedral geometry for the manganese coordination sphere was revealed by X-ray diffraction studies for 1. The antioxidant behavior of this complex and other manganese(III)-salen type complexes was tested through superoxide dismutase and catalase probes, and through the study of their neuroprotective effects in SH-SY5Y neuroblastoma cells. In this human neuronal model, these model complexes were found to improve cell survival in an oxidative stress model. During studies aimed to getting a better understanding of the kinetics of the processes involved in this antioxidant behavior, an important effect on the solvent in the kinetics of reaction of the complexes with H2O2 was revealed that suggests a change in the mechanism of reaction of the complexes. The kinetic data in methanol and buffered aqueous solutions correlate well with the results of the test of catalase activity, thus showing that the rate determining step in the catalytic cycle corresponds to the initial reaction of the complexes with H2O2.
Collapse
Affiliation(s)
- Lara Rouco
- Departamento de Química Inorgánica, Facultade de Ciencias, Campus Terra, Universidade de Santiago de Compostela, Lugo, Spain
| | - Andrea Liberato
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Puerto Real, Cádiz, Spain
| | - M Jesús Fernández-Trujillo
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Puerto Real, Cádiz, Spain
| | - Angeles Máñez
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Puerto Real, Cádiz, Spain
| | - Manuel G Basallote
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Puerto Real, Cádiz, Spain
| | - Rebeca Alvariño
- Departamento. de Farmacología, Facultade de Veterinaria, Campus Terra, Universidade de Santiago de Compostela, Lugo, Spain
| | - Amparo Alfonso
- Departamento. de Farmacología, Facultade de Veterinaria, Campus Terra, Universidade de Santiago de Compostela, Lugo, Spain
| | - Luis M Botana
- Departamento. de Farmacología, Facultade de Veterinaria, Campus Terra, Universidade de Santiago de Compostela, Lugo, Spain
| | - Marcelino Maneiro
- Departamento de Química Inorgánica, Facultade de Ciencias, Campus Terra, Universidade de Santiago de Compostela, Lugo, Spain.
| |
Collapse
|
15
|
Puggioni F, Alves-Correia M, Mohamed MF, Stomeo N, Mager R, Marinoni M, Racca F, Paoletti G, Varricchi G, Giorgis V, Melioli G, Canonica GW, Heffler E. Immunostimulants in respiratory diseases: focus on Pidotimod. Multidiscip Respir Med 2019; 14:31. [PMID: 31700623 PMCID: PMC6827234 DOI: 10.1186/s40248-019-0195-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/23/2019] [Indexed: 12/18/2022] Open
Abstract
Usefulness of Pidotimod and its role as immunostimulant, has been discussed, we know, for several decades. Nevertheless, there is still much to know. Understanding its mechanisms and its potential usefulness in airway infections and its prevention, asthma both Th2 and non Th2 type, bronchiectasis, as adjuvant in vaccination and in allergen immunotherapy still remains to clearly unveil. The aim of this paper was to provide a useful updated review of the role of the main available immunostimulants, giving particular focus on Pidotimod use and its potentials utility in respiratory diseases. Pidotimod showed its usefulness in reducing need for antibiotics in airway infections, increasing the level of immunoglobulins (IgA, IgM, IgG) and T-lymphocyte subsets (CD3+, CD4+) endowed with immunomodulatory activity that affect both innate and adaptive immune responses. Higher expression of TLR2 and of HLA-DR molecules, induction of dendritic cell maturation and release of pro-inflammatory molecules, stimulation of T lymphocyte proliferation and differentiation toward a Th1 phenotype, as well as an increase of the phagocytosis have been demonstrated to be associated with Pidotimod in in vitro studies. All these activities are potentially useful for several respiratory conditions such as asthma, COPD, and recurrent respiratory tract infections.
Collapse
Affiliation(s)
- Francesca Puggioni
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| | - Magna Alves-Correia
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
- Central Hospital of Funchal, SESARAM, EPE, Madeira, Portugal
| | - Manar-Farouk Mohamed
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
- Ain Shams University, Faculty of Medicine, Cairo, Egypt
| | - Niccolò Stomeo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
| | - Riccardo Mager
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
| | | | - Francesca Racca
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| | - Giovanni Paoletti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Veronica Giorgis
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| | - Giovanni Melioli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI Italy
- Personalized Medicine, Allergy and Asthma - Humanitas Clinical and Research Center – IRCCS, Via Alessandro Manzoni 56, 20089 Rozzano, MI Italy
| |
Collapse
|
16
|
Taubenberger JK, Kash JC, Morens DM. The 1918 influenza pandemic: 100 years of questions answered and unanswered. Sci Transl Med 2019; 11:eaau5485. [PMID: 31341062 PMCID: PMC11000447 DOI: 10.1126/scitranslmed.aau5485] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022]
Abstract
The 2018-2019 period marks the centennial of the "Spanish" influenza pandemic, which caused at least 50 million deaths worldwide. The unprecedented nature of the pandemic's sudden appearance and high fatality rate serve as a stark reminder of the threat influenza poses. Unusual features of the 1918-1919 pandemic, including age-specific mortality and the high frequency of severe pneumonias, are still not fully understood. Sequencing and reconstruction of the 1918 virus has allowed scientists to answer many questions about its origin and pathogenicity, although many questions remain. This Review summarizes key findings and still-to-be answered questions about this deadliest of human events.
