1
|
Adams TJ, Schuliga M, Pearce N, Bartlett NW, Liang M. Targeting respiratory virus-induced reactive oxygen species in airways diseases. Eur Respir Rev 2025; 34:240169. [PMID: 40240057 PMCID: PMC12000908 DOI: 10.1183/16000617.0169-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/02/2025] [Indexed: 04/18/2025] Open
Abstract
The immune response to virus infection in the respiratory tract must be carefully balanced to achieve pathogen clearance without excessive immunopathology. For chronic respiratory diseases where there is ongoing inflammation, such as in asthma and COPD, airway immune balance is perturbed, and viral infection frequently worsens (exacerbates) these conditions. Reactive oxygen species (ROS) are critical to the induction and propagation of inflammation, and when appropriately regulated, ROS are vital cell signalling molecules and contribute to innate immunity. However, extended periods of high ROS concentration can cause excessive cellular damage that dysregulates antiviral immunity and promotes inflammation. Traditional antioxidant therapeutics have had limited success treating inflammatory diseases such as viral exacerbations of asthma or COPD, owing to nonspecific pharmacology and poorly understood pharmacokinetic properties. These drawbacks could be addressed with novel drug delivery technologies and pharmacological agents. This review summarises current research on ROS imbalances during virus infection, discusses the commercially available mitochondrial antioxidant drugs that have progressed to clinical trial and assesses novel drug delivery approaches for antioxidant delivery to the airways. Additionally, it provides a perspective on future research into pharmacological targeting of ROS for the treatment of respiratory virus infection and disease.
Collapse
Affiliation(s)
- Thomas J Adams
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Nyoaki Pearce
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Nathan W Bartlett
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| |
Collapse
|
2
|
Zheng Y, Zhang Y, Chen Y, Deng X, Liu B, Xu Q, Qian C, Zhang Z, Wang K, Zeng Y, Liang Z, Sang L, Nong L, Liu X, Xu Y, Li Y, Huang Y. Indoleamine 2,3-dioxygenase 1 drives epithelial cells ferroptosis in influenza-induced acute lung injury. Redox Biol 2025; 81:103572. [PMID: 40023977 PMCID: PMC11915170 DOI: 10.1016/j.redox.2025.103572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening complication of influenza A virus (IAV) infection, characterized by high morbidity and mortality. Recent studies have implicated ferroptosis, a distinct form of regulated cell death characterized by iron-dependent lipid peroxidation, in the pathogenesis of IAV-induced ALI. However, the underlying mechanisms and key regulators of IAV-induced ferroptosis remain largely unknown. In this study, we found that IAV infection induces predominant ferroptosis in alveolar and bronchial epithelial cells, contributing to tissue damage and the development of acute lung injury. Treatment with the ferroptosis inhibitor ferrostatin-1 improved survival, mitigated weight loss, and alleviated lung injury in IAV-infected mice. Mechanistically, IAV-induced ferroptosis was associated with excess lipid peroxidation, nitrative stress, and disrupted iron metabolism. Targeted lipidomic analysis revealed that phospholipid peroxidation is a crucial mechanism in IAV-induced ferroptosis. Importantly, we identified indoleamine 2,3-dioxygenase 1 (IDO1) as a key regulator of IAV-induced ferroptosis. IDO1 knockdown inhibited IAV-induced cell death, and reduced intracellular reactive oxygen species, peroxynitrite, and inducible nitric oxide synthase expression. Furthermore, pharmacological inhibition of IDO1 with 1-methyl-tryptophan improved ALI phenotype in IAV-infected mice. These findings highlight the critical role of ferroptosis in IAV-induced ALI pathogenesis and identify IDO1 as a potential therapeutic target for the treatment of this life-threatening condition.
Collapse
Affiliation(s)
- Yongxin Zheng
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yu Zhang
- Department of Critical Care Medicine, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiumei Deng
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Baiyun Liu
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qiang Xu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Chuyun Qian
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511500, China
| | - Zhihui Zhang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ke Wang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yuan Zeng
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Zhenting Liang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ling Sang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Lingbo Nong
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yimin Li
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| | - Yongbo Huang
- State Key Laboratory of Respiratory Disease, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
3
|
Elajaili HB, Dee NM, Dikalov SI, Kao JPY, Nozik ES. Use of Electron Paramagnetic Resonance (EPR) to Evaluate Redox Status in a Preclinical Model of Acute Lung Injury. Mol Imaging Biol 2024; 26:495-502. [PMID: 37193807 PMCID: PMC10188229 DOI: 10.1007/s11307-023-01826-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023]
Abstract
PURPOSE Patients with hyper- vs. hypo-inflammatory subphenotypes of acute respiratory distress syndrome (ARDS) exhibit different clinical outcomes. Inflammation increases the production of reactive oxygen species (ROS) and increased ROS contributes to the severity of illness. Our long-term goal is to develop electron paramagnetic resonance (EPR) imaging of lungs in vivo to precisely measure superoxide production in ARDS in real time. As a first step, this requires the development of in vivo EPR methods for quantifying superoxide generation in the lung during injury, and testing if such superoxide measurements can differentiate between susceptible and protected mouse strains. PROCEDURES In WT mice, mice lacking total body extracellular superoxide dismutase (EC-SOD) (KO), or mice overexpressing lung EC-SOD (Tg), lung injury was induced with intraperitoneal (IP) lipopolysaccharide (LPS) (10 mg/kg). At 24 h after LPS treatment, mice were injected with the cyclic hydroxylamines 1-hydroxy-3-carboxy-2,2,5,5-tetramethylpyrrolidine hydrochloride (CPH) or 4-acetoxymethoxycarbonyl-1-hydroxy-2,2,5,5-tetramethylpyrrolidine-3-carboxylic acid (DCP-AM-H) probes to detect, respectively, cellular and mitochondrial ROS - specifically superoxide. Several probe delivery strategies were tested. Lung tissue was collected up to one hour after probe administration and assayed by EPR. RESULTS As measured by X-band EPR, cellular and mitochondrial superoxide increased in the lungs of LPS-treated mice compared to control. Lung cellular superoxide was increased in EC-SOD KO mice and decreased in EC-SOD Tg mice compared to WT. We also validated an intratracheal (IT) delivery method, which enhanced the lung signal for both spin probes compared to IP administration. CONCLUSIONS We have developed protocols for delivering EPR spin probes in vivo, allowing detection of cellular and mitochondrial superoxide in lung injury by EPR. Superoxide measurements by EPR could differentiate mice with and without lung injury, as well as mouse strains with different disease susceptibilities. We expect these protocols to capture real-time superoxide production and enable evaluation of lung EPR imaging as a potential clinical tool for subphenotyping ARDS patients based on redox status.
Collapse
Affiliation(s)
- Hanan B Elajaili
- Pediatric Critical Care Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., B131, Aurora, CO, 80045, USA
| | - Nathan M Dee
- Pediatric Critical Care Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., B131, Aurora, CO, 80045, USA
| | - Sergey I Dikalov
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eva S Nozik
- Pediatric Critical Care Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., B131, Aurora, CO, 80045, USA.
| |
Collapse
|
4
|
Kleinehr J, Schöfbänker M, Daniel K, Günl F, Mohamed FF, Janowski J, Brunotte L, Boergeling Y, Liebmann M, Behrens M, Gerdemann A, Klotz L, Esselen M, Humpf HU, Ludwig S, Hrincius ER. Glycolytic interference blocks influenza A virus propagation by impairing viral polymerase-driven synthesis of genomic vRNA. PLoS Pathog 2023; 19:e1010986. [PMID: 37440521 DOI: 10.1371/journal.ppat.1010986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 06/10/2023] [Indexed: 07/15/2023] Open
Abstract
Influenza A virus (IAV), like any other virus, provokes considerable modifications of its host cell's metabolism. This includes a substantial increase in the uptake as well as the metabolization of glucose. Although it is known for quite some time that suppression of glucose metabolism restricts virus replication, the exact molecular impact on the viral life cycle remained enigmatic so far. Using 2-deoxy-d-glucose (2-DG) we examined how well inhibition of glycolysis is tolerated by host cells and which step of the IAV life cycle is affected. We observed that effects induced by 2-DG are reversible and that cells can cope with relatively high concentrations of the inhibitor by compensating the loss of glycolytic activity by upregulating other metabolic pathways. Moreover, mass spectrometry data provided information on various metabolic modifications induced by either the virus or agents interfering with glycolysis. In the presence of 2-DG viral titers were significantly reduced in a dose-dependent manner. The supplementation of direct or indirect glycolysis metabolites led to a partial or almost complete reversion of the inhibitory effect of 2-DG on viral growth and demonstrated that indeed the inhibition of glycolysis and not of N-linked glycosylation was responsible for the observed phenotype. Importantly, we could show via conventional and strand-specific qPCR that the treatment with 2-DG led to a prolonged phase of viral mRNA synthesis while the accumulation of genomic vRNA was strongly reduced. At the same time, minigenome assays showed no signs of a general reduction of replicative capacity of the viral polymerase. Therefore, our data suggest that the significant reduction in IAV replication by glycolytic interference occurs mainly due to an impairment of the dynamic regulation of the viral polymerase which conveys the transition of the enzyme's function from transcription to replication.
Collapse
Affiliation(s)
- Jens Kleinehr
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Michael Schöfbänker
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Katharina Daniel
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Franziska Günl
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Fakry Fahmy Mohamed
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, Sharkia, Egypt
| | - Josua Janowski
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Linda Brunotte
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Yvonne Boergeling
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Marie Liebmann
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany
| | - Matthias Behrens
- Institute of Food Chemistry, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Andrea Gerdemann
- Institute of Food Chemistry, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Muenster, Germany
| | - Melanie Esselen
- Institute of Food Chemistry, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Stephan Ludwig
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Eike R Hrincius
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Muenster, Germany
| |
Collapse
|
5
|
Potential Role of Superoxide Dismutase 3 (SOD3) in Resistance to Influenza A Virus Infection. Antioxidants (Basel) 2023; 12:antiox12020354. [PMID: 36829913 PMCID: PMC9952479 DOI: 10.3390/antiox12020354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Influenza A virus infection induces the production of excessive reactive oxygen species (ROS). Overproduction of ROS can overwhelm the antioxidant defense system, leading to increasing intensive oxidative stress. However, antioxidant defense against oxidative damage induced by influenza A virus infection, and in particular the significance of the SOD3 response in the pathogenesis of influenza virus infection, has not been well characterized. Here, we investigated the potential role of SOD3 in resistance to influenza A virus infection. In this study, SOD3, as an important antioxidant enzyme, was shown to be highly elevated in A549 cells following influenza A virus infection. Furthermore, inhibition of SOD3 impacted viral replication and virulence. We found that SOD3 disrupts IAV replication by impairing the synthesis of vRNA, whereas it did not affect viral ribonucleoprotein nuclear export. In addition, overexpression of SOD3 greatly reduced the levels of ROS caused by influenza A virus infection, regulated the inflammatory response to virus infection by inhibiting the phosphorylation of p65 of the NF-κB signaling pathway, and inhibited virus-induced apoptosis to a certain extent. Taken together, these findings indicate that SOD3 is actively involved in influenza A virus replication. Pharmacological modulation or targeting of SOD3 may pave the way for a novel therapeutic approach to combating influenza A virus infection.
