1
|
Dempsey MP, Conrady CD. The Host-Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment. Microorganisms 2023; 11:2074. [PMID: 37630634 PMCID: PMC10460047 DOI: 10.3390/microorganisms11082074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Ocular infectious diseases are an important cause of potentially preventable vision loss and blindness. In the following manuscript, we will review ocular immunology and the pathogenesis of herpesviruses and Pseudomonas aeruginosa infections of the cornea and posterior segment. We will highlight areas of future research and what is currently known to promote bench-to-bedside discoveries to improve clinical outcomes of these debilitating ocular diseases.
Collapse
Affiliation(s)
- Michael P. Dempsey
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Christopher D. Conrady
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Wolf A, Tabasi M, Zacharek M, Martin G, Hershenson MB, Meyerhoff ME, Sajjan U. S-Nitrosoglutathione Reduces the Density of Staphylococcus aureus Biofilms Established on Human Airway Epithelial Cells. ACS OMEGA 2023; 8:846-856. [PMID: 36643497 PMCID: PMC9835527 DOI: 10.1021/acsomega.2c06212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/20/2022] [Indexed: 05/03/2023]
Abstract
Patients with chronic rhinosinusitis (CRS) often show persistent colonization by bacteria in the form of biofilms which are resistant to antibiotic treatment. One of the most commonly isolated bacteria in CRS is Staphylococcus aureus (S. aureus). Nitric oxide (NO) is a potent antimicrobial agent and disperses biofilms efficiently. We hypothesized that S-nitrosoglutathione (GSNO), an endogenous NO carrier/donor, synergizes with gentamicin to disperse and reduce the bacterial biofilm density. We prepared GSNO formulations which are stable up to 12 months at room temperature and show the maximum amount of NO release within 1 h. We examined the effects of this GSNO formulation on the S. aureus biofilm established on the apical surface of the mucociliary-differentiated airway epithelial cell cultures regenerated from airway basal (stem) cells from cystic fibrosis (CF) and CRS patients. We demonstrate that for CF cells, which are defective in producing NO, treatment with GSNO at 100 μM increased the NO levels on the apical surface and reduced the biofilm bacterial density by 2 log units without stimulating pro-inflammatory effects or inducing epithelial cell death. In combination with gentamicin, GSNO further enhanced the killing of biofilm bacteria. Compared to placebo, GSNO significantly increased the ciliary beat frequency (CBF) in both infected and uninfected CF cell cultures. The combination of GSNO and gentamicin also reduced the bacterial density of biofilms grown on sinonasal epithelial cells from CRS patients and improved the CBF. These findings demonstrate that GSNO in combination with gentamicin may effectively reduce the density of biofilm bacteria in CRS patients. GSNO treatment may also enhance the mucociliary clearance by improving the CBF.
Collapse
Affiliation(s)
- Alex Wolf
- NOTA
Laboratories LLC, Ann Arbor, Michigan 48109, United States
| | - Mohsen Tabasi
- Department
of Microbiology Immunology and Inflammation, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Mark Zacharek
- Deparment
of Otolaryngology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Glenn Martin
- NOTA
Laboratories LLC, Ann Arbor, Michigan 48109, United States
| | - Marc B. Hershenson
- Department
of Pediatrics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mark E. Meyerhoff
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Umadevi Sajjan
- Department
of Microbiology Immunology and Inflammation, Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
of
Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, Pennsylvania 19140, United States
- . Phone: (215) 707-7139
| |
Collapse
|
3
|
Nekrasova LA, Shmakova AA, Samokhodskaya LM, Kirillova KI, Stoyanova SS, Mershina EA, Nazarova GB, Rubina KA, Semina EV, Kamalov AA. The Association of PLAUR Genotype and Soluble suPAR Serum Level with COVID-19-Related Lung Damage Severity. Int J Mol Sci 2022; 23:ijms232416210. [PMID: 36555850 PMCID: PMC9785175 DOI: 10.3390/ijms232416210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Uncovering the risk factors for acute respiratory disease coronavirus 2019 (COVID-19) severity may help to provide a valuable tool for early patient stratification and proper treatment implementation, improving the patient outcome and lowering the burden on the healthcare system. Here we report the results of a single-center retrospective cohort study on 151 severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-infected symptomatic hospitalized adult patients. We assessed the association of several blood test measurements, soluble urokinase receptor (uPAR) serum level and specific single nucleotide polymorphisms of ACE (I/D), NOS3 (rs2070744, rs1799983), SERPINE1 (rs1799768), PLAU (rs2227564) and PLAUR (rs344781, rs2302524) genes, with the disease severity classified by the percentage of lung involvement on computerized tomography scans. Our findings reveal that the T/C genotype of PLAUR rs2302524 was independently associated with a less severe lung damage (odds ratio 0.258 [0.071-0.811]). Along with high C-reactive protein, fibrinogen and soluble uPAR serum levels turned out to be independently associated with more severe lung damage in COVID-19 patients. The identified factors may be further employed as predictors of a possibly severe COVID-19 clinical course.
Collapse
Affiliation(s)
- Ludmila A. Nekrasova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Anna A. Shmakova
- Koltzov Institute of Developmental Biology, 117334 Moscow, Russia
| | - Larisa M. Samokhodskaya
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Karina I. Kirillova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Simona S. Stoyanova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Elena A. Mershina
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Galina B. Nazarova
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Kseniya A. Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Ekaterina V. Semina
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
- Institute of Experimental Cardiology, National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Correspondence:
| | - Armais A. Kamalov
- Medical Scientific and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
4
|
De Simone G, di Masi A, Ascenzi P. Strategies of Pathogens to Escape from NO-Based Host Defense. Antioxidants (Basel) 2022; 11:2176. [PMID: 36358549 PMCID: PMC9686644 DOI: 10.3390/antiox11112176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 06/22/2024] Open
Abstract
Nitric oxide (NO) is an essential signaling molecule present in most living organisms including bacteria, fungi, plants, and animals. NO participates in a wide range of biological processes including vasomotor tone, neurotransmission, and immune response. However, NO is highly reactive and can give rise to reactive nitrogen and oxygen species that, in turn, can modify a broad range of biomolecules. Much evidence supports the critical role of NO in the virulence and replication of viruses, bacteria, protozoan, metazoan, and fungi, thus representing a general mechanism of host defense. However, pathogens have developed different mechanisms to elude the host NO and to protect themselves against oxidative and nitrosative stress. Here, the strategies evolved by viruses, bacteria, protozoan, metazoan, and fungi to escape from the NO-based host defense are overviewed.
Collapse
Affiliation(s)
| | | | - Paolo Ascenzi
- Laboratorio Interdipartimentale di Microscopia Elettronica, Via della Vasca Navale 79, 00146 Roma, Italy
| |
Collapse
|
5
|
Los B, Preußner M, Eschke K, Vidal RM, Abdelgawad A, Olofsson D, Keiper S, Paulo-Pedro M, Grindel A, Meinke S, Trimpert J, Heyd F. Body temperature variation controls pre-mRNA processing and transcription of antiviral genes and SARS-CoV-2 replication. Nucleic Acids Res 2022; 50:6769-6785. [PMID: 35713540 PMCID: PMC9262603 DOI: 10.1093/nar/gkac513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/07/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022] Open
Abstract
Antiviral innate immunity represents the first defense against invading viruses and is key to control viral infections, including SARS-CoV-2. Body temperature is an omnipresent variable but was neglected when addressing host defense mechanisms and susceptibility to SARS-CoV-2 infection. Here, we show that increasing temperature in a 1.5°C window, between 36.5 and 38°C, strongly increases the expression of genes in two branches of antiviral immunity, nitric oxide production and type I interferon response. We show that alternative splicing coupled to nonsense-mediated decay decreases STAT2 expression in colder conditions and suggest that increased STAT2 expression at elevated temperature induces the expression of diverse antiviral genes and SARS-CoV-2 restriction factors. This cascade is activated in a remarkably narrow temperature range below febrile temperature, which reflects individual, circadian and age-dependent variation. We suggest that decreased body temperature with aging contributes to reduced expression of antiviral genes in older individuals. Using cell culture and in vivo models, we show that higher body temperature correlates with reduced SARS-CoV-2 replication, which may affect the different vulnerability of children versus seniors toward severe SARS-CoV-2 infection. Altogether, our data connect body temperature and pre-mRNA processing to provide new mechanistic insight into the regulation of antiviral innate immunity.