Collapse
Affiliation(s)
- Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David M Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Liberato A, Fernández-Trujillo MJ, Máñez Á, Maneiro M, Rodríguez-Silva L, Basallote MG. Pitfalls in the ABTS Peroxidase Activity Test: Interference of Photochemical Processes. Inorg Chem 2018; 57:14471-14475. [DOI: 10.1021/acs.inorgchem.8b02525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Andrea Liberato
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Avda. República Saharahui s/n, Puerto Real, 11510 Cádiz, Spain
| | - M. Jesús Fernández-Trujillo
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Avda. República Saharahui s/n, Puerto Real, 11510 Cádiz, Spain
| | - Ángeles Máñez
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Avda. República Saharahui s/n, Puerto Real, 11510 Cádiz, Spain
| | - Marcelino Maneiro
- Departamento de Química Inorgánica, Facultade de Ciencias, Campus de Lugo, Universidade de Santiago de Compostela, Avda. Alfonso X s/n, Lugo 27002, Spain
| | - Laura Rodríguez-Silva
- Departamento de Química Inorgánica, Facultade de Ciencias, Campus de Lugo, Universidade de Santiago de Compostela, Avda. Alfonso X s/n, Lugo 27002, Spain
| | - Manuel G. Basallote
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Facultad de Ciencias, Universidad de Cádiz, Avda. República Saharahui s/n, Puerto Real, 11510 Cádiz, Spain
| |
Collapse
|
18
|
Redox Biology of Respiratory Viral Infections. Viruses 2018; 10:v10080392. [PMID: 30049972 PMCID: PMC6115776 DOI: 10.3390/v10080392] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/17/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022] Open
Abstract
Respiratory viruses cause infections of the upper or lower respiratory tract and they are responsible for the common cold—the most prevalent disease in the world. In many cases the common cold results in severe illness due to complications, such as fever or pneumonia. Children, old people, and immunosuppressed patients are at the highest risk and require fast diagnosis and therapeutic intervention. However, the availability and efficiencies of existing therapeutic approaches vary depending on the virus. Investigation of the pathologies that are associated with infection by respiratory viruses will be paramount for diagnosis, treatment modalities, and the development of new therapies. Changes in redox homeostasis in infected cells are one of the key events that is linked to infection with respiratory viruses and linked to inflammation and subsequent tissue damage. Our review summarizes current knowledge on changes to redox homeostasis, as induced by the different respiratory viruses.
Collapse
|
19
|
Takizawa N, Kimura T, Watanabe T, Shibasaki M. Anti-influenza virus activity of a salcomine derivative mediated by inhibition of viral RNA synthesis. Arch Virol 2018; 163:1607-1614. [PMID: 29497849 DOI: 10.1007/s00705-018-3779-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/19/2018] [Indexed: 12/25/2022]
Abstract
Influenza virus infection is a major threat to global health. Although vaccines and anti-influenza virus drugs are available, annual influenza virus epidemics result in severe illness, and an influenza pandemic occurs every 20-30 years. To identify candidate anti-influenza virus compounds, we screened approximately 5,000 compounds in an in-house library. We identified MZ7465, a salcomine derivative, as a potent inhibitor of influenza virus propagation. We analyzed the antiviral propagation mechanism of the hit compound by determining the amounts of viral proteins and RNA in infected cells treated with or without the hit compound. Treatment of infected cells with MZ7465 decreased both viral protein and RNA synthesis. In addition, an in vitro assay showed that viral RNA synthesis was directly inhibited by MZ7465. These results suggest that salcomine and its derivatives are potential candidates for the treatment of influenza virus infections.
Collapse
MESH Headings
- Animals
- Antiviral Agents/chemistry
- Antiviral Agents/pharmacology
- Bronchi/drug effects
- Bronchi/pathology
- Bronchi/virology
- Cell Line
- Dogs
- Epithelial Cells/drug effects
- Epithelial Cells/pathology
- Epithelial Cells/virology
- Gene Expression Regulation, Viral
- HEK293 Cells
- High-Throughput Screening Assays
- Humans
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/growth & development
- Influenza A Virus, H3N2 Subtype/drug effects
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/growth & development
- Madin Darby Canine Kidney Cells
- Organometallic Compounds/chemistry
- Organometallic Compounds/pharmacology
- RNA, Viral/antagonists & inhibitors
- RNA, Viral/biosynthesis
- RNA, Viral/genetics
- Small Molecule Libraries/chemistry
- Small Molecule Libraries/pharmacology
- Structure-Activity Relationship
- Viral Proteins/antagonists & inhibitors
- Viral Proteins/biosynthesis
- Viral Proteins/genetics
- Virus Replication/drug effects
Collapse
Affiliation(s)
- Naoki Takizawa
- Laboratory of Virology, Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan.