Collapse
|
6
|
Natural Bioactive Compounds Targeting NADPH Oxidase Pathway in Cardiovascular Diseases. Molecules 2023; 28:molecules28031047. [PMID: 36770715 PMCID: PMC9921542 DOI: 10.3390/molecules28031047] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/04/2022] [Accepted: 12/10/2022] [Indexed: 01/21/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, in both developed and developing countries. According to the WHO report, the morbidity and mortality caused by CVD will continue to rise with the estimation of death going up to 22.2 million in 2030. NADPH oxidase (NOX)-derived reactive oxygen species (ROS) production induces endothelial nitric oxide synthase (eNOS) uncoupling and mitochondrial dysfunction, resulting in sustained oxidative stress and the development of cardiovascular diseases. Seven distinct members of the family have been identified of which four (namely, NOX1, 2, 4 and 5) may have cardiovascular functions. Currently, the treatment and management plan for patients with CVDs mainly depends on the drugs. However, prolonged use of prescribed drugs may cause adverse drug reactions. Therefore, it is crucial to find alternative treatment options with lesser adverse effects. Natural products have been gaining interest as complementary therapy for CVDs over the past decade due to their wide range of medicinal properties, including antioxidants. These might be due to their potent active ingredients, such as flavonoid and phenolic compounds. Numerous natural compounds have been demonstrated to have advantageous effects on cardiovascular disease via NADPH cascade. This review highlights the potential of natural products targeting NOX-derived ROS generation in treating CVDs. Emphasis is put on the activation of the oxidases, including upstream or downstream signalling events.
Collapse
|
7
|
Dulin H, Hendricks N, Xu D, Gao L, Wuang K, Ai H, Hai R. Impact of Protein Nitration on Influenza Virus Infectivity and Immunogenicity. Microbiol Spectr 2022; 10:e0190222. [PMID: 36314966 PMCID: PMC9769652 DOI: 10.1128/spectrum.01902-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/26/2022] [Indexed: 11/06/2022] Open
Abstract
Influenza viruses are deadly respiratory pathogens of special importance due to their long history of global pandemics. During influenza virus infections, the host responds by producing interferons, which activate interferon-stimulated genes (ISGs) inside target cells. One of these ISGs is inducible nitric oxide synthase (iNOS). iNOS produces nitric oxide (NO) from arginine and molecular oxygen inside the cell. NO can react with superoxide radicals to form reactive nitrogen species, principally peroxynitrite. While much work has been done studying the many roles of nitric oxide in influenza virus infections, the direct effect of peroxynitrite on influenza virus proteins has not been determined. Manipulations of NO, either by knocking out iNOS or chemically inhibiting NO, produced no change in virus titers in mouse models of influenza infection. However, peroxynitrite has a known antimicrobial effect on various bacteria and parasites, and the reason for its lack of antimicrobial effect on influenza virus titers in vivo remains unclear. Therefore, we wished to test the direct effect of nitration of influenza virus proteins. We examined the impact of nitration on virus infectivity, replication, and immunogenicity. We observed that the nitration of influenza A virus proteins decreased virus infectivity and replication ex vivo. We also determined that the nitration of influenza virus hemagglutinin protein can reduce antibody responses to native virus protein. However, our study also suggests that nitration of influenza virus proteins in vivo is likely not extensive enough to inhibit virus functions substantially. These findings will help clarify the role of peroxynitrite during influenza virus infections. IMPORTANCE Nitric oxide and peroxynitrite produced during microbial infections have diverse and seemingly paradoxical functions. While nitration of lung tissue during influenza virus infection has been observed in both mice and humans, the direct effect of protein nitration on influenza viruses has remained elusive. We addressed the impact of nitration of influenza virus proteins on virus infectivity, replication, and immunogenicity. We observed that ex vivo nitration of influenza virus proteins reduced virus infectivity and immunogenicity. However, we did not detect nitration of influenza virus hemagglutinin protein in vivo. These results contribute to our understanding of the roles of nitric oxide and peroxynitrite in influenza virus infections.
Collapse
Affiliation(s)
- Harrison Dulin
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, Riverside, California, USA
- Microbiology and Plant Pathology, University of California, Riverside, Riverside, California, USA
| | - Nathan Hendricks
- Proteomics Core, University of California, Riverside, Riverside, California, USA
| | - Duo Xu
- Microbiology and Plant Pathology, University of California, Riverside, Riverside, California, USA
| | - Linfeng Gao
- Microbiology and Plant Pathology, University of California, Riverside, Riverside, California, USA
| | - Keidy Wuang
- Microbiology and Plant Pathology, University of California, Riverside, Riverside, California, USA
| | - Huiwang Ai
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Rong Hai
- Cell, Molecular, and Developmental Biology Graduate Program, University of California, Riverside, Riverside, California, USA
- Microbiology and Plant Pathology, University of California, Riverside, Riverside, California, USA
| |
Collapse
|
8
|
Hong KS, Pagan K, Whalen W, Harris R, Yang J, Stout-Delgado H, Cho SJ. The Role of Glutathione Reductase in Influenza Infection. Am J Respir Cell Mol Biol 2022; 67:438-445. [PMID: 35767671 PMCID: PMC9753556 DOI: 10.1165/rcmb.2021-0372oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 06/29/2022] [Indexed: 02/05/2023] Open
Abstract
Influenza infection induces lung epithelial cell injury via programmed cell death. Glutathione, a potent antioxidant, has been reported to be associated with influenza infection. We hypothesized that lung epithelial cell death during influenza infection is regulated by glutathione metabolism. Eight-week-old male and female BALB/c mice were infected with influenza (PR8: A/PR/8/34 [H1N1]) via intranasal instillation. Metabolomic analyses were performed on whole lung lysate after influenza infection. For in vitro analysis, Beas-2B cells were infected with influenza. RNA was extracted, and QuantiTect Primer Assay was used to assess gene expression. Glutathione concentrations were assessed by colorimetric assay. Influenza infection resulted in increased inflammation and epithelial cell injury in our murine model, leading to increased morbidity and mortality. In both our in vivo and in vitro models, influenza infection was found to induce apoptosis and necroptosis. Influenza infection led to decreased glutathione metabolism and reduced glutathione reductase activity in lung epithelial cells. Genetic inhibition of glutathione reductase suppressed apoptosis and necroptosis of lung epithelial cells. Pharmacologic inhibition of glutathione reductase reduced airway inflammation, lung injury, and cell death in our murine influenza model. Our results demonstrate that glutathione reductase activity is suppressed during influenza. Glutathione reductase inhibition prevents epithelial cell death and morbidity in our murine influenza model. Our results suggest that glutathione reductase-dependent glutathione metabolism may play an important role in the host response to viral infection by regulating lung epithelial cell death.
Collapse
Affiliation(s)
- Kyung Sook Hong
- Division of Critical Care Medicine, Department of Surgery, Ewha Womans University College of Medicine, Seoul, South Korea; and
| | - Kassandra Pagan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - William Whalen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Rebecca Harris
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jianjun Yang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Heather Stout-Delgado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Soo Jung Cho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
9
|
Shchukin IA, Fidler MS, Koltsov IA, Suvorov AY. COVID-19-Associated Stroke. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 52:649-656. [PMID: 36119649 PMCID: PMC9468522 DOI: 10.1007/s11055-022-01291-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/22/2021] [Indexed: 11/25/2022]
Abstract
The COVID-19 pandemic has had significant influences on the incidence of acute cerebrovascular accidents and the structure of mortality. SARS-CoV-2 increases the risks of developing both ischemic and hemorrhagic stroke. The key pathogenetic element underlying the development of cerebral stroke in COVID-19 consists of impairments to the operation of angiotensin 2 receptors, which are accompanied by accumulation of excess quantities of angiotensin 2, endothelial dysfunction, hypercoagulation, overproduction of proinflammatory cytokines, and an oxidative storm. In patients with stroke and COVID-19, lesion severity is associated with dual mechanisms of ischemia - systemic and cerebral. The possibilities of medication-based correction of both systemic impairments associated with coronavirus infection and local impairments due to ischemic or hemorrhagic brain damage, are limited. Substances with antioxidant activity may potentially be effective in patients with stroke and COVID-19. Data from a number of clinical rials indicate that Mexidol significantly improves functional outcomes in ischemic stroke. Use of Mexidol in patients with stroke and COVID-19 is advised.
Collapse
Affiliation(s)
- I. A. Shchukin
- Pirogov Russian National Research Medical University, Russian Ministry of Health, Moscow, Russia
| | - M. S. Fidler
- Pirogov Russian National Research Medical University, Russian Ministry of Health, Moscow, Russia
| | - I. A. Koltsov
- Pirogov Russian National Research Medical University, Russian Ministry of Health, Moscow, Russia
| | - A. Yu. Suvorov
- Federal Center for the Brain and Neurotechnology, Federal Medical Biological Agency of Russia, Moscow, Russia
| |
Collapse
|
10
|
Yang J, Zhang R, Zhao H, Qi H, Li J, Li J, Zhou X, Wang A, Fan K, Yan X, Zhang T. Bioinspired copper single-atom nanozyme as a superoxide dismutase-like antioxidant for sepsis treatment. EXPLORATION (BEIJING, CHINA) 2022; 2:20210267. [PMID: 37325607 PMCID: PMC10191017 DOI: 10.1002/exp.20210267] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Sepsis is a systemic inflammatory response syndrome with high morbidity and mortality mediated by infection-caused oxidative stress. Early antioxidant intervention by removing excessively produced reactive oxygen and nitrogen species (RONS) is beneficial to the prevention and treatment of sepsis. However, traditional antioxidants have failed to improve patient outcomes due to insufficient activity and sustainability. Herein, by mimicking the electronic and structural characteristics of natural Cu-only superoxide dismutase (SOD5), a single-atom nanozyme (SAzyme) featuring coordinately unsaturated and atomically dispersed Cu-N4 site was synthesized for effective sepsis treatment. The de novo-designed Cu-SAzyme exhibits a superior SOD-like activity to efficiently eliminate O2 •-, which is the source of multiple RONS, thus blocking the free radical chain reaction and subsequent inflammatory response in the early stage of sepsis. Moreover, the Cu-SAzyme effectively harnessed systemic inflammation and multi-organ injuries in sepsis animal models. These findings indicate that the developed Cu-SAzyme possesses great potential as therapeutic nanomedicines for the treatment of sepsis.