Collapse
Affiliation(s)
- Bruna Los
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Marco Preußner
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Kathrin Eschke
- Omiqa Bioinformatics, Altensteinstraße 40, 14195 Berlin, Germany
| | | | - Azza Abdelgawad
- Omiqa Bioinformatics, Altensteinstraße 40, 14195 Berlin, Germany
| | - Didrik Olofsson
- Institute of Virology, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Sandra Keiper
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Margarida Paulo-Pedro
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Alica Grindel
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Stefan Meinke
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Jakob Trimpert
- Omiqa Bioinformatics, Altensteinstraße 40, 14195 Berlin, Germany
| | - Florian Heyd
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| |
Collapse
|
6
|
Karunakaran KB, Balakrishnan N, Ganapathiraju MK. Interactome of SARS-CoV-2 Modulated Host Proteins With Computationally Predicted PPIs: Insights From Translational Systems Biology Studies. FRONTIERS IN SYSTEMS BIOLOGY 2022; 2. [DOI: 10.3389/fsysb.2022.815237] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Accelerated efforts to identify intervention strategies for the COVID-19 pandemic caused by SARS-CoV-2 need to be supported by deeper investigations into host invasion and response mechanisms. We constructed the neighborhood interactome network of the 332 human proteins targeted by SARS-CoV-2 proteins, augmenting it with 1,941 novel human protein-protein interactions predicted using our High-precision Protein-Protein Interaction Prediction (HiPPIP) model. Novel interactors, and the interactome as a whole, showed significant enrichment for genes differentially expressed in SARS-CoV-2-infected A549 and Calu-3 cells, postmortem lung samples of COVID-19 patients and blood samples of COVID-19 patients with severe clinical outcomes. The PPIs connected host proteins to COVID-19 blood biomarkers, ACE2 (SARS-CoV-2 entry receptor), genes differentiating SARS-CoV-2 infection from other respiratory virus infections, and SARS-CoV-targeted host proteins. Novel PPIs facilitated identification of the cilium organization functional module; we deduced the potential antiviral role of an interaction between the virus-targeted NUP98 and the cilia-associated CHMP5. Functional enrichment analyses revealed promyelocytic leukaemia bodies, midbody, cell cycle checkpoints and tristetraprolin pathway as potential viral targets. Network proximity of diabetes and hypertension associated genes to host proteins indicated a mechanistic basis for these co-morbidities in critically ill/non-surviving patients. Twenty-four drugs were identified using comparative transcriptome analysis, which include those undergoing COVID-19 clinical trials, showing broad-spectrum antiviral properties or proven activity against SARS-CoV-2 or SARS-CoV/MERS-CoV in cell-based assays. The interactome is available on a webserver at http://severus.dbmi.pitt.edu/corona/.
Collapse
|
7
|
Kamenshchikov NO, Berra L, Carroll RW. Therapeutic Effects of Inhaled Nitric Oxide Therapy in COVID-19 Patients. Biomedicines 2022; 10:biomedicines10020369. [PMID: 35203578 PMCID: PMC8962307 DOI: 10.3390/biomedicines10020369] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 01/08/2023] Open
Abstract
The global COVID-19 pandemic has become the largest public health challenge of recent years. The incidence of COVID-19-related acute hypoxemic respiratory failure (AHRF) occurs in up to 15% of hospitalized patients. Antiviral drugs currently available to clinicians have little to no effect on mortality, length of in-hospital stay, the need for mechanical ventilation, or long-term effects. Inhaled nitric oxide (iNO) administration is a promising new non-standard approach to directly treat viral burden while enhancing oxygenation. Along with its putative antiviral affect in COVID-19 patients, iNO can reduce inflammatory cell-mediated lung injury by inhibiting neutrophil activation, lowering pulmonary vascular resistance and decreasing edema in the alveolar spaces, collectively enhancing ventilation/perfusion matching. This narrative review article presents recent literature on the iNO therapy use for COVID-19 patients. The authors suggest that early administration of the iNO therapy may be a safe and promising approach for the treatment of COVID-19 patients. The authors also discuss unconventional approaches to treatment, continuous versus intermittent high-dose iNO therapy, timing of initiation of therapy (early versus late), and novel delivery systems. Future laboratory and clinical research is required to define the role of iNO as an adjunct therapy against bacterial, viral, and fungal infections.
Collapse
Affiliation(s)
- Nikolay O. Kamenshchikov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
- Correspondence:
| | - Lorenzo Berra
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA;
| | - Ryan W. Carroll
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA;
- Division of Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
8
|
Deek SA. BPC 157 as Potential Treatment for COVID-19. Med Hypotheses 2021; 158:110736. [PMID: 34798584 PMCID: PMC8575535 DOI: 10.1016/j.mehy.2021.110736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 10/07/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023]
Abstract
The emergence of coronavirus disease (COVID-19) in China at the end of 2019 has caused a large global outbreak. COVID-19 is largely seen as a thrombotic and vascular disease targeting endothelial cells (ECs) throughout the body that can provoke the breakdown of central vascular functions. This explains the complications and multi-organ failure seen in COVID-19 patients including acute respiratory distress syndrome, cardiovascular complications, liver damage, and neurological damage. Acknowledging the comorbidities and potential organ injuries throughout the course of COVID-19 is therefore crucial in the clinical management of patients. Here we discuss BPC 157, based primarily on animal model data, as a novel agent that can improve the clinical management of COVID-19. BPC 157 is a peptide that has demonstrated anti-inflammatory, cytoprotective, and endothelial-protective effects in different organ systems in different species. BPC 157 activated endothelial nitric oxide synthase (eNOS) is associated with nitric oxide (NO) release, tissue repair and angiomodulatory properties which can lead to improved vascular integrity and immune response, reduced proinflammatory profile, and reduced critical levels of the disease. As a result, discussion of its use as a potential prophylactic and complementary treatment is critical. All examined treatments, although potentiality effective against COVID-19, need either appropriate drug development or clinical trials in humans to be suitable for clinical use.
Collapse
Affiliation(s)
- Sarah A Deek
- Department of Civil, Architectural and Environmental Engineering, The University of Texas at Austin, 78712 Austin, TX, USA.
| |
Collapse
|
9
|
Maguire G. Stem cells part of the innate and adaptive immune systems as a therapeutic for Covid-19. Commun Integr Biol 2021; 14:186-198. [PMID: 34527167 PMCID: PMC8437473 DOI: 10.1080/19420889.2021.1965356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022] Open
Abstract
Some stem cell types not only release molecules that reduce viral replication, but also reduce the hypercytokinemia and inflammation induced by the immune system, and have been found to be part of the innate and adaptive immune systems. An important component of the stem cell's ability to ameliorate viral diseases, especially the complications post-clearance of the pathogen, is the ability of adult stem cells to reset the innate and adaptive immune systems from an inflammatory state to a repair state. Thus, the molecules released from certain stem cell types found to be safe and efficacious, may be an important new means for therapeutic development in Covid-19, especially for late-stage inflammation and tissue damage once the virus has cleared, particularly in the aged population.