| | - Tomoyuki Kimura
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
| | - Takumi Watanabe
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
| | - Masakatsu Shibasaki
- Laboratory of Virology, Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
- Laboratory of Synthetic Organic Chemistry, Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
| |
Collapse
|
20
|
Differential Ability of Pandemic and Seasonal H1N1 Influenza A Viruses To Alter the Function of Human Neutrophils. mSphere 2018; 3:mSphere00567-17. [PMID: 29299535 PMCID: PMC5750393 DOI: 10.1128/mspheredirect.00567-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
A long-standing notion is that IAV inhibits normal neutrophil function and thereby predisposes individuals to secondary bacterial infections. Here we report that seasonal H1N1 IAV primes human neutrophils for enhanced killing of Staphylococcus aureus. Moreover, we provide a comprehensive view of the changes in neutrophil gene expression during interaction with seasonal or pandemic IAV and report how these changes relate to functions such as bactericidal activity. This study expands our knowledge of IAV interactions with human neutrophils. Neutrophils are essential cells of host innate immunity. Although the role of neutrophils in defense against bacterial and fungal infections is well characterized, there is a relative paucity of information about their role against viral infections. Influenza A virus (IAV) infection can be associated with secondary bacterial coinfection, and it has long been posited that the ability of IAV to alter normal neutrophil function predisposes individuals to secondary bacterial infections. To better understand this phenomenon, we evaluated the interaction of pandemic or seasonal H1N1 IAV with human neutrophils isolated from healthy persons. These viruses were ingested by human neutrophils and elicited changes in neutrophil gene expression that are consistent with an interferon-mediated immune response. The viability of neutrophils following coculture with either pandemic or seasonal H1N1 IAV was similar for up to 18 h of culture. Notably, neutrophil exposure to seasonal (but not pandemic) IAV primed these leukocytes for enhanced functions, including production of reactive oxygen species and bactericidal activity. Taken together, our results are at variance with the universal idea that IAV impairs neutrophil function directly to predispose individuals to secondary bacterial infections. Rather, we suggest that some strains of IAV prime neutrophils for enhanced bacterial clearance. IMPORTANCE A long-standing notion is that IAV inhibits normal neutrophil function and thereby predisposes individuals to secondary bacterial infections. Here we report that seasonal H1N1 IAV primes human neutrophils for enhanced killing of Staphylococcus aureus. Moreover, we provide a comprehensive view of the changes in neutrophil gene expression during interaction with seasonal or pandemic IAV and report how these changes relate to functions such as bactericidal activity. This study expands our knowledge of IAV interactions with human neutrophils.
Collapse
|
21
|
Liu JX, Zhang Y, Hu QP, Li JQ, Liu YT, Wu QG, Wu JG, Lai XP, Zhang ZD, Li X, Li G. Anti-inflammatory effects of rosmarinic acid-4-O-β-D-glucoside in reducing acute lung injury in mice infected with influenza virus. Antiviral Res 2017; 144:34-43. [PMID: 28461072 DOI: 10.1016/j.antiviral.2017.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 03/29/2017] [Accepted: 04/03/2017] [Indexed: 11/29/2022]
Abstract
Rosmarinic acid-4-O-β-D-glucoside (RAG) is a dicaffeoyl phenolic compound isolated from Sarcandra glabra (Thunb.) Nakai. Preliminary studies show that RAG has significant anti-inflammatory properties and can alleviate ear swelling in mice and the paw swelling in rats. Here, the anti-influenza effects of RAG were investigated in mice infected with A/FM/1/47 H1N1 virus. The survival rate and body weight were observed, the lung edema, virus copies, inflammatory cytokines (including IL-4, IL-5, TNF-α and IFN-γ) and oxidative damage indexes (including SOD, MDA, NO, and CAT) were measured. Moreover, immune cell recruitment in alveoli was measured with white blood cells and differential counts. Therapeutic RAG concentrations substantially improve the symptoms, mitigate body weight loss and alleviate lung edema induced by virus, thus improve survival protection effects. Furthermore, RAG was shown to regulate influenza virus-induced inflammatory cytokine expression, specifically by downregulating the Th1 cell cytokines IFN-γ, TNF-α and upregulating the Th2 cell cytokines IL-4, IL-5. Cell migration and infiltration were also diminished after RAG administration.
Collapse
Affiliation(s)
- Jian-Xing Liu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ying Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qiu-Ping Hu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ji-Qiang Li
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Yun-Tao Liu
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Qing-Guang Wu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jian-Guo Wu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiao-Ping Lai
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Dongguan, 523808, China
| | - Zhong-de Zhang
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Xiong Li
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China.
| | - Geng Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
22
|
Morris DE, Cleary DW, Clarke SC. Secondary Bacterial Infections Associated with Influenza Pandemics. Front Microbiol 2017; 8:1041. [PMID: 28690590 PMCID: PMC5481322 DOI: 10.3389/fmicb.2017.01041] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/24/2017] [Indexed: 12/16/2022] Open
Abstract
Lower and upper respiratory infections are the fourth highest cause of global mortality (Lozano et al., 2012). Epidemic and pandemic outbreaks of respiratory infection are a major medical concern, often causing considerable disease and a high death toll, typically over a relatively short period of time. Influenza is a major cause of epidemic and pandemic infection. Bacterial co/secondary infection further increases morbidity and mortality of influenza infection, with Streptococcus pneumoniae, Haemophilus influenzae, and Staphylococcus aureus reported as the most common causes. With increased antibiotic resistance and vaccine evasion it is important to monitor the epidemiology of pathogens in circulation to inform clinical treatment and development, particularly in the setting of an influenza epidemic/pandemic.