Collapse
Affiliation(s)
- Ji Yang
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Ruofei Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Hanqing Zhao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Haifeng Qi
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Jingyun Li
- Key Laboratory of Infection and ImmunityInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Jian‐Feng Li
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
| | - Xinyao Zhou
- School of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Aiqin Wang
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
- Nanozyme Medical Center, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
- Nanozyme Medical Center, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Tao Zhang
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| |
Collapse
|
11
|
Kumova OK, Galani IE, Rao A, Johnson H, Triantafyllia V, Matt SM, Pascasio J, Gaskill PJ, Andreakos E, Katsikis PD, Carey AJ. Severity of neonatal influenza infection is driven by type I interferon and oxidative stress. Mucosal Immunol 2022; 15:1309-1320. [PMID: 36352099 PMCID: PMC9724789 DOI: 10.1038/s41385-022-00576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 09/26/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
Neonates exhibit increased susceptibility to respiratory viral infections, attributed to inflammation at the developing pulmonary air-blood interface. IFN I are antiviral cytokines critical to control viral replication, but also promote inflammation. Previously, we established a neonatal murine influenza virus (IV) model, which demonstrates increased mortality. Here, we sought to determine the role of IFN I in this increased mortality. We found that three-day-old IFNAR-deficient mice are highly protected from IV-induced mortality. In addition, exposure to IFNβ 24 h post IV infection accelerated death in WT neonatal animals but did not impact adult mortality. In contrast, IFN IIIs are protective to neonatal mice. IFNβ induced an oxidative stress imbalance specifically in primary neonatal IV-infected pulmonary type II epithelial cells (TIIEC), not in adult TIIECs. Moreover, neonates did not have an infection-induced increase in antioxidants, including a key antioxidant, superoxide dismutase 3, as compared to adults. Importantly, antioxidant treatment rescued IV-infected neonatal mice, but had no impact on adult morbidity. We propose that IFN I exacerbate an oxidative stress imbalance in the neonate because of IFN I-induced pulmonary TIIEC ROS production coupled with developmentally regulated, defective antioxidant production in response to IV infection. This age-specific imbalance contributes to mortality after respiratory infections in this vulnerable population.
Collapse
Affiliation(s)
- Ogan K. Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ioanna-Evdokia Galani
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Abhishek Rao
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Hannah Johnson
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Stephanie M. Matt
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Judy Pascasio
- Pathology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Peter J. Gaskill
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Peter D. Katsikis
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alison J. Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States.,Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
12
|
Viral PB1-F2 and host IFN-γ guide ILC2 and T cell activity during influenza virus infection. Proc Natl Acad Sci U S A 2022; 119:2118535119. [PMID: 35169077 PMCID: PMC8872759 DOI: 10.1073/pnas.2118535119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2022] [Indexed: 12/28/2022] Open
Abstract
The regulation of functional immune cell plasticity is poorly understood. Host environmental cues are critical, but the possible influence of pathogen-derived virulence factors has not been described. We have used reverse-engineered influenza A viruses that differ in PB1-F2 activity to analyze influenza in mice in the presence or absence of host interferon (IFN)-γ. In the absence of functional PB1-F2 and IFN-γ, lung ILC2s initiated robust IL-5 responses following viral challenge, which led to improved tissue integrity and survival. Conversely, functional PB1-F2 suppressed IL-5+ ILC2 responses and induced a dominant IL-13+ CD8 T cell response regardless of host IFN-γ. These findings demonstrate the critical interplay between the viral virulence factors and host cytokines in regulating protective pulmonary immunity during influenza virus infection. Functional plasticity of innate lymphoid cells (ILCs) and T cells is regulated by host environmental cues, but the influence of pathogen-derived virulence factors has not been described. We now report the interplay between host interferon (IFN)-γ and viral PB1-F2 virulence protein in regulating the functions of ILC2s and T cells that lead to recovery from influenza virus infection of mice. In the absence of IFN-γ, lung ILC2s from mice challenged with the A/California/04/2009 (CA04) H1N1 virus, containing nonfunctional viral PB1-F2, initiated a robust IL-5 response, which also led to improved tissue integrity and increased survival. Conversely, challenge with Puerto Rico/8/1934 (PR8) H1N1 virus expressing fully functional PB1-F2, suppressed IL-5+ ILC2 responses, and induced a dominant IL-13+ CD8 T cell response, regardless of host IFN-γ expression. IFN-γ–deficient mice had increased survival and improved tissue integrity following challenge with lethal doses of CA04, but not PR8 virus, and increased resistance was dependent on the presence of IFN-γR+ ILC2s. Reverse-engineered influenza viruses differing in functional PB1-F2 activity induced ILC2 and T cell phenotypes similar to the PB1-F2 donor strains, demonstrating the potent role of viral PB1-F2 in host resistance. These results show the ability of a pathogen virulence factor together with host IFN-γ to regulate protective pulmonary immunity during influenza infection.
Collapse
|
13
|
Liu J, Dean DA. Gene Therapy for Acute Respiratory Distress Syndrome. Front Physiol 2022; 12:786255. [PMID: 35111077 PMCID: PMC8801611 DOI: 10.3389/fphys.2021.786255] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating clinical syndrome that leads to acute respiratory failure and accounts for over 70,000 deaths per year in the United States alone, even prior to the COVID-19 pandemic. While its molecular details have been teased apart and its pathophysiology largely established over the past 30 years, relatively few pharmacological advances in treatment have been made based on this knowledge. Indeed, mortality remains very close to what it was 30 years ago. As an alternative to traditional pharmacological approaches, gene therapy offers a highly controlled and targeted strategy to treat the disease at the molecular level. Although there is no single gene or combination of genes responsible for ARDS, there are a number of genes that can be targeted for upregulation or downregulation that could alleviate many of the symptoms and address the underlying mechanisms of this syndrome. This review will focus on the pathophysiology of ARDS and how gene therapy has been used for prevention and treatment. Strategies for gene delivery to the lung, such as barriers encountered during gene transfer, specific classes of genes that have been targeted, and the outcomes of these approaches on ARDS pathogenesis and resolution will be discussed.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| | - David A. Dean
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
14
|
Effect of Reactive Oxygen Species on the Endoplasmic Reticulum and Mitochondria during Intracellular Pathogen Infection of Mammalian Cells. Antioxidants (Basel) 2021; 10:antiox10060872. [PMID: 34071633 PMCID: PMC8229183 DOI: 10.3390/antiox10060872] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress, particularly reactive oxygen species (ROS), are important for innate immunity against pathogens. ROS directly attack pathogens, regulate and amplify immune signals, induce autophagy and activate inflammation. In addition, production of ROS by pathogens affects the endoplasmic reticulum (ER) and mitochondria, leading to cell death. However, it is unclear how ROS regulate host defense mechanisms. This review outlines the role of ROS during intracellular pathogen infection, mechanisms of ROS production and regulation of host defense mechanisms by ROS. Finally, the interaction between microbial pathogen-induced ROS and the ER and mitochondria is described.
Collapse
|
15
|
Intracellular Redox-Modulated Pathways as Targets for Effective Approaches in the Treatment of Viral Infection. Int J Mol Sci 2021; 22:ijms22073603. [PMID: 33808471 PMCID: PMC8036776 DOI: 10.3390/ijms22073603] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Host-directed therapy using drugs that target cellular pathways required for virus lifecycle or its clearance might represent an effective approach for treating infectious diseases. Changes in redox homeostasis, including intracellular glutathione (GSH) depletion, are one of the key events that favor virus replication and contribute to the pathogenesis of virus-induced disease. Redox homeostasis has an important role in maintaining an appropriate Th1/Th2 balance, which is necessary to mount an effective immune response against viral infection and to avoid excessive inflammatory responses. It is known that excessive production of reactive oxygen species (ROS) induced by viral infection activates nuclear factor (NF)-kB, which orchestrates the expression of viral and host genes involved in the viral replication and inflammatory response. Moreover, redox-regulated protein disulfide isomerase (PDI) chaperones have an essential role in catalyzing formation of disulfide bonds in viral proteins. This review aims at describing the role of GSH in modulating redox sensitive pathways, in particular that mediated by NF-kB, and PDI activity. The second part of the review discusses the effectiveness of GSH-boosting molecules as broad-spectrum antivirals acting in a multifaceted way that includes the modulation of immune and inflammatory responses.
Collapse
|
16
|
Shchukin I, Fidler M, Koltsov I, Suvorov A. COVID-19 related stroke. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:69-76. [DOI: 10.17116/jnevro202112112269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
17
|
Yee M, David Cohen E, Haak J, Dylag AM, O'Reilly MA. Neonatal hyperoxia enhances age-dependent expression of SARS-CoV-2 receptors in mice. Sci Rep 2020; 10:22401. [PMID: 33372179 PMCID: PMC7769981 DOI: 10.1038/s41598-020-79595-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/06/2020] [Indexed: 12/28/2022] Open
Abstract
The severity of COVID-19 lung disease is higher in the elderly and people with pre-existing co-morbidities. People who were born preterm may be at greater risk for COVID-19 because their early exposure to oxygen (hyperoxia) at birth increases the severity of respiratory viral infections. Hyperoxia at birth increases the severity of influenza A virus infections in adult mice by reducing the number of alveolar epithelial type 2 (AT2) cells. Since AT2 cells express the SARS-CoV-2 receptors angiotensin converting enzyme (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2), their expression should decline as AT2 cells are depleted by hyperoxia. Instead, ACE2 was detected in airway Club cells and endothelial cells at birth, and then AT2 cells at one year of age. Neonatal hyperoxia stimulated expression of ACE2 in Club cells and in AT2 cells by 2 months of age. It also stimulated expression of TMPRSS2 in the lung. Increased expression of SARS-CoV-2 receptors was blocked by mitoTEMPO, a mitochondrial superoxide scavenger that reduced oxidative stress and DNA damage seen in oxygen-exposed mice. Our finding that hyperoxia enhances the age-dependent expression of SARS-CoV-2 receptors in mice helps explain why COVID-19 lung disease is greater in the elderly and people with pre-existing co-morbidities.