Collapse
Affiliation(s)
- Greg Maguire
- Dept. of Preventative and Medicinal Chemistry, NeoGenesis Inc. And BioRegenerative Sciences Inc, San Diego, CA, USA
| |
Collapse
|
10
|
Bath PM, Coleman CM, Gordon AL, Lim WS, Webb AJ. Nitric oxide for the prevention and treatment of viral, bacterial, protozoal and fungal infections. F1000Res 2021; 10:536. [PMID: 35685687 PMCID: PMC9171293 DOI: 10.12688/f1000research.51270.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Although the antimicrobial potential of nitric oxide (NO) is widely published, it is little used clinically. NO is a key signalling molecule modulating vascular, neuronal, inflammatory and immune responses. Endogenous antimicrobial activity is largely mediated by high local NO concentrations produced by cellular inducible nitric oxide synthase, and by derivative reactive nitrogen oxide species including peroxynitrite and S-nitrosothiols. NO may be taken as dietary substrate (inorganic nitrate, L-arginine), and therapeutically as gaseous NO, and transdermal, sublingual, oral, intranasal and intravenous nitrite or nitrate. Numerous preclinical studies have demonstrated that NO has generic static and cidal activities against viruses (including β-coronaviruses such as SARS-CoV-2), bacteria, protozoa and fungi/yeasts in vitro. Therapeutic effects have been seen in animal models in vivo, and phase II trials have demonstrated that NO donors can reduce microbial infection. Nevertheless, excess NO, as occurs in septic shock, is associated with increased morbidity and mortality. In view of the dose-dependent positive and negative effects of NO, safety and efficacy trials of NO and its donors are needed for assessing their role in the prevention and treatment of infections. Trials should test dietary inorganic nitrate for pre- or post-exposure prophylaxis and gaseous NO or oral, topical or intravenous nitrite and nitrate for treatment of mild-to-severe infections, including due to SARS-CoV-2 (COVID-19). This review summarises the evidence base from in vitro, in vivo and early phase clinical studies of NO activity in viral, bacterial, protozoal and fungal infections.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
- Stroke, Nottingham University Hospitals NHS Trust, Nottingham, Notts, NG7 2UH, UK
| | - Christopher M Coleman
- Division of Infection, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
| | - Adam L Gordon
- Unit of Injury, Inflammation and Recovery Sciences, University of Nottingham, Derby, Derbyshire, DE22 3NE, UK
- NIHR Applied Research Collaboration-East Midlands (ARC-EM), Nottingham, Notts, UK
| | - Wei Shen Lim
- Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Andrew J Webb
- Clinical Pharmacology, School of Cardiovascular Medicine & Sciences, Kings College London British Heart Foundation Centre of Research Excellence, St Thomas' Hospital, London, SE1 7EH, UK
| |
Collapse
|
11
|
Bath PM, Coleman CM, Gordon AL, Lim WS, Webb AJ. Nitric oxide for the prevention and treatment of viral, bacterial, protozoal and fungal infections. F1000Res 2021; 10:536. [PMID: 35685687 PMCID: PMC9171293 DOI: 10.12688/f1000research.51270.1] [Citation(s) in RCA: 2] [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] [Accepted: 06/21/2021] [Indexed: 12/18/2023] Open
Abstract
Although the antimicrobial potential of nitric oxide (NO) is widely published, it is little used clinically. NO is a key signalling molecule modulating vascular, neuronal, inflammatory and immune responses. Endogenous antimicrobial activity is largely mediated by high local NO concentrations produced by cellular inducible nitric oxide synthase, and by derivative reactive nitrogen oxide species including peroxynitrite and S-nitrosothiols. NO may be taken as dietary substrate (inorganic nitrate, L-arginine), and therapeutically as gaseous NO, and transdermal, sublingual, oral, intranasal and intravenous nitrite or nitrate. Numerous preclinical studies have demonstrated that NO has generic static and cidal activities against viruses (including β-coronaviruses such as SARS-CoV-2), bacteria, protozoa and fungi/yeasts in vitro. Therapeutic effects have been seen in animal models in vivo, and phase II trials have demonstrated that NO donors can reduce microbial infection. Nevertheless, excess NO, as occurs in septic shock, is associated with increased morbidity and mortality. In view of the dose-dependent positive and negative effects of NO, safety and efficacy trials of NO and its donors are needed for assessing their role in the prevention and treatment of infections. Trials should test dietary inorganic nitrate for pre- or post-exposure prophylaxis and gaseous NO or oral, topical or intravenous nitrite and nitrate for treatment of mild-to-severe infections, including due to SARS-CoV-2 (COVID-19). This review summarises the evidence base from in vitro, in vivo and early phase clinical studies of NO activity in viral, bacterial, protozoal and fungal infections.
Collapse
Affiliation(s)
- Philip M. Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
- Stroke, Nottingham University Hospitals NHS Trust, Nottingham, Notts, NG7 2UH, UK
| | - Christopher M. Coleman
- Division of Infection, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
| | - Adam L. Gordon
- Unit of Injury, Inflammation and Recovery Sciences, University of Nottingham, Derby, Derbyshire, DE22 3NE, UK
- NIHR Applied Research Collaboration-East Midlands (ARC-EM), Nottingham, Notts, UK
| | - Wei Shen Lim
- Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Andrew J. Webb
- Clinical Pharmacology, School of Cardiovascular Medicine & Sciences, Kings College London British Heart Foundation Centre of Research Excellence, St Thomas' Hospital, London, SE1 7EH, UK
| |
Collapse
|
12
|
Implications of SARS-Cov-2 infection on eNOS and iNOS activity: Consequences for the respiratory and vascular systems. Nitric Oxide 2021; 111-112:64-71. [PMID: 33831567 PMCID: PMC8021449 DOI: 10.1016/j.niox.2021.04.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/18/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023]
Abstract
Symptoms of COVID-19 range from asymptomatic/mild symptoms to severe illness and death, consequence of an excessive inflammatory process triggered by SARS-CoV-2 infection. The diffuse inflammation leads to endothelium dysfunction in pulmonary blood vessels, uncoupling eNOS activity, lowering NO production, causing pulmonary physiological alterations and coagulopathy. On the other hand, iNOS activity is increased, which may be advantageous for host defense, once NO plays antiviral effects. However, overproduction of NO may be deleterious, generating a pro-inflammatory effect. In this review, we discussed the role of endogenous NO as a protective or deleterious agent of the respiratory and vascular systems, the most affected in COVID-19 patients, focusing on eNOS and iNOS roles. We also reviewed the currently available NO therapies and pointed out possible alternative treatments targeting NO metabolism, which could help mitigate health crises in the present and future CoV's spillovers.
Collapse
|
13
|
Garren MR, Ashcraft M, Qian Y, Douglass M, Brisbois EJ, Handa H. Nitric oxide and viral infection: Recent developments in antiviral therapies and platforms. APPLIED MATERIALS TODAY 2021; 22:100887. [PMID: 38620577 PMCID: PMC7718584 DOI: 10.1016/j.apmt.2020.100887] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/11/2020] [Accepted: 11/14/2020] [Indexed: 05/09/2023]
Abstract
Nitric oxide (NO) is a gasotransmitter of great significance to developing the innate immune response to many bacterial and viral infections, while also modulating vascular physiology. The generation of NO from the upregulation of endogenous nitric oxide synthases serves as an efficacious method for inhibiting viral replication in host defense and warrants investigation for the development of antiviral therapeutics. With increased incidence of global pandemics concerning several respiratory-based viral infections, it is necessary to develop broad therapeutic platforms for inhibiting viral replication and enabling more efficient host clearance, as well as to fabricate new materials for deterring viral transmission from medical devices. Recent developments in creating stabilized NO donor compounds and their incorporation into macromolecular scaffolds and polymeric substrates has created a new paradigm for developing NO-based therapeutics for long-term NO release in applications for bactericidal and blood-contacting surfaces. Despite this abundance of research, there has been little consideration of NO-releasing scaffolds and substrates for reducing passive transmission of viral infections or for treating several respiratory viral infections. The aim of this review is to highlight the recent advances in developing gaseous NO, NO prodrugs, and NO donor compounds for antiviral therapies; discuss the limitations of NO as an antiviral agent; and outline future prospects for guiding materials design of a next generation of NO-releasing antiviral platforms.