Collapse
Affiliation(s)
- Denise E. Morris
- Infectious Disease Epidemiology Group, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, University Hospital Southampton Foundation NHS TrustSouthampton, United Kingdom
| | - David W. Cleary
- Infectious Disease Epidemiology Group, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, University Hospital Southampton Foundation NHS TrustSouthampton, United Kingdom
| | - Stuart C. Clarke
- Infectious Disease Epidemiology Group, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, University Hospital Southampton Foundation NHS TrustSouthampton, United Kingdom
- Global Health Research Institute, University of SouthamptonSouthampton, United Kingdom
- NIHR Southampton Respiratory Biomedical Research UnitSouthampton, United Kingdom
| |
Collapse
|
23
|
Kobzik L. Searching for a Lifeline: Transcriptome Profiling Studies of Influenza Susceptibility and Resistance. J Innate Immun 2017; 9:232-242. [PMID: 28249256 DOI: 10.1159/000457902] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/24/2017] [Indexed: 11/19/2022] Open
Abstract
Excess or dysregulated host inflammatory responses cause much of the morbidity and mortality caused by severe influenza. Given the limitations of vaccines and antiviral drugs, novel therapeutics to modulate host responses and improve outcomes in severe influenza are needed. One strategy is to learn from the direct comparison of high-survivor versus high-mortality animal models. This review surveys the results of lung transcriptome profiling studies in murine models that directly compare susceptible versus resistant hosts challenged with identical influenza infections. The potential contributions and limitations of these studies are discussed. To amplify their power, the studies are subjected to a meta-analysis, which helps identify frequently dysregulated pathways and potentially novel areas for investigation. Using connectivity map-based tools (LINCS), transcriptome signatures linked to susceptibility can identify candidate drugs that merit testing for in vivo efficacy.
Collapse
Affiliation(s)
- Lester Kobzik
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, and Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
24
|
Wu X, Wu X, Sun Q, Zhang C, Yang S, Li L, Jia Z. Progress of small molecular inhibitors in the development of anti-influenza virus agents. Am J Cancer Res 2017; 7:826-845. [PMID: 28382157 PMCID: PMC5381247 DOI: 10.7150/thno.17071] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/18/2016] [Indexed: 02/05/2023] Open
Abstract
The influenza pandemic is a major threat to human health, and highly aggressive strains such as H1N1, H5N1 and H7N9 have emphasized the need for therapeutic strategies to combat these pathogens. Influenza anti-viral agents, especially active small molecular inhibitors play important roles in controlling pandemics while vaccines are developed. Currently, only a few drugs, which function as influenza neuraminidase (NA) inhibitors and M2 ion channel protein inhibitors, are approved in clinical. However, the acquired resistance against current anti-influenza drugs and the emerging mutations of influenza virus itself remain the major challenging unmet medical needs for influenza treatment. It is highly desirable to identify novel anti-influenza agents. This paper reviews the progress of small molecular inhibitors act as antiviral agents, which include hemagglutinin (HA) inhibitors, RNA-dependent RNA polymerase (RdRp) inhibitors, NA inhibitors and M2 ion channel protein inhibitors etc. Moreover, we also summarize new, recently reported potential targets and discuss strategies for the development of new anti-influenza virus drugs.
Collapse
|
25
|
Hu J, Gao Z, Wang X, Gu M, Liang Y, Liu X, Hu S, Liu H, Liu W, Chen S, Peng D, Liu X. iTRAQ-based quantitative proteomics reveals important host factors involved in the high pathogenicity of the H5N1 avian influenza virus in mice. Med Microbiol Immunol 2016; 206:125-147. [PMID: 28000052 DOI: 10.1007/s00430-016-0489-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023]
Abstract
We previously reported a pair of H5N1 avian influenza viruses which are genetically similar but differ greatly in their virulence in mice. A/Chicken/Jiangsu/k0402/2010 (CK10) is highly lethal to mice, whereas A/Goose/Jiangsu/k0403/2010 (GS10) is avirulent. In this study, to investigate the host factors that account for their virulence discrepancy, we compared the pathology and host proteome of the CK10- or GS10-infected mouse lung. Moderate lung injury was observed from CK10-infected animals as early as the first day of infection, and the pathology steadily progressed at later time point. However, only mild lesions were observed in GS10-infected mouse lung at the late infection stage. Using the quantitative iTRAQ coupled LC-MS/MS method, we first found that more significantly differentially expressed (DE) proteins were stimulated by GS10 compared with CK10. However, bio-function analysis of the DE proteins suggested that CK10 induced much stronger inflammatory response-related functions than GS10. Canonical pathway analysis also demonstrated that CK10 highly activated the "Acute Phase Response Signaling," which results in a wide range of biological activities in response to viral infection, including many inflammatory processes. Further in-depth analysis showed that CK10 exacerbated acute lung injury-associated responses, including inflammatory response, cell death, reactive oxygen species production and complement response. In addition, some of these identified proteins that associated with the lung injury were further confirmed to be regulated in vitro. Therefore, our findings suggest that the early increased lung injury-associated host response induced by CK10 may contribute to the lung pathology and the high virulence of this virus in mice.
Collapse
Affiliation(s)
- Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Zhao Gao
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Min Gu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Yanyan Liang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Huimou Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Wenbo Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Sujuan Chen
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Daxin Peng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu Province, China. .,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
26
|
Troy NM, Bosco A. Respiratory viral infections and host responses; insights from genomics. Respir Res 2016; 17:156. [PMID: 27871304 PMCID: PMC5117516 DOI: 10.1186/s12931-016-0474-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/10/2016] [Indexed: 01/23/2023] Open
Abstract
Respiratory viral infections are a leading cause of disease and mortality. The severity of these illnesses can vary markedly from mild or asymptomatic upper airway infections to severe wheezing, bronchiolitis or pneumonia. In this article, we review the viral sensing pathways and organizing principles that govern the innate immune response to infection. Then, we reconstruct the molecular networks that differentiate symptomatic from asymptomatic respiratory viral infections, and identify the underlying molecular drivers of these networks. Finally, we discuss unique aspects of the biology and pathogenesis of infections with respiratory syncytial virus, rhinovirus and influenza, drawing on insights from genomics.