Collapse
Affiliation(s)
- Min Yee
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - E David Cohen
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Jeannie Haak
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Andrew M Dylag
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Michael A O'Reilly
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.
| |
Collapse
|
18
|
Tang Y, Li Y, Sun J, Pan H, Yao F, Jiao X. Selection of an Optimal Combination Panel to Better Triage COVID-19 Hospitalized Patients. J Inflamm Res 2020; 13:773-787. [PMID: 33149652 PMCID: PMC7602889 DOI: 10.2147/jir.s273193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE It is difficult to predict the prognosis of COVID-19 patients at the disease onset. This study was designed to add new biomarkers into conventional inflammatory panels to build an optimal combination panel, to better triage patients and predict their outcomes. PATIENTS AND METHODS Biochemical parameters representing multi-organ functions, cytokines, acute-phase proteins, and other inflammatory markers were measured in COVID-19 patients on hospital admission. Receiver operating characteristic (ROC) curves, logistic regression, event-free survival (EFS), and Cox analyses were performed to screen and compare the predictive capabilities of the new panel in patients with different illness severity and outcome. RESULTS This study included 120 patients with COVID-19, consisting of 32 critical, 28 severe, and 60 mild/moderate patients. Initial levels of the selected biomarkers showed a significant difference in the three groups, all of which influenced patient outcome and EFS to varying degrees. Cox proportional hazard model revealed that procalcitonin (PCT) and interleukin 10 (IL-10) were independent risk factors, while superoxide dismutase (SOD) was an independent protective factor influencing EFS. In discriminating the critical and mild patients, a panel combining PCT, IL-6, and neutrophil (NEUT) yielded the best diagnostic performance with an AUC of 0.99, the sensitivity of 90.60% and specificity of 100%. In distinguishing between severe and mild patients, SOD's AUC of 0.89 was higher than any other single biomarker. In differentiating the critical and severe patients, the combination of white blood cell count (WBC), PCT, IL-6, IL-10, and SOD achieved the highest AUC of 0.95 with a sensitivity of 75.00% and specificity of 100%. CONCLUSION The optimal combination panel has a substantial potential to better triage COVID-19 patients on admission. Better triage of patients will benefit the rational use of medical resources.
Collapse
Affiliation(s)
- Yueting Tang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Yirong Li
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Jiayu Sun
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Huaqin Pan
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| |
Collapse
|
19
|
Tackle the free radicals damage in COVID-19. Nitric Oxide 2020; 102:39-41. [PMID: 32562746 PMCID: PMC7837363 DOI: 10.1016/j.niox.2020.06.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/27/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
COVID-19 is a severe pandemic which has caused a devastating amount of loss in lives around the world, and yet we still don't know how to appropriately treat this disease. We know very little about the pathogenesis of SARS-CoV-2, the virus which induces the COVID-19. However, COVID-19 does share many similar symptoms with SARS and influenza. Previous scientific discoveries learned from lab animal models and clinical practices shed light on possible pathogenic mechanisms in COVID-19. In the past decades, accumulated scientific findings confirmed the pathogenic role of free radicals damage in respiratory virus infection. Astonishingly very few medical professionals mention the crucial role of free radical damage in COVID-19. This hypothesis aims to summarize the crucial pathogenic role of free radical damage in respiratory virus induced pneumonia and suggest an antioxidative therapeutic strategy for COVID-19. Additional scheme figure is attached.
Collapse
|
20
|
Bortz E, Wu TT, Patel P, Whitelegge JP, Sun R. Proteomics of Bronchoalveolar Lavage Fluid Reveals a Lung Oxidative Stress Response in Murine Herpesvirus-68 Infection. Viruses 2018; 10:v10120670. [PMID: 30486363 PMCID: PMC6316452 DOI: 10.3390/v10120670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
Murine herpesvirus-68 (MHV-68) productively infects mouse lungs, exhibiting a complex pathology characteristic of both acute viral infections and chronic respiratory diseases. We sought to discover proteins differentially expressed in bronchoalveolar lavage (BAL) from mice infected with MHV-68. Mice were infected intranasally with MHV-68. After nine days, as the lytic phase of infection resolved, differential BAL proteins were identified by two-dimensional (2D) electrophoresis and mass spectrometry. Of 23 unique proteins, acute phase proteins, vitamin A transport, and oxidative stress response factors Pdx6 and EC-SOD (Sod3) were enriched. Correspondingly, iNOS2 was induced in lung tissue by seven days post-infection. Oxidative stress was partly a direct result of MHV-68 infection, as reactive oxygen species (ROS) were induced in cultured murine NIH3T3 fibroblasts and human lung A549 cells infected with MHV-68. Finally, mice infected with a recombinant MHV-68 co-expressing inflammatory cytokine murine interleukin 6 (IL6) showed exacerbated oxidative stress and soluble type I collagen characteristic of tissue recovery. Thus, oxidative stress appears to be a salient feature of MHV-68 pathogenesis, in part caused by lytic replication of the virus and IL6. Proteins and small molecules in lung oxidative stress networks therefore may provide new therapeutic targets to ameliorate respiratory virus infections.
Collapse
Affiliation(s)
- Eric Bortz
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA.
| | - Ting-Ting Wu
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Parthive Patel
- Center for Molecular Biology and German Cancer Research Center (DKFZ), University of Heidelberg (ZMBH), 69120 Heidelberg, Germany.
| | - Julian P Whitelegge
- The Pasarow Mass Spectrometry Laboratory & the Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Ren Sun
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
21
|
Zablockienė B, Kačergius T, Ambrozaitis A, Žurauskas E, Bratchikov M, Jurgauskienė L, Zablockis R, Gravenstein S. Zanamivir Diminishes Lung Damage in Influenza A Virus-infected Mice by Inhibiting Nitric Oxide Production. ACTA ACUST UNITED AC 2018; 32:473-478. [PMID: 29695548 DOI: 10.21873/invivo.11263] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIM Severe pulmonary influenza A virus (IAV) infection causes lung inflammation and expression of inducible nitric oxide synthase (iNOS), leading to overproduction of nitric oxide (NO). We studied whether zanamivir reduces pulmonary inflammation through inhibition of NO production in mice. MATERIALS AND METHODS We treated IAV-infected mice daily with intranasal zanamivir. Controls were infected and either placebo-treated or untreated, or not infected and placebo-treated. Mice were weighed daily. After euthanasia on day 3, lungs were excised and bronchoalveolar lavage was performed and fluid nitrite concentration was determined. Lungs were analyzed microscopically. iNOS and IAV RNA levels in lungs were assessed using quantitative reverse transcription-polymerase chain reaction (RT-qPCR). RESULTS Mice undergoing zanamivir treatment had less weight loss, viral replication, and lung damage, as well as significant reductions of local NO and iNOS mRNA synthesis (p<0.05). CONCLUSION Zanamivir is associated with an anti-inflammatory effect mediated through inhibition of NO production in IAV-infected mice.
Collapse
Affiliation(s)
- Birutė Zablockienė
- Clinic of Infectious and Chest Diseases, Dermatovenerology and Allergology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Tomas Kačergius
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Department of Internal Medicine, Glennan Center for Geriatrics and Gerontology, Eastern Virginia Medical School, Norfolk, VA, U.S.A
| | - Arvydas Ambrozaitis
- Clinic of Infectious and Chest Diseases, Dermatovenerology and Allergology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Edvardas Žurauskas
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Maksim Bratchikov
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Laimutė Jurgauskienė
- Clinic of Cardiovascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rolandas Zablockis
- Clinic of Infectious and Chest Diseases, Dermatovenerology and Allergology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Stefan Gravenstein
- Department of Internal Medicine, Glennan Center for Geriatrics and Gerontology, Eastern Virginia Medical School, Norfolk, VA, U.S.A.,Department of Medicine, Warren Alpert Medical School, School of Public Health, Brown University, and Providence Veterans Administration Hospital, Providence, RI, U.S.A.,Department of Health Services Policy and Practice, School of Public Health, Brown University, and Providence Veterans Administration Hospital, Providence, RI, U.S.A
| |
Collapse
|
22
|
Khomich OA, Kochetkov SN, Bartosch B, Ivanov AV. Redox Biology of Respiratory Viral Infections. Viruses 2018; 10:392. [PMID: 30049972 PMCID: PMC6115776 DOI: 10.3390/v10080392] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
Affiliation(s)
- Olga A Khomich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str, 32, 119991 Moscow, Russia.
- Inserm U1052, Cancer Research Center Lyon, University of Lyon, 69000 Lyon, France.
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str, 32, 119991 Moscow, Russia.
| | - Birke Bartosch
- Inserm U1052, Cancer Research Center Lyon, University of Lyon, 69000 Lyon, France.
- DevWeCan Laboratories of Excellence Network (Labex), 69003 Lyon, France.
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str, 32, 119991 Moscow, Russia.
| |
Collapse
|
23
|
Wang QW, Su Y, Sheng JT, Gu LM, Zhao Y, Chen XX, Chen C, Li WZ, Li KS, Dai JP. Anti-influenza A virus activity of rhein through regulating oxidative stress, TLR4, Akt, MAPK, and NF-κB signal pathways. PLoS One 2018; 13:e0191793. [PMID: 29385192 PMCID: PMC5791991 DOI: 10.1371/journal.pone.0191793] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
Rhein, an anthraquinone compound existing in many traditional herbal medicines, has anti-inflammatory, antioxidant, antitumor, antiviral, hepatoprotective, and nephroprotective activities, but its anti-influenza A virus (IAV) activity is ambiguous. In the present study, through plaque inhibition assay, time-of-addition assay, antioxidant assay, qRT-PCR, ELISA, and western blotting assays, we investigated the anti-IAV effect and mechanism of action of rhein in vitro and in vivo. The results showed that rhein could significantly inhibit IAV adsorption and replication, decrease IAV-induced oxidative stress, activations of TLR4, Akt, p38, JNK MAPK, and NF-κB pathways, and production of inflammatory cytokines and matrix metalloproteinases in vitro. Oxidant H2O2 and agonists of TLR4, Akt, p38/JNK and IKK/NF-κB could significantly antagonize the inhibitory effects of rhein on IAV-induced cytopathic effect (CPE) and IAV replication. Through an in vivo test in mice, we also found that rhein could significantly improve the survival rate, lung index, pulmonary cytokines, and pulmonary histopathological changes. Rhein also significantly decreased pulmonary viral load at a high dose. In conclusion, rhein can inhibit IAV adsorption and replication, and the mechanism of action to inhibit IAV replication may be due to its ability to suppress IAV-induced oxidative stress and activations of TLR4, Akt, p38, JNK MAPK, and NF-κB signal pathways.