Collapse
Key Words
- ACE, angiotensin converting enzyme
- AP1, activator protein 1
- COVID-19
- COVID-19, coronavirus disease 2019
- ECMO, extracorporeal membrane oxygenation, FDA, United States Food and Drug Administration
- GNSO, S-nitrosoglutathione
- H1N1, influenza A virus subtype H1N1
- HI, Host Immunology
- HIV, human immunodeficiency virus
- HPV, human papillomavirus
- HSV, herpes simplex virus
- I/R, pulmonary ischemia-reperfusion
- IC50, inhibitory concentration 50
- IFN, interferon
- IFNγ, interferon gamma
- IKK, inhibitor of nuclear factor kappa B kinase
- IRF-1, interferon regulatory factor 1
- Inhalation therapy
- Medical Terminology: ARDS, acute respiratory distress syndrome
- NF-κB, nuclear factor kappa-light-chain enhancer of activated B cells
- NO, nitric oxide
- NOS, nitric oxide synthase
- Nitric Oxide and Related Compounds: eNOS/NOS 3, endothelial nitric oxide synthase
- Nitric oxide
- Other: DNA, deoxyribonucleic acid
- P38-MAPK, P38 mitogen-activated protein kinases
- PAMP, pathogen-associated molecular pattern
- PCV2, porcine circovirus type 2
- PHT, pulmonary hypertension
- PKR, protein kinase R
- RNA, ribonucleic acid
- RNI, reactive nitrogen intermediate
- RSNO, S-nitrosothiol
- SARS, severe acute respiratory syndrome
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SNAP, S-nitroso-N-acetyl-penicillamine
- STAT-1, signal transducer and activator of transcription 1
- Severe acute respiratory distress
- TAK1, transforming growth factor β-activated kinases-1
- TLR, toll-like receptor
- VAP, ventilator associated pneumonia
- Viral infection
- Viruses: CVB3, coxsackievirus
- dsRNA, double stranded (viral) ribonucleic acid
- gNO, gaseous nitric oxide
- iNOS/NOS 2, inducible nitric oxide synthase
- mtALDH, mitochondrial aldehyde dehydrogenase
- nNOS/NOS 1, neuronal nitric oxide synthase
Collapse
Affiliation(s)
- Mark R Garren
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Morgan Ashcraft
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Yun Qian
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Megan Douglass
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Elizabeth J Brisbois
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Hitesh Handa
- School of Chemical, Materials, and Biochemical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| |
Collapse
|
14
|
Guan SP, Seet RCS, Kennedy BK. Does eNOS derived nitric oxide protect the young from severe COVID-19 complications? Ageing Res Rev 2020; 64:101201. [PMID: 33157320 PMCID: PMC7609225 DOI: 10.1016/j.arr.2020.101201] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Aging is the largest risk factors for severity and mortality in adult COVID-19. Severe cases of COVID-19 are related to vascular damage with evidence of direct viral infection in the endothelial cells. Increase risk of COVID-19 death are also highly related to disease with lower vascular Nitric Oxide (NO) level. Vascular viral defence by endothelial nitric oxide synthase (eNOS) derive NO may be the protecting factor for the young. eNOS polymorphism could potentially explain the disparity of COVID-19 mortality between Asian and non-Asian countries.
The COVID-19 pandemic poses an imminent threat to humanity, especially to the elderly. The molecular mechanisms underpinning the age-dependent disparity for disease progression is not clear. COVID-19 is both a respiratory and a vascular disease in severe patients. The damage endothelial system provides a good explanation for the various complications seen in COVID-19 patients. These observations lead us to suspect that endothelial cells are a barrier that must be breached before progression to severe disease. Endothelial intracellular defences are largely dependent of the activation of the interferon (IFN) system. Nevertheless, low type I and III IFNs are generally observed in COVID-19 patients suggesting that other intracellular viral defence systems are also activated to protect the young. Intriguingly, Nitric oxide (NO), which is the main intracellular antiviral defence, has been shown to inhibit a wide array of viruses, including SARS-CoV-1. Additionally, the increased risk of death with diseases that have underlying endothelial dysfunction suggest that endothelial NOS-derived nitric oxide could be the main defence mechanism. NO decreases dramatically in the elderly, the hyperglycaemic and the patients with low levels of vitamin D. However, eNOS derived NO occurs at low levels, unless it is during inflammation and co-stimulated by bradykinin. Regrettably, the bradykinin-induced vasodilation also progressively declines with age, thereby decreasing anti-viral NO production as well. Intriguingly, the inverse correlation between the percentage of WT eNOS haplotype and death per 100K population could potentially explain the disparity of COVID-19 mortality between Asian and non-Asian countries. These changes with age, low bradykinin and NO, may be the fundamental reasons that intracellular innate immunity declines with age leading to more severe COVID-19 complications.
Collapse
|
15
|
Barh D, Tiwari S, Weener ME, Azevedo V, Góes-Neto A, Gromiha MM, Ghosh P. Multi-omics-based identification of SARS-CoV-2 infection biology and candidate drugs against COVID-19. Comput Biol Med 2020; 126:104051. [PMID: 33131530 PMCID: PMC7547373 DOI: 10.1016/j.compbiomed.2020.104051] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/24/2022]
Abstract
SARS-CoV-2 has ushered a global pandemic with no effective drug being available at present. Although several FDA-approved drugs are currently under clinical trials for drug repositioning, there is an on-going global effort for new drug identification. In this paper, using multi-omics (interactome, proteome, transcriptome, and bibliome) data and subsequent integrated analysis, we present the biological events associated with SARS-CoV-2 infection and identify several candidate drugs against this viral disease. We found that: (i) Interactome-based infection pathways differ from the other three omics-based profiles. (ii) Viral process, mRNA splicing, cytokine and interferon signaling, and ubiquitin mediated proteolysis are important pathways in SARS-CoV-2 infection. (iii) SARS-CoV-2 infection also shares pathways with Influenza A, Epstein-Barr virus, HTLV-I, Measles, and Hepatitis virus. (iv) Further, bacterial, parasitic, and protozoan infection pathways such as Tuberculosis, Malaria, and Leishmaniasis are also shared by this virus. (v) A total of 50 candidate drugs, including the prophylaxis agents and pathway specific inhibitors are identified against COVID-19. (vi) Betamethasone, Estrogen, Simvastatin, Hydrocortisone, Tositumomab, Cyclosporin A etc. are among the important drugs. (vii) Ozone, Nitric oxide, plasma components, and photosensitizer drugs are also identified as possible therapeutic candidates. (viii) Curcumin, Retinoic acids, Vitamin D, Arsenic, Copper, and Zinc may be the candidate prophylaxis agents. Nearly 70% of our identified agents are previously suggested to have anti-COVID-19 effects or under clinical trials. Among our identified drugs, the ones that are not yet tested, need validation with caution while an appropriate drug combination from these candidate drugs along with a SARS-CoV-2 specific antiviral agent is needed for effective COVID-19 management.
Collapse
Affiliation(s)
- Debmalya Barh
- Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, WB, India.
| | - Sandeep Tiwari
- Laboratório de Genética Celular e Molecular, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marianna E Weener
- Clinical Research Center, Oftalmic, CRO, 119334, Bardina Str.22/4, Moscow, Russia
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Aristóteles Góes-Neto
- Laboratório de Biologia Molecular e Computacional de Fungos, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT-M), Chennai, 600036, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, 23284, USA
| |
Collapse
|
16
|
Al-Hatamleh MAI, Hatmal MM, Sattar K, Ahmad S, Mustafa MZ, Bittencourt MDC, Mohamud R. Antiviral and Immunomodulatory Effects of Phytochemicals from Honey against COVID-19: Potential Mechanisms of Action and Future Directions. Molecules 2020; 25:E5017. [PMID: 33138197 PMCID: PMC7672575 DOI: 10.3390/molecules25215017] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
The new coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has recently put the world under stress, resulting in a global pandemic. Currently, there are no approved treatments or vaccines, and this severe respiratory illness has cost many lives. Despite the established antimicrobial and immune-boosting potency described for honey, to date there is still a lack of evidence about its potential role amid COVID-19 outbreak. Based on the previously explored antiviral effects and phytochemical components of honey, we review here evidence for its role as a potentially effective natural product against COVID-19. Although some bioactive compounds in honey have shown potential antiviral effects (i.e., methylglyoxal, chrysin, caffeic acid, galangin and hesperidinin) or enhancing antiviral immune responses (i.e., levan and ascorbic acid), the mechanisms of action for these compounds are still ambiguous. To the best of our knowledge, this is the first work exclusively summarizing all these bioactive compounds with their probable mechanisms of action as antiviral agents, specifically against SARS-CoV-2.