Collapse
Affiliation(s)
- Niamh M Troy
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia.
| |
Collapse
|
27
|
Hu J, Mo Y, Gao Z, Wang X, Gu M, Liang Y, Cheng X, Hu S, Liu W, Liu H, Chen S, Liu X, Peng D, Liu X. PA-X-associated early alleviation of the acute lung injury contributes to the attenuation of a highly pathogenic H5N1 avian influenza virus in mice. Med Microbiol Immunol 2016; 205:381-95. [PMID: 27289459 PMCID: PMC7086737 DOI: 10.1007/s00430-016-0461-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/31/2016] [Indexed: 12/18/2022]
Abstract
PA-X is a novel discovered accessory protein encoded by the PA mRNA. Our previous study demonstrated that PA-X decreases the virulence of a highly pathogenic H5N1 strain A/Chicken/Jiangsu/k0402/2010 in mice. However, the underlying mechanism of virulence attenuation associated with PA-X is still unknown. In this study, we compared two PA-X-deficient mutant viruses and the parental virus in terms of induction of pathology and manipulation of host response in the mouse lung, stimulation of cell death and PA nuclear accumulation. We first found that down-regulated PA-X expression markedly aggravated the acute lung injury of the infected mice early on day 1 post-infection (p.i.). We then determined that loss of PA-X expression induced higher levels of cytokines, chemokines and complement-derived peptides (C3a and C5a) in the lung, especially at early time point’s p.i. In addition, in vitro assays showed that the PA-X-deficient viruses enhanced cell death and increased expression of reactive oxygen species (ROS) in mammalian cells. Moreover, we also found that PA nuclear accumulation of the PA-X-null viruses accelerated in MDCK cells. These results demonstrate that PA-X decreases the level of complement components, ROS, cell death and inflammatory response, which may together contribute to the alleviated lung injury and the attenuation of the virulence of H5N1 virus in mice.
Collapse
Affiliation(s)
- Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Yiqun Mo
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Zhao Gao
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Min Gu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Yanyan Liang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xin Cheng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Wenbo Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Huimou Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Sujuan Chen
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Daxing Peng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu Province, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
28
|
Walters KA, D'Agnillo F, Sheng ZM, Kindrachuk J, Schwartzman LM, Kuestner RE, Chertow DS, Golding BT, Taubenberger JK, Kash JC. 1918 pandemic influenza virus and Streptococcus pneumoniae co-infection results in activation of coagulation and widespread pulmonary thrombosis in mice and humans. J Pathol 2015; 238:85-97. [PMID: 26383585 DOI: 10.1002/path.4638] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/25/2015] [Accepted: 09/07/2015] [Indexed: 12/15/2022]
Abstract
To study bacterial co-infection following 1918 H1N1 influenza virus infection, mice were inoculated with the 1918 influenza virus, followed by Streptococcus pneumoniae (SP) 72 h later. Co-infected mice exhibited markedly more severe disease, shortened survival time and more severe lung pathology, including widespread thrombi. Transcriptional profiling revealed activation of coagulation only in co-infected mice, consistent with the extensive thrombogenesis observed. Immunohistochemistry showed extensive expression of tissue factor (F3) and prominent deposition of neutrophil elastase on endothelial and epithelial cells in co-infected mice. Lung sections of SP-positive 1918 autopsy cases showed extensive thrombi and prominent staining for F3 in alveolar macrophages, monocytes, neutrophils, endothelial and epithelial cells, in contrast to co-infection-positive 2009 pandemic H1N1 autopsy cases. This study reveals that a distinctive feature of 1918 influenza virus and SP co-infection in mice and humans is extensive expression of tissue factor and activation of the extrinsic coagulation pathway leading to widespread pulmonary thrombosis.
Collapse
Affiliation(s)
| | - Felice D'Agnillo
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - Jason Kindrachuk
- Critical Care Medicine Department, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Louis M Schwartzman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | | | - Daniel S Chertow
- Critical Care Medicine Department, National Institutes of Health (NIH), Bethesda, MD, USA.,Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - Basil T Golding
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
29
|
Tovmasyan A, Maia CGC, Weitner T, Carballal S, Sampaio RS, Lieb D, Ghazaryan R, Ivanovic-Burmazovic I, Ferrer-Sueta G, Radi R, Reboucas JS, Spasojevic I, Benov L, Batinic-Haberle I. A comprehensive evaluation of catalase-like activity of different classes of redox-active therapeutics. Free Radic Biol Med 2015; 86:308-21. [PMID: 26026699 PMCID: PMC4554972 DOI: 10.1016/j.freeradbiomed.2015.05.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/12/2015] [Accepted: 05/12/2015] [Indexed: 01/20/2023]
Abstract