Collapse
Affiliation(s)
- Qian-Wen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jiang-Tao Sheng
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Li-Ming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiao-Xuan Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Cheng Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Wei-Zhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jian-Ping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
24
|
Pavlova EL, Simeonova LS, Gegova GA. Combined efficacy of oseltamivir, isoprinosine and ellagic acid in influenza A(H3N2)-infected mice. Biomed Pharmacother 2017; 98:29-35. [PMID: 29241072 DOI: 10.1016/j.biopha.2017.12.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 11/25/2022] Open
Abstract
Influenza pathogenesis comprises a complex cascade of impaired cellular processes resulting from the viral replication and exaggerated immune response accompanied by reactive oxygen species (ROS) burst and oxidative stress, destructing membranous structures and tissues. By classical virological and biochemical methods we compared and evaluated the therapeutic effects of 2.5mg/kg/day of the antiviral drug - oseltamivir (OS), 500mg/kg/day of the immune modulator - isoprinosine (IP) and 500mg/kg/day of the antioxidant agent ellagic acid (EA) with a focus on their combined activities in influenza H3N2 virus-infected mice. The survival, lung pathology and titers, as well as the oxidative stress biomarker thiobarbituric acid reactive substances (TBARS) in the lungs, liver and blood plasma, correlated to the activities of the antioxidant enzymes superoxide dismutase (SOD) and glutathione reductase (GR) were assessed. We found that the viral inhibitor applied together with the immune modulator and the antioxidant exhibited strong therapeutic effects on the survival of the influenza-challenged mice. That effect was mostly pronounced for the triple combination - protection index (PI) of 75.2%, mean survival time (MST) extended by 5.8 days compared to the PBS control and significant reduction of the lung titers by 1.38 Δlg; 2.3 scores lower lung pathology and 8 times reduction of the accumulated TBARS in the lungs and liver on the 5-th day p.i. The enzymatic assays revealed that this combination demonstrated very good protection against the damaging superoxide radicals (83% efficiency of SOD, in comparison to healthy controls 100%). The double combinations of OS with IP and EA also showed protective effects according to the virological analysis - PI of 53.1% and 54.5%. Ten times higher GR activity was observed when the combination EA+OS and monotherapy of EA were applied (96% in comparison to healthy controls 100%). The best antioxidant effect in blood plasma was observed in the EA+IP group - 4 times reduction in the TBARS-content compared to infected controls but it did not have any efficacy on the survival and lung injury.
Collapse
Affiliation(s)
- Elitsa L Pavlova
- Biophysics & Medical Physics, Sofia University "St. Kliment Ohridski", 5 James Boucher Blvd., 1164, Sofia, Bulgaria.
| | - Lora S Simeonova
- Department of Virology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Georgi Bonchev Str., 1113, Sofia, Bulgaria
| | - Galina A Gegova
- Department of Virology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Georgi Bonchev Str., 1113, Sofia, Bulgaria
| |
Collapse
|
25
|
Camp JV, Jonsson CB. A Role for Neutrophils in Viral Respiratory Disease. Front Immunol 2017; 8:550. [PMID: 28553293 PMCID: PMC5427094 DOI: 10.3389/fimmu.2017.00550] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 04/24/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are immune cells that are well known to be present during many types of lung diseases associated with acute respiratory distress syndrome (ARDS) and may contribute to acute lung injury. Neutrophils are poorly studied with respect to viral infection, and specifically to respiratory viral disease. Influenza A virus (IAV) infection is the cause of a respiratory disease that poses a significant global public health concern. Influenza disease presents as a relatively mild and self-limiting although highly pathogenic forms exist. Neutrophils increase in the respiratory tract during infection with mild seasonal IAV, moderate and severe epidemic IAV infection, and emerging highly pathogenic avian influenza (HPAI). During severe influenza pneumonia and HPAI infection, the number of neutrophils in the lower respiratory tract is correlated with disease severity. Thus, comparative analyses of the relationship between IAV infection and neutrophils provide insights into the relative contribution of host and viral factors that contribute to disease severity. Herein, we review the contribution of neutrophils to IAV disease pathogenesis and to other respiratory virus infections.
Collapse
Affiliation(s)
- Jeremy V Camp
- Institute of Virology, University of Veterinary Medicine at Vienna, Vienna, Austria
| | - Colleen B Jonsson
- Department of Microbiology, University of Tennessee-Knoxville, Knoxville, TN, USA
| |
Collapse
|
26
|
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
|
27
|
Awogbindin IO, Olaleye DO, Farombi EO. Mechanistic perspective of the oxido-immunopathologic resolution property of kolaviron in mice influenza pneumonitis. APMIS 2017; 125:184-196. [PMID: 28116826 DOI: 10.1111/apm.12640] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 10/23/2016] [Indexed: 12/18/2022]
Abstract
Implicated in influenza-associated pathology are innate defence overzealousness and unabated secretion of oxidative tissue-sensitive antimicrobial agents. At different time points, mice were pre-treated with kolaviron (400 mg/kg), a natural antioxidant and anti-inflammatory agent, and subsequently challenged with 2 LD50 influenza A/H3N2/Perth/16/09 virus. After euthanasia at day 6, blood, lungs, liver and spleen were collected and processed for biochemical, immunohistochemical and flow cytometric assessment of redo-inflammatory imbalance, cytokine storm indices and T helper 1 host response. Previously kolaviron was reported to delay mortality onset, improve morbidity and attenuate myeloperoxidase activity and nitric oxide production with minimal impact on viral clearance. This study additionally confirmed nitric oxide, but not hydrogen peroxide, as the major culprit implicated in influenza virus-induced oxido-pathology. Systemic effect of the sustained inflammation and nitrosative stress was more prominent in the spleen and lung than in the liver of mice infected with A/H3N2/Perth/16/09. Influential to immunopathology was heightened pulmonary expression of IL-1β, RANTES, IL-10, MCP-1, NF-κB, iNOS and COX-2. However, kolaviron combated the influenza-established nitrative stress, reversed the elicited cytokine storm and restored the oxidized environment to a reductive milieu. Our data also suggest that kolaviron administration early in infection may foster CD4+ response. These data indicate that kolaviron may confer disease-dwindling properties during acute influenza infection via a system-wide protective approach involving multiple targets especially at the early stage of the infection.
Collapse
Affiliation(s)
| | - David O Olaleye
- Department of Virology, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
28
|
Implications of oxidative stress on viral pathogenesis. Arch Virol 2016; 162:907-917. [PMID: 28039563 DOI: 10.1007/s00705-016-3187-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 12/08/2016] [Indexed: 12/12/2022]
Abstract
Reactive species are frequently formed after viral infections. Antioxidant defences, including enzymatic and non-enzymatic components, protect against reactive species, but sometimes these defences are not completely adequate. An imbalance in the production of reactive species and the body's inability to detoxify these reactive species is referred to as oxidative stress. The aim of this review is to analyse the role of oxidative stress in the pathogenesis of viral infections and highlight some major therapeutic approaches that have gained importance, with regards to controlling virus-induced oxidative injury. Attention will be focused on DNA viruses (papillomaviruses, hepadnaviruses), RNA viruses (flaviviruses, orthomyxoviruses, paramyxoviruses, togaviruses) and retroviruses (human immunodeficiency virus). In general, viruses cause an imbalance in the cellular redox environment, which depending on the virus and the cell can result in different responses, e.g. cell signaling, antioxidant defences, reactive species, and other processes. Therefore, the modulation of reactive species production and oxidative stress potentially represents a novel pharmacological approach for reducing the consequences of viral pathogenesis.
Collapse
|
29
|
Rong R, Li RR, Hou YB, Li J, Ding JX, Zhang CB, Yang Y. Mahuang-Xixin-Fuzi decoction reduces the infection of influenza A virus in Kidney-Yang deficiency syndrome mice. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:217-224. [PMID: 27401293 DOI: 10.1016/j.jep.2016.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/19/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mahuang-Xixin-Fuzi Decoction (MXF) as a famous formula for the treatment of colds, fever, nasal congestion and headache with elder people, has always been widely used in traditional Chinese medicine. The present study is aimed at investigating the treatment effect of MXF on Kidney-Yang deficiency syndrome in mice simultaneously infected with H1N1 virus. MATERIALS AND METHODS We employed the Kidney-Yang deficiency mouse model to investigate the effect of MXF against influenza A virus (A/FM/1/47, H1N1). Mice were infected with the virus after fifteen days Kidney-Yang deficiency syndrome was established (intraperitoneal injection of estradiol benzoate), while MXF was orally administrated with 1.2-4.7g/kg/d for 6 consecutive days after inoculation. Body weight, rectal temperature, morbidity, and mortality were recorded daily. Histopathologic changes, antioxidant activity of SOD and MDA were detected. Moreover, levels of inflammatory cytokines including IL-6, IL-10, MCP-1, TNF-α were measured in the sera of mice. RESULTS We found that the extract of MXF at dosages of 2.3-4.7g/kg could effectively diminish mortality rate, ameliorate lung edema and inflammation. Administration of MXF decoction significantly depressed the expression of IL-6, MCP-1 and TNF-α, and markedly increased expression of IL-10 in serum. Simultaneously, the extract was also found to reduce MDA and increase SOD in the lung tissue of mice. CONCLUSION These data support the notion that the extract of MXF could treat Kidney-Yang deficiency syndrome in mice simultaneously infected with influenza A virus by reducing inflammation and increasing antioxidant activities.
Collapse
Affiliation(s)
- Rong Rong
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Rong-Rong Li
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Municipal Hospital of Yantai City Government, Yantai 264000, China
| | - Yan-Bao Hou
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jing Li
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jia-Xing Ding
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Lvye Pharmaceutical Co., Ltd., Yantai 264003, China
| | - Cheng-Bo Zhang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yong Yang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
30
|
Talmi-Frank D, Altboum Z, Solomonov I, Udi Y, Jaitin D, Klepfish M, David E, Zhuravlev A, Keren-Shaul H, Winter D, Gat-Viks I, Mandelboim M, Ziv T, Amit I, Sagi I. Extracellular Matrix Proteolysis by MT1-MMP Contributes to Influenza-Related Tissue Damage and Mortality. Cell Host Microbe 2016; 20:458-470. [DOI: 10.1016/j.chom.2016.09.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 06/19/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022]
|
31
|
Garozzo A, Tempera G, Ungheri D, Timpanaro R, Castro A. N-Acetylcysteine Synergizes with Oseltamivir in Protecting Mice from Lethal Influenza Infection. Int J Immunopathol Pharmacol 2016; 20:349-54. [PMID: 17624247 DOI: 10.1177/039463200702000215] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Many studies have shown that oxidative stress is important in the pathogenesis of pulmonary damage during influenza virus infections. Antioxidant molecules are therefore potentially useful against viral infection. Our previous studies show that N-acetylcysteine (NAC) has a protective effect in a model of lethal influenza infection in mice. NAC administration significantly decreased the mortality in infected mice. Further studies have demonstrated that NAC enhanced survival in combination with the antiviral agent ribavirin. In the present study, we report the effect of combined treatment with NAC and Oseltamivir, clinically used in the treatment and prevention of influenza virus infection, in a murine model of lethal influenza infection. NAC was given as a single daily dose of 1000 mg/Kg starting from 4 h before infection and until day 4 after infection; Oseltamivir was given twice daily at dose of 1 mg/Kg/die for 5 days, starting from 4 h before infection. End-point evaluation was 21-days' survival. NAC alone was slightly effective (20%), since a suboptimal treatment was used. Survival increased to 60% with Oseltamivir and to 100% with Oseltamivir and NAC used in combination. Since NAC alone does not show any antiviral action, the present findings suggest that antioxidant therapy increase survival by an improvement in host defense mechanisms, and/or by a direct antioxidant effect against oxidative stress associated with viral infection. Our studies demonstrate the effectiveness of combining agents acting through different mechanisms, such as antiviral drugs oseltamivir and the antioxidant NAC, indicating a possible advantage of combining the two treatments.