Collapse
Affiliation(s)
- Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia; (M.A.I.A.-H.); (S.A.)
| | - Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, Zarqa 13133, Jordan;
| | - Kamran Sattar
- Department of Medical Education, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia;
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia; (M.A.I.A.-H.); (S.A.)
| | - Mohd Zulkifli Mustafa
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
- Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Marcelo De Carvalho Bittencourt
- Université de Lorraine, CNRS, UMR 7365, IMoPA, F-54000 Nancy, France;
- Université de Lorraine, CHRU-Nancy, Laboratoire d’Immunologie, F-54000 Nancy, France
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia; (M.A.I.A.-H.); (S.A.)
- Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| |
Collapse
|
17
|
Heck T, Ludwig M, Frizzo M, Rasia-Filho A, Homem de Bittencourt PI. Suppressed anti-inflammatory heat shock response in high-risk COVID-19 patients: lessons from basic research (inclusive bats), light on conceivable therapies. Clin Sci (Lond) 2020; 134:1991-2017. [PMID: 32749472 PMCID: PMC7403894 DOI: 10.1042/cs20200596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/05/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
The major risk factors to fatal outcome in COVID-19 patients, i.e., elderliness and pre-existing metabolic and cardiovascular diseases (CVD), share in common the characteristic of being chronic degenerative diseases of inflammatory nature associated with defective heat shock response (HSR). The molecular components of the HSR, the principal metabolic pathway leading to the physiological resolution of inflammation, is an anti-inflammatory biochemical pathway that involves molecular chaperones of the heat shock protein (HSP) family during homeostasis-threatening stressful situations (e.g., thermal, oxidative and metabolic stresses). The entry of SARS coronaviruses in target cells, on the other hand, aggravates the already-jeopardized HSR of this specific group of patients. In addition, cellular counterattack against virus involves interferon (IFN)-mediated inflammatory responses. Therefore, individuals with impaired HSR cannot resolve virus-induced inflammatory burst physiologically, being susceptible to exacerbated forms of inflammation, which leads to a fatal "cytokine storm". Interestingly, some species of bats that are natural reservoirs of zoonotic viruses, including SARS-CoV-2, possess an IFN-based antiviral inflammatory response perpetually activated but do not show any sign of disease or cytokine storm. This is possible because bats present a constitutive HSR that is by far (hundreds of times) more intense and rapid than that of human, being associated with a high core temperature. Similarly in humans, fever is a physiological inducer of HSR while antipyretics, which block the initial phase of inflammation, impair the resolution phase of inflammation through the HSR. These findings offer a rationale for the reevaluation of patient care and fever reduction in SARS, including COVID-19.
Collapse
Affiliation(s)
- Thiago Gomes Heck
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS), Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
| | - Mirna Stela Ludwig
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS), Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
| | - Matias Nunes Frizzo
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS), Regional University of Northwestern Rio Grande do Sul State (UNIJUI), Ijuí, RS, 98700-000 Brazil
| | - Alberto Antonio Rasia-Filho
- Federal University of Health Sciences of Porto Alegre (UFCSPA), Graduate Program in Biosciences, Porto Alegre, RS, 90050-170 Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90050-170 Brazil
| |
Collapse
|
18
|
Tatoyan MR, Izmailyan RA, Semerjyan AB, Karalyan NY, Sahakyan CT, Mkrtchyan GL, Ghazaryan HK, Arzumanyan HH, Semerjyan ZB, Karalova EM, Karalyan ZA. Patterns of alveolar macrophage activation upon attenuated and virulent African swine fever viruses in vitro. Comp Immunol Microbiol Infect Dis 2020; 72:101513. [PMID: 32569898 DOI: 10.1016/j.cimid.2020.101513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/09/2020] [Accepted: 06/14/2020] [Indexed: 12/01/2022]
Abstract
The pattern of porcine alveolar macrophage (AM) activation upon classical stimuli of two strains of African swine fever (ASF) viruses, an attenuated ASFV-BA71V and virulent ASFV-Georgia2007 were investigated. In an in vitro experiment ASFV-Georgia2007-infected AM showed M1 polarization pattern different from the one induced by classical stimuli. Altered morphology, appearance of binuclear cells, decreased synthesis of IFN-alpha as well as IFN-epsilon was observed compared with attenuated ASFV-BA71V, and decreased synthesis of IFN-omega compared with intact cells. However, CD68 level did not significantly differ between alveolar macrophage populations infected by ASFV-Georgia2007 and control group, while both LPS/IFN-gamma stimulation and non-pathogenic ASFV-BA71V virus increased the level of CD68 soluble receptor. AM infection with ASFV-Georgia2007 resulted in remarkable DNA proliferation whereas LPS/IFN-gamma and ASFV-BA71V induced less expressed DNA proliferation in activated cells. The higher value of nitric oxide was obvious in the cells infected with ASFV-BA71V, compared to ASFV-Georgia2007 and LPS/IFN-gamma activated cells. In conclusion, pattern of activation of alveolar macrophages induced by ASFV-Georgia2007 virus differs from the one expressed in LPS/IFN-gamma- and ASFV-BA71V-activated cells. ASFV-BA71V and LPS/IFN-gamma share similar antiviral response of porcine AM. Therefore we assume that wild type virulent ASFV can partially down regulate antiviral response of AM and conclude that evolutionary decrease of virulence in ASFV is related to alterations of control of the host cell antiviral response.
Collapse
Affiliation(s)
| | - Roza A Izmailyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia
| | | | | | | | | | - Hovsep K Ghazaryan
- Laboratory of Human Genomics and Immunomics, Institute of Molecular Biology of NAS RA, Yerevan, Armenia
| | - Hranush H Arzumanyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia
| | - Zara B Semerjyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia; Experimental Laboratory, Yerevan State Medical University, Yerevan, Armenia
| | - Elena M Karalova
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia; Experimental Laboratory, Yerevan State Medical University, Yerevan, Armenia
| | - Zaven A Karalyan
- Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, Yerevan, Armenia; Yerevan State Medical University, Yerevan, Armenia.
| |
Collapse
|
19
|
Karunakaran KB, Balakrishnan N, Ganapathiraju M. Potentially repurposable drugs for COVID-19 identified from SARS-CoV-2 Host Protein Interactome. RESEARCH SQUARE 2020:rs.3.rs-30363. [PMID: 32702734 PMCID: PMC7336709 DOI: 10.21203/rs.3.rs-30363/v1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We previously presented the protein-protein interaction network - the 'HoP' or the host protein interactome - of 332 host proteins that were identified to interact with 27 nCoV19 viral proteins by Gordon et al. Here, we studied drugs targeting the proteins in this interactome to identify whether any of them may potentially be repurposable against SARS-CoV-2. We studied each of the drugs using the BaseSpace Correlation Engine and identified those that induce gene expression profiles negatively correlated with SARS-associated expression profile. This analysis resulted in 20 drugs whose differential gene expression (drug versus normal) had an anti-correlation with differential expression for SARS (viral infection versus normal). These included drugs that were already being tested for their clinical activity against SARS-CoV-2, those with proven activity against SARS-CoV/MERS-CoV, broad-spectrum antiviral drugs, and those identified/prioritized by other computational re-purposing studies. In summary, our integrated computational analysis of the HoP interactome in conjunction with drug-induced transcriptomic data resulted in drugs that may be repurposable for COVID-19.