Because of the increased insight into the biological role of hydrogen peroxide (H2O2) under physiological and pathological conditions and the role it presumably plays in the action of natural and synthetic redox-active drugs, there is a need to accurately define the type and magnitude of reactions that may occur with this intriguing and key species of redoxome. Historically, and frequently incorrectly, the impact of catalase-like activity has been assigned to play a major role in the action of many redox-active drugs, mostly SOD mimics and peroxynitrite scavengers, and in particular MnTBAP(3-) and Mn salen derivatives. The advantage of one redox-active compound over another has often been assigned to the differences in catalase-like activity. Our studies provide substantial evidence that Mn(III) N-alkylpyridylporphyrins couple with H2O2 in actions other than catalase-related. Herein we have assessed the catalase-like activities of different classes of compounds: Mn porphyrins (MnPs), Fe porphyrins (FePs), Mn(III) salen (EUK-8), and Mn(II) cyclic polyamines (SOD-active M40403 and SOD-inactive M40404). Nitroxide (tempol), nitrone (NXY-059), ebselen, and MnCl2, which have not been reported as catalase mimics, were used as negative controls, while catalase enzyme was a positive control. The dismutation of H2O2 to O2 and H2O was followed via measuring oxygen evolved with a Clark oxygen electrode at 25°C. The catalase enzyme was found to have kcat(H2O2)=1.5×10(6)M(-1) s(-1). The yield of dismutation, i.e., the maximal amount of O2 evolved, was assessed also. The magnitude of the yield reflects an interplay between the kcat(H2O2) and the stability of compounds toward H2O2-driven oxidative degradation, and is thus an accurate measure of the efficacy of a catalyst. The kcat(H2O2) values for 12 cationic Mn(III) N-substituted (alkyl and alkoxyalkyl) pyridylporphyrin-based SOD mimics and Mn(III) N,N'-dialkylimidazolium porphyrin, MnTDE-2-ImP(5+), ranged from 23 to 88M(-1) s(-1). The analogous Fe(III) N-alkylpyridylporphyrins showed ~10-fold higher activity than the corresponding MnPs, but the values of kcat(H2O2) are still ~4 orders of magnitude lower than that of the enzyme. While the kcat(H2O2) values for Fe ethyl and n-octyl analogs were 803.5 and 368.4M(-1) s(-1), respectively, the FePs are more prone to H2O2-driven oxidative degradation, therefore allowing for similar yields in H2O2 dismutation as analogous MnPs. The kcat(H2O2) values are dependent on the electron deficiency of the metal site as it controls the peroxide binding in the first step of the dismutation process. SOD-like activities depend on electron deficiency of the metal site also, as it controls the first step of O2(●-) dismutation. In turn, the kcat(O2(●-)) parallels the kcat(H2O2). Therefore, the electron-rich anionic non-SOD mimic MnTBAP(3-) has essentially very low catalase-like activity, kcat(H2O2)=5.8M(-1) s(-1). The catalase-like activities of Mn(III) and Fe(III) porphyrins are at most, 0.0004 and 0.05% of the enzyme activity, respectively. The kcat(H2O2) values of 8.2 and 6.5M(-1) s(-1) were determined for electron-rich Mn(II) cyclic polyamine-based compounds, M40403 and M40404, respectively. The EUK-8, with modest SOD-like activity, has only slightly higher kcat(H2O2)=13.5M(-1) s(-1). The biological relevance of kcat(H2O2) of MnTE-2-PyP(5+), MnTDE-2-ImP(5+), MnTBAP(3-), FeTE-2-PyP(5+), M40403, M40404, and Mn salen was evaluated in wild-type and peroxidase/catalase-deficient E. coli.
Collapse
Affiliation(s)
- Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Clarissa G C Maia
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA; Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Tin Weitner
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sebastián Carballal
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Romulo S Sampaio
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA; Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Dominik Lieb
- Friedrich-Alexander Universitat, Erlangen-Nurnberg, Germany
| | - Robert Ghazaryan
- Department of Organic Chemistry, Faculty of Pharmacy, Yerevan State Medical University, Armenia
| | | | - Gerardo Ferrer-Sueta
- Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias and Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Julio S Reboucas
- Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Pharmaceutical Research Shared Resource, PK/PD Core Laboratory, Durham, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
30
|
Pleiotropic Effects of Levofloxacin, Fluoroquinolone Antibiotics, against Influenza Virus-Induced Lung Injury. PLoS One 2015; 10:e0130248. [PMID: 26086073 PMCID: PMC4473075 DOI: 10.1371/journal.pone.0130248] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 05/19/2015] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) and nitric oxide (NO) are major pathogenic molecules produced during viral lung infections, including influenza. While fluoroquinolones are widely used as antimicrobial agents for treating a variety of bacterial infections, including secondary infections associated with the influenza virus, it has been reported that they also function as anti-oxidants against ROS and as a NO regulator. Therefore, we hypothesized that levofloxacin (LVFX), one of the most frequently used fluoroquinolone derivatives, may attenuate pulmonary injuries associated with influenza virus infections by inhibiting the production of ROS species such as hydroxyl radicals and neutrophil-derived NO that is produced during an influenza viral infection. The therapeutic impact of LVFX was examined in a PR8 (H1N1) influenza virus-induced lung injury mouse model. ESR spin-trapping experiments indicated that LVFX showed scavenging activity against neutrophil-derived hydroxyl radicals. LVFX markedly improved the survival rate of mice that were infected with the influenza virus in a dose-dependent manner. In addition, the LVFX treatment resulted in a dose-dependent decrease in the level of 8-hydroxy-2'-deoxyguanosine (a marker of oxidative stress) and nitrotyrosine (a nitrative marker) in the lungs of virus-infected mice, and the nitrite/nitrate ratio (NO metabolites) and IFN-γ in BALF. These results indicate that LVFX may be of substantial benefit in the treatment of various acute inflammatory disorders such as influenza virus-induced pneumonia, by inhibiting inflammatory cell responses and suppressing the overproduction of NO in the lungs.