Collapse
Affiliation(s)
- A Garozzo
- Department of Microbiological and Gynaecological Sciences, University of Catania, Italy
| | | | | | | | | |
Collapse
|
32
|
Abstract
Pulmonary oxidant stress plays an important pathogenetic role in disease conditions including acute lung injury/adult respiratory distress syndrome (ALI/ARDS), hyperoxia, ischemia-reperfusion, sepsis, radiation injury, lung transplantation, COPD, and inflammation. Reactive oxygen species (ROS), released from activated macrophages and leukocytes or formed in the pulmonary epithelial and endothelial cells, damage the lungs and initiate cascades of pro-inflammatory reactions propagating pulmonary and systemic stress. Diverse molecules including small organic compounds (e.g. gluthatione, tocopherol (vitamin E), flavonoids) serve as natural antioxidants that reduce oxidized cellular components, decompose ROS and detoxify toxic oxidation products. Antioxidant enzymes can either facilitate these antioxidant reactions (e.g. peroxidases using glutathione as a reducing agent) or directly decompose ROS (e.g. superoxide dismutases [SOD] and catalase). Many antioxidant agents are being tested for treatment of pulmonary oxidant stress. The administration of small antioxidants via the oral, intratracheal and vascular routes for the treatment of short- and long-term oxidant stress showed rather modest protective effects in animal and human studies. Intratracheal and intravascular administration of antioxidant enzymes are being currently tested for the treatment of acute oxidant stress. For example, intratracheal administration of recombinant human SOD is protective in premature infants exposed to hyperoxia. However, animal and human studies show that more effective delivery of drugs to cells experiencing oxidant stress is needed to improve protection. Diverse delivery systems for antioxidants including liposomes, chemical modifications (e.g. attachment of masking pegylated [PEG]-groups) and coupling to affinity carriers (e.g. antibodies against cellular adhesion molecules) are being employed and currently tested, mostly in animal and, to a limited extent, in humans, for the treatment of oxidant stress. Further studies are needed, however, in order to develop and establish effective applications of pulmonary antioxidant interventions useful in clinical practice. Although beyond the scope of this review, antioxidant gene therapies may eventually provide a strategy for the management of subacute and chronic pulmonary oxidant stress.
Collapse
Affiliation(s)
- Melpo Christofidou-Solomidou
- Institute of Environmental Medicine and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
33
|
Complex compounds of zinc and copper(ii) ions with dihydroquercetin and their antiviral activity. Russ Chem Bull 2016. [DOI: 10.1007/s11172-015-1027-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
Lei XG, Zhu JH, Cheng WH, Bao Y, Ho YS, Reddi AR, Holmgren A, Arnér ESJ. Paradoxical Roles of Antioxidant Enzymes: Basic Mechanisms and Health Implications. Physiol Rev 2016; 96:307-64. [PMID: 26681794 DOI: 10.1152/physrev.00010.2014] [Citation(s) in RCA: 277] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated from aerobic metabolism, as a result of accidental electron leakage as well as regulated enzymatic processes. Because ROS/RNS can induce oxidative injury and act in redox signaling, enzymes metabolizing them will inherently promote either health or disease, depending on the physiological context. It is thus misleading to consider conventionally called antioxidant enzymes to be largely, if not exclusively, health protective. Because such a notion is nonetheless common, we herein attempt to rationalize why this simplistic view should be avoided. First we give an updated summary of physiological phenotypes triggered in mouse models of overexpression or knockout of major antioxidant enzymes. Subsequently, we focus on a series of striking cases that demonstrate "paradoxical" outcomes, i.e., increased fitness upon deletion of antioxidant enzymes or disease triggered by their overexpression. We elaborate mechanisms by which these phenotypes are mediated via chemical, biological, and metabolic interactions of the antioxidant enzymes with their substrates, downstream events, and cellular context. Furthermore, we propose that novel treatments of antioxidant enzyme-related human diseases may be enabled by deliberate targeting of dual roles of the pertaining enzymes. We also discuss the potential of "antioxidant" nutrients and phytochemicals, via regulating the expression or function of antioxidant enzymes, in preventing, treating, or aggravating chronic diseases. We conclude that "paradoxical" roles of antioxidant enzymes in physiology, health, and disease derive from sophisticated molecular mechanisms of redox biology and metabolic homeostasis. Simply viewing antioxidant enzymes as always being beneficial is not only conceptually misleading but also clinically hazardous if such notions underpin medical treatment protocols based on modulation of redox pathways.
Collapse
Affiliation(s)
- Xin Gen Lei
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jian-Hong Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Wen-Hsing Cheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yongping Bao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ye-Shih Ho
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Amit R Reddi
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Arne Holmgren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Elias S J Arnér
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
The Influenza Virus H5N1 Infection Can Induce ROS Production for Viral Replication and Host Cell Death in A549 Cells Modulated by Human Cu/Zn Superoxide Dismutase (SOD1) Overexpression. Viruses 2016; 8:v8010013. [PMID: 26761025 PMCID: PMC4728573 DOI: 10.3390/v8010013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022] Open
Abstract
Highly pathogenic H5N1 infections are often accompanied by excessive pro-inflammatory response, high viral titer, and apoptosis; as such, the efficient control of these infections poses a great challenge. The pathogenesis of influenza virus infection is also related to oxidative stress. However, the role of endogenic genes with antioxidant effect in the control of influenza viruses, especially H5N1 viruses, should be further investigated. In this study, the H5N1 infection in lung epithelial cells decreased Cu/Zn superoxide dismutase (SOD1) expression at mRNA and protein levels. Forced SOD1 expression significantly inhibited the H5N1-induced increase in reactive oxygen species, decreased pro-inflammatory response, prevented p65 and p38 phosphorylation, and impeded viral ribonucleoprotein nuclear export and viral replication. The SOD1 overexpression also rescued H5N1-induced cellular apoptosis and alleviated H5N1-caused mitochondrial dysfunction. Therefore, this study described the role of SOD1 in the replication of H5N1 influenza virus and emphasized the relevance of this enzyme in the control of H5N1 replication in epithelial cells. Pharmacological modulation or targeting SOD1 may open a new way to fight H5N1 influenza virus.
Collapse
|
36
|
Preventive Activity against Influenza (H1N1) Virus by Intranasally Delivered RNA-Hydrolyzing Antibody in Respiratory Epithelial Cells of Mice. Viruses 2015; 7:5133-44. [PMID: 26402693 PMCID: PMC4584307 DOI: 10.3390/v7092863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/10/2015] [Accepted: 09/14/2015] [Indexed: 01/22/2023] Open
Abstract
The antiviral effect of a catalytic RNA-hydrolyzing antibody, 3D8 scFv, for intranasal administration against avian influenza virus (H1N1) was described. The recombinant 3D8 scFv protein prevented BALB/c mice against H1N1 influenza virus infection by degradation of the viral RNA genome through its intrinsic RNA-hydrolyzing activity. Intranasal administration of 3D8 scFv (50 μg/day) for five days prior to infection demonstrated an antiviral activity (70% survival) against H1N1 infection. The antiviral ability of 3D8 scFv to penetrate into epithelial cells from bronchial cavity via the respiratory mucosal layer was confirmed by immunohistochemistry, qRT-PCR, and histopathological examination. The antiviral activity of 3D8 scFv against H1N1 virus infection was not due to host immune cytokines or chemokines, but rather to direct antiviral RNA-hydrolyzing activity of 3D8 scFv against the viral RNA genome. Taken together, our results suggest that the RNase activity of 3D8 scFv, coupled with its ability to penetrate epithelial cells through the respiratory mucosal layer, directly prevents H1N1 virus infection in a mouse model system.
Collapse
|
37
|
Sgarbanti R, Amatore D, Celestino I, Marcocci ME, Fraternale A, Ciriolo MR, Magnani M, Saladino R, Garaci E, Palamara AT, Nencioni L. Intracellular redox state as target for anti-influenza therapy: are antioxidants always effective? Curr Top Med Chem 2015; 14:2529-41. [PMID: 25478883 PMCID: PMC4435240 DOI: 10.2174/1568026614666141203125211] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/29/2014] [Accepted: 11/02/2014] [Indexed: 12/12/2022]
Abstract
Influenza virus infections represent a big issue for public health since effective treatments are still lacking. In particular, the emergence of strains resistant to drugs limits the effectiveness of anti-influenza agents. For this reason, many efforts have been dedicated to the identification of new therapeutic strategies aimed at targeting the virus-host cell interactions. Oxidative stress is a characteristic of some viral infections including influenza. Because antioxidants defend cells from damage caused by reactive oxygen species induced by different stimuli including pathogens, they represent interesting molecules to fight infectious diseases. However, most of the available studies have found that these would-be panaceas could actually exacerbate the diseases they claim to prevent, and have thus revealed "the dark side" of these molecules. This review article discusses the latest opportunities and drawbacks of the antioxidants used in anti-influenza therapy and new perspectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
38
|
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
|
39
|
Cram DL, Blount JD, York JE, Young AJ. Immune response in a wild bird is predicted by oxidative status, but does not cause oxidative stress. PLoS One 2015; 10:e0122421. [PMID: 25815888 PMCID: PMC4376632 DOI: 10.1371/journal.pone.0122421] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/20/2015] [Indexed: 12/26/2022] Open
Abstract
The immune system provides vital protection against pathogens, but extensive evidence suggests that mounting immune responses can entail survival and fecundity costs. The physiological mechanisms that underpin these costs remain poorly understood, despite their potentially important role in shaping life-histories. Recent studies involving laboratory models highlight the possibility that oxidative stress could mediate these costs, as immune-activation can increase the production of reactive oxygen species leading to oxidative stress. However, this hypothesis has rarely been tested in free-ranging wild populations, where natural oxidative statuses and compensatory strategies may moderate immune responses and their impacts on oxidative status. Furthermore, the possibility that individuals scale their immune responses according to their oxidative status, conceivably to mitigate such costs, remains virtually unexplored. Here, we experimentally investigate the effects of a phytohaemagglutinin (PHA) immune-challenge on oxidative status in wild male and female white-browed sparrow weavers, Plocepasser mahali. We also establish whether baseline oxidative status prior to challenge predicts the scale of the immune responses. Contrary to previous work on captive animals, our findings suggest that PHA-induced immune-activation does not elicit oxidative stress. Compared with controls (n = 25 birds), PHA-injected birds (n = 27 birds) showed no evidence of a differential change in markers of oxidative damage or enzymatic and non-enzymatic antioxidant protection 24 hours after challenge. We did, however, find that the activity of a key antioxidant enzyme (superoxide dismutase, SOD) prior to immune-activation predicted the scale of the resulting swelling: birds with stronger initial SOD activity subsequently produced smaller swellings. Our findings (i) suggest that wild birds can mount immune responses without suffering from systemic oxidative stress, and (ii) lend support to biomedical evidence that baseline oxidative status can impact the scale of immune responses; a possibility not yet recognised in ecological studies of immunity.