Collapse
|
20
|
Boylan M, O’Brien MB, Beynon C, Meade KG. 1,25(OH)D vitamin D promotes NOS2 expression in response to bacterial and viral PAMPs in primary bovine salivary gland fibroblasts. Vet Res Commun 2020; 44:83-88. [DOI: 10.1007/s11259-020-09775-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/30/2020] [Indexed: 12/21/2022]
|
21
|
Traboulsi H, Cherian M, Abou Rjeili M, Preteroti M, Bourbeau J, Smith BM, Eidelman DH, Baglole CJ. Inhalation Toxicology of Vaping Products and Implications for Pulmonary Health. Int J Mol Sci 2020; 21:E3495. [PMID: 32429092 PMCID: PMC7278963 DOI: 10.3390/ijms21103495] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/28/2020] [Accepted: 05/12/2020] [Indexed: 12/17/2022] Open
Abstract
E-cigarettes have a liquid that may contain flavors, solvents, and nicotine. Heating this liquid generates an aerosol that is inhaled into the lungs in a process commonly referred to as vaping. E-cigarette devices can also contain cannabis-based products including tetrahydrocannabinol (THC), the psychoactive component of cannabis (marijuana). E-cigarette use has rapidly increased among current and former smokers as well as youth who have never smoked. The long-term health effects are unknown, and emerging preclinical and clinical studies suggest that e-cigarettes may not be harmless and can cause cellular alterations analogous to traditional tobacco smoke. Here, we review the historical context and the components of e-cigarettes and discuss toxicological similarities and differences between cigarette smoke and e-cigarette aerosol, with specific reference to adverse respiratory outcomes. Finally, we outline possible clinical disorders associated with vaping on pulmonary health and the recent escalation of acute lung injuries, which led to the declaration of the vaping product use-associated lung injury (EVALI) outbreak. It is clear there is much about vaping that is not understood. Consequently, until more is known about the health effects of vaping, individual factors that need to be taken into consideration include age, current and prior use of combustible tobacco products, and whether the user has preexisting lung conditions such as asthma and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Hussein Traboulsi
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
| | - Mathew Cherian
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
| | - Mira Abou Rjeili
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Matthew Preteroti
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jean Bourbeau
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Benjamin M. Smith
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
| | - Carolyn J. Baglole
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
22
|
Vats A, Gautam D, Maharana J, Singh Chera J, Kumar S, Rout PK, Werling D, De S. Poly I:C stimulation in-vitro as a marker for an antiviral response in different cell types generated from Buffalo (Bubalus bubalis). Mol Immunol 2020; 121:136-143. [PMID: 32200171 DOI: 10.1016/j.molimm.2020.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/06/2020] [Accepted: 03/09/2020] [Indexed: 01/17/2023]
Abstract
The innate immune system is activated upon virus invasion of a host cell by recognizing viral component, such as dsRNA through specific receptors, resulting in the production of type- I IFNs, which confer an antiviral state within the invaded as well as surrounding cells. In the present study, fibroblast, monocyte and macrophage cells derived from water Buffalo (Bubalus bubalis) were exposed to a synthetic dsRNA analogue, poly I:C to mimic viral invasion in each cell type. Recognition of poly I:C through cytosolic helicase receptors RIG-I and MDA5 molecule lead to the activation of the RLR pathway, subsequently activating the MAVS-IRF3/7 cascade and the production of antiviral effector molecule like IFNβ and ISGs. Within the different cell types, we identified variability in RLR receptor and IFNβ expression after poly I:C administration. Fibroblasts responded quickly and strongly with IFNβ production, followed by macrophages and monocytes. Despite absolute expression variability among different cell types the expression trend of RLRs pathway genes were similar. Length of poly I:C molecule also influence IFNβ expression in response of RLR pathway. Short (LMW) poly I:C induce stronger IFN-β expression in myeloid (macrophage and monocyte) cells. In contrast long (HMW) poly I:C preferably elicit higher IFNβ expression in non-myeloid (fibroblast) cell. Therefore, MDA5 and RIG-1 plays an indispensable role in eliciting antiviral response in non- immune (fibroblast) host cell. Thus, stimulation of RLR pathway with suitable and potentially cell-type specific agonist molecules successfully elicit antiviral state in the host animal, with fibroblasts conferring a stronger antiviral state compared with the monocytes and macrophages.
Collapse
Affiliation(s)
- Ashutosh Vats
- Animal Genomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Devika Gautam
- Animal Genomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Jitendra Maharana
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Jatinder Singh Chera
- Animal Genomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Sushil Kumar
- Animal Genomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Pramod K Rout
- ICAR-Central Institute for Research on Goats, Mathura, Uttar Pradesh, India
| | - Dirk Werling
- The Royal Veterinary College, University of London, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, UK
| | - Sachinandan De
- Animal Genomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India.
| |
Collapse
|
23
|
Daliri EB, Choi S, Cho B, Jo HY, Kim S, Chelliah R, Rubab M, Kim J, Oh H, Lee O, Oh D. Biological activities of a garlic- Cirsium setidens Nakai blend fermented with Leuconostoc mesenteroides. Food Sci Nutr 2019; 7:2024-2032. [PMID: 31289650 PMCID: PMC6593477 DOI: 10.1002/fsn3.1032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 01/17/2023] Open
Abstract
In this study, we investigated the antioxidant- and immune-stimulating activities of various garlic-Cirsium setidens Nakai blends (fermented and unfermented). The levels of S-allyl cysteine increased by 2.5 times while pectolinarigenin (an anti-inflammatory compound) increased about six times (from 1.1 ± 0.04 mg/g to 6.70 ± 0.12 mg/g) after the garlic-Cirsium setidens Nakai (80:20%, respectively) blend (S4) was fermented with Leuconostoc mesenteroides KCTC 13302. The ferric reducing ability and DPPH radical scavenging activities of all the samples increased significantly after fermentation. Ethanolic extracts of the fermented samples significantly enhanced RAW 264.7 macrophage proliferation in a dose-dependent manner and induced nitric oxide production. Among the samples, S6 and S8 stimulated the highest levels of nitric oxide (NO) production. S6 significantly induced proinflammatory cytokines TNF-α and IL-1β as well as an anti-inflammatory cytokine IL-10 relative to control. Since the resolution of an infection would require a harmonized interplay of proinflammatory factors and anti-inflammatory cytokines, consumption of S6 could be helpful in promoting health.
Collapse
Affiliation(s)
| | - Sun‐Il Choi
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Bong‐Yeon Cho
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Hyeon Yeong Jo
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Se‐Hun Kim
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Ramachandran Chelliah
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Momna Rubab
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Joong‐Hark Kim
- Erom R&D CenterErom Co., LtdChuncheon‐si, Gangwon‐doSouth Korea
| | - Hyun‐Taek Oh
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
- Erom R&D CenterErom Co., LtdChuncheon‐si, Gangwon‐doSouth Korea
| | - Ok‐Hwan Lee
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| | - Deog‐Hwan Oh
- Department of Food Science and BiotechnologyKangwon National UniversityChuncheonSouth Korea
| |
Collapse
|
24
|
Immunomodulating dose of levamisole stimulates innate immune response and prevents intestinal damage in porcine rotavirus diarrhea: a restricted-randomized, single-blinded, and placebo-controlled clinical trial. Trop Anim Health Prod 2019; 51:1455-1465. [DOI: 10.1007/s11250-019-01833-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/23/2019] [Indexed: 11/25/2022]
|
25
|
Xue T, Li J, Liu C. A radical form of nitric oxide inhibits porcine circovirus type 2 replication in vitro. BMC Vet Res 2019; 15:47. [PMID: 30709350 PMCID: PMC6359798 DOI: 10.1186/s12917-019-1796-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/24/2019] [Indexed: 11/29/2022] Open
Abstract
Background Porcine circovirus type 2 (PCV2) is the causal agent of postweaning multisystemic wasting syndrome (PMWS), causing large economical losses of the global swine industry. Nitric oxide (NO), as an important signaling molecule, has antiviral activity on some viruses. To date, there is little information on the role of NO during PCV2 infection. Results We used indirect fluorescence assay (IFA), TCID50, real-time RT-qPCR and western blot assay to reveal the role of NO in restricting PCV2 replication. PCV2 replication was inhibited by a form of NO, NO•, whereas PCV2 was not susceptible to another form of NO, NO+. Conclusion Our findings indicate that the form of NO• has a potential role in the fight against PCV2 infection.