Collapse
|
31
|
Wang J, Tang C, Wang Q, Li R, Chen Z, Han X, Wang J, Xu X. Apoptosis induction and release of inflammatory cytokines in the oviduct of egg-laying hens experimentally infected with H9N2 avian influenza virus. Vet Microbiol 2015; 177:302-14. [DOI: 10.1016/j.vetmic.2015.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 03/31/2015] [Accepted: 04/04/2015] [Indexed: 02/04/2023]
|
32
|
Kash JC, Taubenberger JK. The role of viral, host, and secondary bacterial factors in influenza pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1528-36. [PMID: 25747532 DOI: 10.1016/j.ajpath.2014.08.030] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 12/20/2022]
Abstract
Influenza A virus infections in humans generally cause self-limited infections, but can result in severe disease, secondary bacterial pneumonias, and death. Influenza viruses can replicate in epithelial cells throughout the respiratory tree and can cause tracheitis, bronchitis, bronchiolitis, diffuse alveolar damage with pulmonary edema and hemorrhage, and interstitial and airspace inflammation. The mechanisms by which influenza infections result in enhanced disease, including development of pneumonia and acute respiratory distress, are multifactorial, involving host, viral, and bacterial factors. Host factors that enhance risk of severe influenza disease include underlying comorbidities, such as cardiac and respiratory disease, immunosuppression, and pregnancy. Viral parameters enhancing disease risk include polymerase mutations associated with host switch and adaptation, viral proteins that modulate immune and antiviral responses, and virulence factors that increase disease severity, which can be especially prominent in pandemic viruses and some zoonotic influenza viruses causing human infections. Influenza viral infections result in damage to the respiratory epithelium that facilitates secondary infection with common bacterial pneumopathogens and can lead to secondary bacterial pneumonias that greatly contribute to respiratory distress, enhanced morbidity, and death. Understanding the molecular mechanisms by which influenza and secondary bacterial infections, coupled with the role of host risk factors, contribute to enhanced morbidity and mortality is essential to develop better therapeutic strategies to treat severe influenza.
Collapse
Affiliation(s)
- John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
33
|
Inhibition of reactive oxygen species production ameliorates inflammation induced by influenza A viruses via upregulation of SOCS1 and SOCS3. J Virol 2014; 89:2672-83. [PMID: 25520513 DOI: 10.1128/jvi.03529-14] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Highly pathogenic avian influenza virus infection is associated with severe mortality in both humans and poultry. The mechanisms of disease pathogenesis and immunity are poorly understood although recent evidence suggests that cytokine/chemokine dysregulation contributes to disease severity following H5N1 infection. Influenza A virus infection causes a rapid influx of inflammatory cells, resulting in increased reactive oxygen species production, cytokine expression, and acute lung injury. Proinflammatory stimuli are known to induce intracellular reactive oxygen species by activating NADPH oxidase activity. We therefore hypothesized that inhibition of this activity would restore host cytokine homeostasis following avian influenza virus infection. A panel of airway epithelial and immune cells from mammalian and avian species were infected with A/Puerto Rico/8/1934 H1N1 virus, low-pathogenicity avian influenza H5N3 virus (A/duck/Victoria/0305-2/2012), highly pathogenic avian influenza H5N1 virus (A/chicken/Vietnam/0008/2004), or low-pathogenicity avian influenza H7N9 virus (A/Anhui/1/2013). Quantitative real-time reverse transcriptase PCR showed that H5N1 and H7N9 viruses significantly stimulated cytokine (interleukin-6, beta interferon, CXCL10, and CCL5) production. Among the influenza-induced cytokines, CCL5 was identified as a potential marker for overactive immunity. Apocynin, a Nox2 inhibitor, inhibited influenza-induced cytokines and reactive oxygen species production, although viral replication was not significantly altered in vitro. Interestingly, apocynin treatment significantly increased influenza virus-induced mRNA and protein expression of SOCS1 and SOCS3, enhancing negative regulation of cytokine signaling. These findings suggest that apocynin or its derivatives (targeting host responses) could be used in combination with antiviral strategies (targeting viruses) as therapeutic agents to ameliorate disease severity in susceptible species. IMPORTANCE Highly pathogenic avian influenza virus infection causes severe morbidity and mortality in both humans and poultry. Wide-spread antiviral resistance necessitates the need for the development of additional novel therapeutic measures to modulate overactive host immune responses after infection. Disease severity following avian influenza virus infection can be attributed in part to hyperinduction of inflammatory mediators such as cytokines, chemokines, and reactive oxygen species. Our study shows that highly pathogenic avian influenza H5N1 virus and low-pathogenicity avian influenza H7N9 virus (both associated with human fatalities) promote inactivation of FoxO3 and downregulation of the TAM receptor tyrosine kinase, Tyro3, leading to augmentation of the inflammatory cytokine response. Inhibition of influenza-induced reactive oxygen species with apocynin activated FoxO3 and stimulated SOCS1 and SOCS3 proteins, restoring cytokine homeostasis. We conclude that modulation of host immune responses with antioxidant and/or anti-inflammatory agents in combination with antiviral therapy may have important therapeutic benefits.