Collapse
Affiliation(s)
- Dominic L. Cram
- Centre for Ecology & Conservation, College of Life & Environmental Sciences, University of Exeter, Penryn Campus, Cornwall, United Kingdom
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan D. Blount
- Centre for Ecology & Conservation, College of Life & Environmental Sciences, University of Exeter, Penryn Campus, Cornwall, United Kingdom
| | - Jennifer E. York
- Centre for Ecology & Conservation, College of Life & Environmental Sciences, University of Exeter, Penryn Campus, Cornwall, United Kingdom
- Department of Zoology and Entomology, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Andrew J. Young
- Centre for Ecology & Conservation, College of Life & Environmental Sciences, University of Exeter, Penryn Campus, Cornwall, United Kingdom
| |
Collapse
|
40
|
Vlahos R, Selemidis S. NADPH oxidases as novel pharmacologic targets against influenza A virus infection. Mol Pharmacol 2014; 86:747-59. [PMID: 25301784 DOI: 10.1124/mol.114.095216] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Influenza A viruses represent a major global health care challenge, with imminent pandemics, emerging antiviral resistance, and long lag times for vaccine development, raising a pressing need for novel pharmacologic strategies that ideally target the pathology irrespective of the infecting strain. Reactive oxygen species (ROS) pervade all facets of cell biology with both detrimental and protective properties. Indeed, there is compelling evidence that activation of the NADPH oxidase 2 (NOX2) isoform of the NADPH oxidase family of ROS-producing enzymes promotes lung oxidative stress, inflammation, injury, and dysfunction resulting from influenza A viruses of low to high pathogenicity, as well as impeding virus clearance. By contrast, the dual oxidase isoforms produce ROS that provide vital protective antiviral effects for the host. In this review, we propose that inhibitors of NOX2 are better alternatives than broad-spectrum antioxidant approaches for treatment of influenza pathologies, for which clinical efficacy may have been limited owing to poor bioavailability and inadvertent removal of beneficial ROS. Finally, we briefly describe the current suite of NADPH oxidase inhibitors and the molecular features of the NADPH oxidase enzymes that could be exploited by drug discovery for development of more specific and novel inhibitors to prevent or treat disease caused by influenza.
Collapse
Affiliation(s)
- Ross Vlahos
- Respiratory Research Group, Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne (R.V.), and Oxidant and Inflammation Biology Group, Department of Pharmacology, Monash University (S.S.), Victoria, Australia
| | - Stavros Selemidis
- Respiratory Research Group, Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne (R.V.), and Oxidant and Inflammation Biology Group, Department of Pharmacology, Monash University (S.S.), Victoria, Australia
| |
Collapse
|
41
|
Tanaka R, Ishima Y, Enoki Y, Kimachi K, Shirai T, Watanabe H, Chuang VTG, Maruyama T, Otagiri M. Therapeutic impact of human serum albumin-thioredoxin fusion protein on influenza virus-induced lung injury mice. Front Immunol 2014; 5:561. [PMID: 25414704 PMCID: PMC4220708 DOI: 10.3389/fimmu.2014.00561] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/21/2014] [Indexed: 01/07/2023] Open
Abstract
Reactive oxygen species (ROS) are the primary pathogenic molecules produced in viral lung infections. We previously reported on the use of a recombinant human serum albumin (HSA)–thioredoxin 1 (Trx) fusion protein (HSA–Trx) for extending the half-life Trx, an endogenous protein with anti-oxidant properties. As a result, it was possible to overcome the unfavorable pharmacokinetic and short pharmacological properties of Trx. We hypothesized that HSA–Trx would attenuate the enhanced ROS production of species such as hydroxyl radicals by neutrophils during an influenza viral infection. The levels of 8-hydroxy-2′-deoxyguanosine and 3-nitrotyrosine were used as indices of the anti-oxidant activity of HSA–Trx. In addition, the cytoprotective effects of HSA–Trx were examined in PR8 (H1N1) influenza virus-induced lung injured mice. The findings show that HSA–Trx reduced the number of total cells, neutrophils, and total protein in BALF of influenza virus-induced lung injured mice. The HSA–Trx treatment significantly decreased the level of 8-hydroxy-2′-deoxyguanosine and 3-nitrotyrosine, but failed to inhibit inducible nitric oxide synthase expression, in the lungs of the virus-infected mice. On the other hand, Tamiflu® treatment also significantly suppressed the production of inflammatory cells and neutrophil infiltration, as well as the protein level in BALF and lung histopathological alterations caused by the influenza virus. The suppressive effect of Tamiflu® was slightly stronger than that of HSA–Trx. Interestingly, Tamiflu® significantly decreased virus proliferation, while HSA–Trx had no effect. These results indicate that HSA–Trx may be of therapeutic value for 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 lung.
Collapse
Affiliation(s)
- Ryota Tanaka
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University , Kumamoto , Japan
| | - Yu Ishima
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University , Kumamoto , Japan ; Center for Clinical Pharmaceutical Sciences, School of Pharmacy, Kumamoto University , Kumamoto , Japan
| | - Yuki Enoki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University , Kumamoto , Japan
| | - Kazuhiko Kimachi
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN) , Kumamoto , Japan
| | - Tatsuya Shirai
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN) , Kumamoto , Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University , Kumamoto , Japan ; Center for Clinical Pharmaceutical Sciences, School of Pharmacy, Kumamoto University , Kumamoto , Japan
| | - Victor T G Chuang
- Curtin Health Innovation Research Institute, School of Pharmacy, Faculty of Health Sciences, Curtin University , Perth, WA , Australia
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University , Kumamoto , Japan ; Center for Clinical Pharmaceutical Sciences, School of Pharmacy, Kumamoto University , Kumamoto , Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University , Kumamoto , Japan ; DDS Research Institute, Sojo University , Kumamoto , Japan
| |
Collapse
|
42
|
NADPH oxidases: an overview from structure to innate immunity-associated pathologies. Cell Mol Immunol 2014; 12:5-23. [PMID: 25263488 DOI: 10.1038/cmi.2014.89] [Citation(s) in RCA: 681] [Impact Index Per Article: 61.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022] Open
Abstract
Oxygen-derived free radicals, collectively termed reactive oxygen species (ROS), play important roles in immunity, cell growth, and cell signaling. In excess, however, ROS are lethal to cells, and the overproduction of these molecules leads to a myriad of devastating diseases. The key producers of ROS in many cells are the NOX family of NADPH oxidases, of which there are seven members, with various tissue distributions and activation mechanisms. NADPH oxidase is a multisubunit enzyme comprising membrane and cytosolic components, which actively communicate during the host responses to a wide variety of stimuli, including viral and bacterial infections. This enzymatic complex has been implicated in many functions ranging from host defense to cellular signaling and the regulation of gene expression. NOX deficiency might lead to immunosuppression, while the intracellular accumulation of ROS results in the inhibition of viral propagation and apoptosis. However, excess ROS production causes cellular stress, leading to various lethal diseases, including autoimmune diseases and cancer. During the later stages of injury, NOX promotes tissue repair through the induction of angiogenesis and cell proliferation. Therefore, a complete understanding of the function of NOX is important to direct the role of this enzyme towards host defense and tissue repair or increase resistance to stress in a timely and disease-specific manner.
Collapse
|
43
|
Barbosa RPA, Salgado APC, Garcia CC, Filho BG, Gonçalves APDF, Lima BHF, Lopes GAO, Rachid MA, Peixoto ACC, de Oliveira DB, Ataíde MA, Zirke CA, Cotrim TM, Costa ÉA, Almeida GMDF, Russo RC, Gazzinelli RT, Machado ADMV. Protective immunity and safety of a genetically modified influenza virus vaccine. PLoS One 2014; 9:e98685. [PMID: 24927156 PMCID: PMC4057169 DOI: 10.1371/journal.pone.0098685] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 05/07/2014] [Indexed: 12/01/2022] Open
Abstract
Recombinant influenza viruses are promising viral platforms to be used as antigen delivery vectors. To this aim, one of the most promising approaches consists of generating recombinant viruses harboring partially truncated neuraminidase (NA) segments. To date, all studies have pointed to safety and usefulness of this viral platform. However, some aspects of the inflammatory and immune responses triggered by those recombinant viruses and their safety to immunocompromised hosts remained to be elucidated. In the present study, we generated a recombinant influenza virus harboring a truncated NA segment (vNA-Δ) and evaluated the innate and inflammatory responses and the safety of this recombinant virus in wild type or knock-out (KO) mice with impaired innate (Myd88 -/-) or acquired (RAG -/-) immune responses. Infection using truncated neuraminidase influenza virus was harmless regarding lung and systemic inflammatory response in wild type mice and was highly attenuated in KO mice. We also demonstrated that vNA-Δ infection does not induce unbalanced cytokine production that strongly contributes to lung damage in infected mice. In addition, the recombinant influenza virus was able to trigger both local and systemic virus-specific humoral and CD8+ T cellular immune responses which protected immunized mice against the challenge with a lethal dose of homologous A/PR8/34 influenza virus. Taken together, our findings suggest and reinforce the safety of using NA deleted influenza viruses as antigen delivery vectors against human or veterinary pathogens.