Collapse
Affiliation(s)
- Tao Xue
- School of Pharmacy, Linyi University, Linyi, 276000, China
| | - Jizong Li
- School of Pharmacy, Linyi University, Linyi, 276000, China.,Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences,Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Chuanmin Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences,Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China.
| |
Collapse
|
26
|
Rios-Ibarra CP, Torres-De La Cruz V, Ochoa-Ruiz AG, Rivas-Estilla AM. Quantification of nitric oxide by high-performance liquid chromatography-fluorometric method in subgenomic hepatitis C virus-replicon expressing Huh7 cells upon treatment with acetylsalicylic acid. Exp Ther Med 2018; 16:2621-2626. [PMID: 30186494 PMCID: PMC6122491 DOI: 10.3892/etm.2018.6515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
As nitric oxide (NO) expression levels are lower in hepatocytes compared with other cell types, it is difficult to quantify this compound via Griess assay. The aim of the present study was to quantify NO concentration in the cell culture medium from a subgenomic hepatitis C virus (HCV)-replicon expressing Huh-7 cell system using a high-performance liquid chromatography (HPLC)-fluorescence detector in the presence or absence of acetylsalicylic acid (ASA) treatment. HCV-replicon cells were incubated with ASA (4 mM) for 24, 48 and 72 h. Thereafter, the medium was collected to measure nitrites (NO2-) as an indirect indicator of NO levels using diaminonaphtalene as a derivate agent. NO levels were significantly higher (1.7-fold) in Huh-7 replicon cells treated with ASA (72 h post-treatment) than untreated cells (P<0.05); NO inhibitor reduced ~30% the level of NO in Huh-7 replicon cells treated with ASA (48 h post-treatment; P<0.05). The findings suggested that the HPLC-fluorescence method provided an accurate and efficient measurement of NO production in Huh-7-HCV-replicon cells culture medium.
Collapse
Affiliation(s)
- Clara Patricia Rios-Ibarra
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo León 64460, México
- Department of Bioengineering, Tecnologico de Monterrey, Campus Guadalajara, Zapopan, Jalisco 45138, México
| | - Victor Torres-De La Cruz
- Biomedical Research Center, Northeast Mexican Social Security Institute, Monterrey, Nuevo León 64720, México
| | - Andrea Gabriela Ochoa-Ruiz
- Department of Bioengineering, Tecnologico de Monterrey, Campus Guadalajara, Zapopan, Jalisco 45138, México
| | - Ana María Rivas-Estilla
- Department of Biochemistry and Molecular Medicine, School of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo León 64460, México
| |
Collapse
|
27
|
Who's Driving? Human Cytomegalovirus, Interferon, and NFκB Signaling. Viruses 2018; 10:v10090447. [PMID: 30134546 PMCID: PMC6163874 DOI: 10.3390/v10090447] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 12/16/2022] Open
Abstract
As essential components of the host's innate immune response, NFκB and interferon signaling are critical determinants of the outcome of infection. Over the past 25 years, numerous Human Cytomegalovirus (HCMV) genes have been identified that antagonize or modulate the signaling of these pathways. Here we review the biology of the HCMV factors that alter NFκB and interferon signaling, including what is currently known about how these viral genes contribute to infection and persistence, as well as the major outstanding questions that remain.
Collapse
|
28
|
Ahmed-Hassan H, Abdul-Cader MS, Ahmed Sabry M, Hamza E, Sharif S, Nagy E, Abdul-Careem MF. Double-Stranded Ribonucleic Acid-Mediated Antiviral Response Against Low Pathogenic Avian Influenza Virus Infection. Viral Immunol 2018; 31:433-446. [PMID: 29813000 DOI: 10.1089/vim.2017.0142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptor (TLR)3 signaling pathway is known to induce type 1 interferons (IFNs) and proinflammatory mediators leading to antiviral response against many viral infections. Double-stranded ribonucleic acid (dsRNA) has been shown to act as a ligand for TLR3 and, as such, has been a focus as a potential antiviral agent in many host-viral infection models. Yet, its effectiveness and involved mechanisms as a mediator against low pathogenic avian influenza virus (LPAIV) have not been investigated adequately. In this study, we used avian fibroblasts to verify whether dsRNA induces antiviral response against H4N6 LPAIV and clarify whether type 1 IFNs and proinflammatory mediators such as interleukin (IL)-1β are contributing to the dsRNA-mediated antiviral response against H4N6 LPAIV. We found that dsRNA induces antiviral response in avian fibroblasts against H4N6 LPAIV infection. The treatment of avian fibroblasts with dsRNA increases the expressions of TLR3, IFN-α, IFN-β, and IL-1β. We also confirmed that this antiviral response elicited against H4N6 LPAIV infection correlates, but is not attributable to type 1 IFNs or IL-1β. Our findings imply that the TLR3 ligand, dsRNA, can elicit antiviral response in avian fibroblasts against LPAIV infection, highlighting potential value of dsRNA as an antiviral agent against LPAIV infections. However, further investigations are required to determine the potential role of other innate immune mediators or combination of the tested cytokines in the dsRNA-mediated antiviral response against H4N6 LPAIV infection.
Collapse
Affiliation(s)
- Hanaa Ahmed-Hassan
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada .,2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Mohamed Sarjoon Abdul-Cader
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada
| | - Maha Ahmed Sabry
- 2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Eman Hamza
- 2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Shayan Sharif
- 3 Department of Pathobiology, University of Guelph , Guelph, Ontario, Canada
| | - Eva Nagy
- 3 Department of Pathobiology, University of Guelph , Guelph, Ontario, Canada
| | - Mohamed Faizal Abdul-Careem
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada
| |
Collapse
|
29
|
S-nitrosylation drives cell senescence and aging in mammals by controlling mitochondrial dynamics and mitophagy. Proc Natl Acad Sci U S A 2018; 115:E3388-E3397. [PMID: 29581312 DOI: 10.1073/pnas.1722452115] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
S-nitrosylation, a prototypic redox-based posttranslational modification, is frequently dysregulated in disease. S-nitrosoglutathione reductase (GSNOR) regulates protein S-nitrosylation by functioning as a protein denitrosylase. Deficiency of GSNOR results in tumorigenesis and disrupts cellular homeostasis broadly, including metabolic, cardiovascular, and immune function. Here, we demonstrate that GSNOR expression decreases in primary cells undergoing senescence, as well as in mice and humans during their life span. In stark contrast, exceptionally long-lived individuals maintain GSNOR levels. We also show that GSNOR deficiency promotes mitochondrial nitrosative stress, including excessive S-nitrosylation of Drp1 and Parkin, thereby impairing mitochondrial dynamics and mitophagy. Our findings implicate GSNOR in mammalian longevity, suggest a molecular link between protein S-nitrosylation and mitochondria quality control in aging, and provide a redox-based perspective on aging with direct therapeutic implications.