Collapse
|
34
|
Contemporary avian influenza A virus subtype H1, H6, H7, H10, and H15 hemagglutinin genes encode a mammalian virulence factor similar to the 1918 pandemic virus H1 hemagglutinin. mBio 2014; 5:e02116. [PMID: 25406382 PMCID: PMC4251996 DOI: 10.1128/mbio.02116-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Zoonotic avian influenza virus infections may lead to epidemics or pandemics. The 1918 pandemic influenza virus has an avian influenza virus-like genome, and its H1 hemagglutinin was identified as a key mammalian virulence factor. A chimeric 1918 virus expressing a contemporary avian H1 hemagglutinin, however, displayed murine pathogenicity indistinguishable from that of the 1918 virus. Here, isogenic chimeric avian influenza viruses were constructed on an avian influenza virus backbone, differing only by hemagglutinin subtype expressed. Viruses expressing the avian H1, H6, H7, H10, and H15 subtypes were pathogenic in mice and cytopathic in normal human bronchial epithelial cells, in contrast to H2-, H3-, H5-, H9-, H11-, H13-, H14-, and H16-expressing viruses. Mouse pathogenicity was associated with pulmonary macrophage and neutrophil recruitment. These data suggest that avian influenza virus hemagglutinins H1, H6, H7, H10, and H15 contain inherent mammalian virulence factors and likely share a key virulence property of the 1918 virus. Consequently, zoonotic infections with avian influenza viruses bearing one of these hemagglutinins may cause enhanced disease in mammals. Influenza viruses from birds can cause outbreaks in humans and may contribute to the development of pandemics. The 1918 pandemic influenza virus has an avian influenza virus-like genome, and its main surface protein, an H1 subtype hemagglutinin, was identified as a key mammalian virulence factor. In a previous study, a 1918 virus expressing an avian H1 gene was as virulent in mice as the reconstructed 1918 virus. Here, a set of avian influenza viruses was constructed, differing only by hemagglutinin subtype. Viruses with the avian H1, H6, H7, H10, and H15 subtypes caused severe disease in mice and damaged human lung cells. Consequently, infections with avian influenza viruses bearing one of these hemagglutinins may cause enhanced disease in mammals, and therefore surveillance for human infections with these subtypes may be important in controlling future outbreaks.
Collapse
|
35
|
Lin L, Qin Y, Wu H, Chen Y, Wu S, Si X, Wang H, Wang T, Zhong X, Zhai X, Tong L, Pan B, Zhang F, Zhong Z, Wang Y, Zhao W. Pyrrolidine dithiocarbamate inhibits enterovirus 71 replication by down-regulating ubiquitin-proteasome system. Virus Res 2014; 195:207-16. [PMID: 25456405 DOI: 10.1016/j.virusres.2014.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/25/2014] [Accepted: 10/10/2014] [Indexed: 12/22/2022]
Abstract
Enterovirus 71 (EV71) is the main causative pathogen of hand, foot, and mouth disease (HFMD). The severe neurological complications caused by EV71 infection and the lack of effective therapeutic medicine underline the importance of searching for antiviral substances. Pyrrolidine dithiocarbamate (PDTC), an antioxidant, has been reported to inhibit the replication of coxsackievirus B (CVB) through dysregulating ubiquitin-proteasome system (UPS). In this study, we demonstrated that PDTC exerted potent antiviral effect on EV71. Viral RNA synthesis, viral protein expression, and the production of viral progeny were significantly reduced by the treatment of PDTC in Vero cells infected with EV71. Similar to the previous report about the inhibitory effect of PDTC on UPS, we found that PDTC treatment led to decreased levels of polyubiquitinated proteins in EV71-infected cells. The inhibitory effect of PDTC on UPS was further confirmed by the increased accumulation of cell cycle regulatory proteins p21 and p53, which are normally degraded through UPS, while the expression levels of both proteins remained unchanged. We also showed that PDTC had no impact on the activity of proteasome. Thus, we demonstrated that the down-regulation of PDTC on UPS was the result of its inhibition on ubiquitination. More importantly, this study provides evidence that the inhibition on UPS was required for the antiviral activity of PDTC, since MG132, a potent proteasome inhibitor, significantly inhibited the cytopathic effect and viral protein synthesis in EV71-infected cells. We also found that the antioxidant property of PDTC did not contribute to its antiviral effect, since N-acetyl-l-cysteine, a potent antioxidant, could not inhibit viral replication. In addition, CPE and viral protein synthesis were not inhibited in the cells pretreated with PDTC 2h before viral infection and then cultured in the media with no PDTC supplement, while the antioxidant effect of PDTC was retained. PDTC also showed significant inhibition on apoptosis induced by EV71 infection when it was applied at the early stage of viral infection. Our results collectively suggest that PDTC could be a potential anti-EV71 compound which possesses both antiviral and anti-apoptotic capacity.
Collapse
Affiliation(s)
- Lexun Lin
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Ying Qin
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Heng Wu
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Yang Chen
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Shuo Wu
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Xiaoning Si
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Hui Wang
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Tianying Wang
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Xiaoyan Zhong
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Xia Zhai
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Lei Tong
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Bo Pan
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Zhaohua Zhong
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China
| | - Yan Wang
- Department of Microbiology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China.
| | - Wenran Zhao
- Department of Cell Biology, Harbin Medical University, 196 Xuefu Road, 150086 Harbin, China.
| |
Collapse
|
36
|
Vlahos R, Selemidis S. NADPH Oxidases as Novel Pharmacologic Targets against Influenza A Virus Infection. Mol Pharmacol 2014; 86:747-59. [DOI: 10.1124/mol.114.095216] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
37
|
To EE, Broughton BRS, Hendricks KS, Vlahos R, Selemidis S. Influenza A virus and TLR7 activation potentiate NOX2 oxidase-dependent ROS production in macrophages. Free Radic Res 2014; 48:940-7. [DOI: 10.3109/10715762.2014.927579] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|