Collapse
Affiliation(s)
- Rafael Polidoro Alves Barbosa
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunoparasitologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Ana Paula Carneiro Salgado
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Cristiana Couto Garcia
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunologia e Mecânica Pulmonar, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Bruno Galvão Filho
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | | | - Braulio Henrique Freire Lima
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunologia e Mecânica Pulmonar, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Gabriel Augusto Oliveira Lopes
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunologia e Mecânica Pulmonar, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Milene Alvarenga Rachid
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Andiara Cristina Cardoso Peixoto
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunologia e Mecânica Pulmonar, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Danilo Bretas de Oliveira
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Marco Antônio Ataíde
- Laboratório de Imunoparasitologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Carla Aparecida Zirke
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Tatiane Marques Cotrim
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Érica Azevedo Costa
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
| | - Gabriel Magno de Freitas Almeida
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Remo Castro Russo
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunologia e Mecânica Pulmonar, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Ricardo Tostes Gazzinelli
- Laboratório de Imunopatologia, Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brasil
- Laboratório de Imunoparasitologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | | |
Collapse
|
44
|
The c10orf10 gene product is a new link between oxidative stress and autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1076-88. [DOI: 10.1016/j.bbamcr.2014.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 01/21/2014] [Accepted: 02/06/2014] [Indexed: 01/12/2023]
|
45
|
Reshi ML, Su YC, Hong JR. RNA Viruses: ROS-Mediated Cell Death. Int J Cell Biol 2014; 2014:467452. [PMID: 24899897 PMCID: PMC4034720 DOI: 10.1155/2014/467452] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 03/18/2014] [Accepted: 03/20/2014] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) are well known for being both beneficial and deleterious. The main thrust of this review is to investigate the role of ROS in ribonucleic acid (RNA) virus pathogenesis. Much evidences has accumulated over the past decade, suggesting that patients infected with RNA viruses are under chronic oxidative stress. Changes to the body's antioxidant defense system, in relation to SOD, ascorbic acid, selenium, carotenoids, and glutathione, have been reported in various tissues of RNA-virus infected patients. This review focuses on RNA viruses and retroviruses, giving particular attention to the human influenza virus, Hepatitis c virus (HCV), human immunodeficiency virus (HIV), and the aquatic Betanodavirus. Oxidative stress via RNA virus infections can contribute to several aspects of viral disease pathogenesis including apoptosis, loss of immune function, viral replication, inflammatory response, and loss of body weight. We focus on how ROS production is correlated with host cell death. Moreover, ROS may play an important role as a signal molecule in the regulation of viral replication and organelle function, potentially providing new insights in the prevention and treatment of RNA viruses and retrovirus infections.
Collapse
Affiliation(s)
- Mohammad Latif Reshi
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Yi-Che Su
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Jiann-Ruey Hong
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
46
|
Longoni SS, Marín C, Sánchez-Moreno M. Excreted Leishmania peruviana and Leishmania amazonensis iron-superoxide dismutase purification: specific antibody detection in Colombian patients with cutaneous leishmaniasis. Free Radic Biol Med 2014; 69:26-34. [PMID: 24440468 DOI: 10.1016/j.freeradbiomed.2014.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 11/30/2022]
Abstract
Leishmania sp. survival in the vertebrate host depends on the host macrophage immune response as well as on the parasite's defense against free radicals. Iron-superoxide dismutase (Fe-SOD) is a key antioxidant enzyme that contributes to radical superoxide dismutation, preventing the disease from surging and propagating itself. Leishmania sp. has various Fe-SOD isoforms, one of which (Fe-SODe) is excreted into the medium and, being highly immunogenic, can be considered a very good molecular marker. In this work, we purified the Fe-SOD enzymes excreted by L. peruviana and L. amazonensis and studied them as antigens in serodiagnosis. We used ELISA and Western blot techniques to test 51 human cutaneous leishmaniasis sera from Colombia. All 51 patients presented with dermal injuries caused by unknown Leishmania species. The results observed with the purified proteins were compared with those obtained when total soluble lysate and unpurified Fe-SODe were used as the antigen fraction. Thus, we conclude that the purified enzymes are more sensitive and specific than their unpurified counterparts and that there is no cross-reactivity between them.
Collapse
Affiliation(s)
- Silvia Stefania Longoni
- Departamento de Parasitología, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain.
| | - Clotilde Marín
- Departamento de Parasitología, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain
| | - Manuel Sánchez-Moreno
- Departamento de Parasitología, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain
| |
Collapse
|
47
|
Abstract
Reactive oxygen species (ROS) formation is part of normal cellular aerobic metabolism, due to respiration and oxidation of nutrients in order to generate energy. Low levels of ROS are involved in cellular signaling and are well controlled by the cellular antioxidant defense system. Elevated levels of ROS generation due to pollutants, toxins and radiation exposure, as well as infections, are associated with oxidative stress causing cellular damage. Several respiratory viruses, including respiratory syncytial virus (RSV), human metapneumovirus (hMPV) and influenza, induce increased ROS formation, both intracellularly and as a result of increased inflammatory cell recruitment at the site of infection. They also reduce antioxidant enzyme (AOE) levels and/or activity, leading to unbalanced oxidative-antioxidant status and subsequent oxidative cell damage. Expression of several AOE is controlled by the activation of the nuclear transcription factor NF-E2-related factor 2 (Nrf2), through binding to the antioxidant responsive element (ARE) present in the AOE gene promoters. While exposure to several pro-oxidant stimuli usually leads to Nrf2 activation and upregulation of AOE expression, respiratory viral infections are associated with inhibition of AOE expression/activity, which in the case of RSV and hMPV is associated with reduced Nrf2 nuclear localization, decreased cellular levels and reduced ARE-dependent gene transcription. Therefore, administration of antioxidant mimetics or Nrf2 inducers represents potential viable therapeutic approaches to viral-induced diseases, such as respiratory infections and other infections associated with decreased cellular antioxidant capacity.
Collapse
Affiliation(s)
- Narayana Komaravelli
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA ; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA ; Department of Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
48
|
Miller VM, Zhu Y, Bucher C, McGinnis W, Ryan LK, Siegel A, Zalcman S. Gestational flu exposure induces changes in neurochemicals, affiliative hormones and brainstem inflammation, in addition to autism-like behaviors in mice. Brain Behav Immun 2013; 33:153-63. [PMID: 23880236 DOI: 10.1016/j.bbi.2013.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/27/2013] [Accepted: 07/09/2013] [Indexed: 12/23/2022] Open
Abstract
The prevalence of neurodevelopmental disorders such as autism is increasing, however the etiology of these disorders is unclear and thought to involve a combination of genetic, environmental and immune factors. A recent epidemiological study found that gestational viral exposure during the first trimester increases risk of autism in offspring by twofold. In mice gestational viral exposures alter behavior of offspring, but the biological mechanisms which underpin these behavioral changes are unclear. We hypothesized that gestational viral exposure induces changes in affiliative hormones, brainstem autonomic nuclei and neurotransmitters which are associated with behavioral alterations in offspring. To address this hypothesis, we exposed pregnant mice to influenza A virus (H3N2) on gestational day 9 and determined behavioral, hormonal and brainstem changes in male and female offspring. We found that gestational flu exposure induced dose-dependent alterations in social and aggressive behaviors (p≤0.05) in male and female offspring and increases in locomotor behaviors particularly in male offspring (p≤0.05). We found that flu exposure was also associated with reductions in oxytocin and serotonin (p≤0.05) levels in male and female offspring and sex-specific changes in dopamine metabolism. In addition we found changes in catecholaminergic and microglia density in brainstem tissues of male flu exposed offspring only (p≤0.05). This study demonstrates that gestational viral exposure induces behavioral changes in mice, which are associated with alterations in affiliative hormones. In addition we found sex-specific changes in locomotor behavior, which may be associated with sex-specific alterations in dopamine metabolism and brainstem inflammation. Further investigations into maternal immune responses are necessary to unravel the molecular mechanisms which underpin abnormal hormonal, immune and behavioral responses in offspring after gestational viral exposure.
Collapse
Affiliation(s)
- V M Miller
- Clinical Medical Sciences Building, Wadsworth Center, New York State Department of Health, New Scotland Avenue, Albany, NY 12208, USA; Department of Environmental Health Science, School of Public Health, State University of New York at Albany, Wadsworth Center, Empire State Plaza, NY 12201-0509, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Genome-wide fitness profiling reveals adaptations required by Haemophilus in coinfection with influenza A virus in the murine lung. Proc Natl Acad Sci U S A 2013; 110:15413-8. [PMID: 24003154 DOI: 10.1073/pnas.1311217110] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bacterial coinfection represents a major cause of morbidity and mortality in epidemics of influenza A virus (IAV). The bacterium Haemophilus influenzae typically colonizes the human upper respiratory tract without causing disease, and yet in individuals infected with IAV, it can cause debilitating or lethal secondary pneumonia. Studies in murine models have detected immune components involved in susceptibility and pathology, and yet few studies have examined bacterial factors contributing to coinfection. We conducted genome-wide profiling of the H. influenzae genes that promote its fitness in a murine model of coinfection with IAV. Application of direct, high-throughput sequencing of transposon insertion sites revealed fitness phenotypes of a bank of H. influenzae mutants in viral coinfection in comparison with bacterial infection alone. One set of virulence genes was required in nonvirally infected mice but not in coinfection, consistent with a defect in anti-bacterial defenses during coinfection. Nevertheless, a core set of genes required in both in vivo conditions indicated that many bacterial countermeasures against host defenses remain critical for coinfection. The results also revealed a subset of genes required in coinfection but not in bacterial infection alone, including the iron-sulfur cluster regulator gene, iscR, which was required for oxidative stress resistance. Overexpression of the antioxidant protein Dps in the iscR mutant restored oxidative stress resistance and ability to colonize in coinfection. The results identify bacterial stress and metabolic adaptations required in an IAV coinfection model, revealing potential targets for treatment or prevention of secondary bacterial pneumonia after viral infection.
Collapse
|
50
|
Dundaroz R, Erenberk U, Turel O, Demir AD, Ozkaya E, Erel O. Oxidative and antioxidative status of children with acute bronchiolitis. J Pediatr (Rio J) 2013; 89:407-11. [PMID: 23796358 DOI: 10.1016/j.jped.2012.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 12/05/2012] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Oxidative stress has been shown to contribute to the pathogenesis of acute and chronic lung inflammatory diseases. This article aimed to evaluate the oxidant/antioxidant status of children with acute bronchiolitis through the measurement of plasma total antioxidant capacity, total oxidant status, and oxidative stress index. METHODS Children with acute bronchiolitis admitted to the pediatric emergency department of a university hospital between January and April of 2012 were compared with age-matched healthy controls. Patients with acute bronchiolitis were classified as mild and moderate bronchiolitis. Oxidative and antioxidative status were assessed by measurement of plasma total antioxidant capacity, total oxidant status, and oxidative stress index. RESULTS Thirty-one children with acute bronchiolitis aged between 3 months and 2 years, and 39 healthy children were included. Total oxidative status (TOS) was higher in patients with acute bronchiolitis than the control group (5.16±1.99 μmol H2O2 versus 3.78±1.78 μmol H2O2 [p=0.004]). Total antioxidant capacity (TAC) was lower in children with bronchiolitis than the control group (2.51±0.37 μmol Trolox eqv/L versus 2.75±0.39 μmol Trolox eqv/L [p=0.013]). Patients with moderate bronchiolitis presented higher TOS levels than those with mild bronchiolitis and the control group (p=0.03, p<0.001, respectively). Patients with moderate bronchiolitis had higher oxidative stress index levels than the control group (p=0.015). Oxygen saturation level of bronchiolitis patients was inversely correlated with TOS (r=-0.476, p<0.05). CONCLUSION The balance between oxidant and antioxidant systems is disrupted in children with moderate bronchiolitis, which indicates that this stress factor may have a role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Rusen Dundaroz
- Department of Pediatrics, Bezmialem Vakif University Faculty of Medicine, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|