Collapse
|
30
|
Ahmed-Hassan H, Abdul-Cader MS, De Silva Senapathi U, Sabry MA, Hamza E, Nagy E, Sharif S, Abdul-Careem MF. Potential mediators of in ovo delivered double stranded (ds) RNA-induced innate response against low pathogenic avian influenza virus infection. Virol J 2018. [PMID: 29530062 PMCID: PMC5848551 DOI: 10.1186/s12985-018-0954-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Toll like receptor (TLR) 3 is a critically important innate pattern recognizing receptor that senses many viral infections. Although, it has been shown that double stranded (ds) RNA can be used for the stimulation of TLR3 signaling pathway in a number of host-viral infection models, it’s effectiveness as an antiviral agent against low pathogenic avian influenza virus (LPAIV) needs further investigation. Methods In this study, first, we delivered TLR3 ligand, dsRNA, in ovo at embryo day (ED)18 since in ovo route is routinely used for vaccination against poultry viral and parasitic infections and infected with H4N6 LPAIV 24-h post-treatment. A subset of in ovo dsRNA treated and control groups were observed for the expressions of TLR3 and type I interferon (IFN)s, mRNA expression of interleukin (IL)-1β and macrophage recruitment coinciding with the time of H4N6 LPAIV infection (24 h post-treatment). Additionally, Day 1 chickens were given dsRNA intra-tracheally along with a control group and a subset of chickens were infected with H4N6 LPAIV 24-h post-treatment whereas the rest of the animals were observed for macrophage and type 1 IFN responses coinciding with the time of viral infection. Results Our results demonstrate that the pre-hatch treatment of eggs with dsRNA reduces H4N6 replication in lungs. Further studies revealed that in ovo delivery of dsRNA increases TLR3 expression, type I IFN production and number of macrophages in addition to mRNA expression of IL-1β in lung 24-h post-treatment. The same level of induction of innate response was not evident in the spleen. Moreover, we discovered that dsRNA elicits antiviral response against LPAIV correlating with type I IFN activity in macrophages in vitro. Post-hatch, we found no difference in H4N6 LPAIV genome loads between dsRNA treated and control chickens although we observed higher macrophage recruitment and IFN-β response coinciding with the time of viral infection. Conclusions Our findings imply that the TLR3 ligand, dsRNA has antiviral activity in ovo and in vitro but not in chickens post-hatch and dsRNA-mediated innate host response is characterized by macrophage recruitment and expressions of TLR3 and type 1 IFNs.
Collapse
Affiliation(s)
- Hanaa Ahmed-Hassan
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.,Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed Sarjoon Abdul-Cader
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Upasama De Silva Senapathi
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Maha Ahmed Sabry
- Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Eman Hamza
- Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Eva Nagy
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Mohamed Faizal Abdul-Careem
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
31
|
Liu C, Wen L, Xiao Q, He K. Nitric oxide-generating compound GSNO suppresses porcine circovirus type 2 infection in vitro and in vivo. BMC Vet Res 2017; 13:59. [PMID: 28222773 PMCID: PMC5320642 DOI: 10.1186/s12917-017-0976-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/15/2017] [Indexed: 01/25/2023] Open
Abstract
Background Nitric oxide (NO), an important signaling molecule with biological functions, has antimicrobial activity against a variety of pathogens including viruses. To our knowledge, little information is available about the regulatory effect of NO on porcine circovirus type 2 (PCV2) infection. This study was conducted to investigate the antiviral activity of NO generated from S-nitrosoglutathione (GSNO), during PCV2 infection of PK-15 cells and BALB/c mice. Results GSNO released considerable NO in the culture medium of PK-15 cells, and NO was scavenged by its scavenger hemoglobin (Hb) in a dose-dependent manner. NO strongly inhibited PCV2 replication in PK-15 cells, and the antiviral effect was reversed by Hb. An in vivo assay indicated that GSNO treatment reduced the progression of PCV2 infection in mice, evident as reductions in the percentages of PCV2-positive sera and tissue samples and in the viral DNA copies in serum samples. GSNO also improved the growth performance and immune organs (spleens and thymuses) of the PCV2-infected mice to some degree. Conclusions Our data demonstrate that the NO-generating compound GSNO suppresses PCV2 infection in PK-15 cells and BALB/c mice, indicating that NO and its donor, GSNO, have potential value as antiviral drugs against PCV2 infection.
Collapse
Affiliation(s)
- Chuanmin Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,Key laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,National Center for Engineering Research of Veterinary Bio-products, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 Wen-Hui East Road, Hanjiang District, Yangzhou, 225009, China
| | - Libin Wen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,Key laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,National Center for Engineering Research of Veterinary Bio-products, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 Wen-Hui East Road, Hanjiang District, Yangzhou, 225009, China
| | - Qi Xiao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,Key laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,National Center for Engineering Research of Veterinary Bio-products, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 Wen-Hui East Road, Hanjiang District, Yangzhou, 225009, China
| | - Kongwang He
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China. .,Key laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China. .,National Center for Engineering Research of Veterinary Bio-products, 50 Zhong-ling Street, Xuanwu District, Nanjing, 210014, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 Wen-Hui East Road, Hanjiang District, Yangzhou, 225009, China.
| |
Collapse
|
32
|
Mohamed SH, Mahmoud NF, Mohamed AF, Kotb NS. Post HCV Infection Due to MX Gene Stimulation Produced Post Treatment with Imported and Locally Produced Egyptian Biosimilar IFN. Asian Pac J Cancer Prev 2016; 16:5635-41. [PMID: 26320428 DOI: 10.7314/apjcp.2015.16.14.5635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cirrhosis is regarded as a possible end stage of many liver diseases, including viral infection. It occurs when healthy liver tissue becomes damaged and is replaced by scar tissue and finally may lead to hepatocellular carcinoma. Interferons (IFNs)are two general categories, type I and II. Type I includes one beta interferon and over 20 different alpha interferons. Alpha interferons are very similar in how they work, interacting with other proteins on cells like receptors. The main objective of this study was to compare Mx gene productivity post different cell line treatment with imported and Egyptian biosimilar locally produced IFNs, as well as the efficacy of those tested IFNs. Also, an assessment was made of sensitivity of different cell lines as alternatives to that recommended for evaluation of antiviral activity. MATERIALS AND METHODS Different cell lines (Vero, MDBK and Wish) were employed to evaluate cytotoxicity using the MTT assay. Antiviral activity was evaluated compared with standard IFN against VSV, Indiana strain -156, on tested rh-IFNs (imported; innovated and Egyptian biosimilar locally produced IFNs) in the pre-treated cell lines previously mentioned. The virus was propagated in the Wish cell line as recommended. Finally we estimated up-regulation of the Mx gene as a biomarker. RESULTS Data recorded revealed that test IFNs were safe in test cell lines. Viability was around 100%. Locally tested interferon did not realize the international potency limits, while the imported one was accepted compared with the standard IFN. These results were the same either using infectivity titer reduction assay or crystal violet staining of residual non- infected cells. Mx protein production was cell type related and confirmed by the detected Mx gene expressed in imported and locally produced IFN pre-treated cell lines. The expression of the gene was arranged in the order of Vero> wish > MDBK for the imported IFN, while for the Egyptian biosimillar locally produced one it was MDBK>Vero> wish. With regard to the antiviral activity there was a significant difference of imported IFN potency compared with the locally produced IFN (P<0.05), the IFN potential (antiviral activity) was not cell line related and showed non-significant difference for each separate product. CONCLUSIONS Vero cells can be used as an alternative cell line for evaluation of IFN potency in case of unavailable USP recommended cell lines. Alternative potency evaluation assay could be used and proved significant difference in IFN potency in case of local and imported agents. Evaluation of antiviral activity could be used in parallel to viral infectivity reduction assay for better accuracy. Mx gene can be used as a marker for IFN potential.
Collapse
Affiliation(s)
- Shereen H Mohamed
- National Organization for Research and Control of Biological (NORCB), Dokki, Giza, Egypt E-mail :
| | | | | | | |
Collapse
|
33
|
Abdul-Cader MS, Amarasinghe A, Abdul-Careem MF. Activation of toll-like receptor signaling pathways leading to nitric oxide-mediated antiviral responses. Arch Virol 2016; 161:2075-86. [PMID: 27233799 PMCID: PMC7087267 DOI: 10.1007/s00705-016-2904-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs), well-characterized pattern-recognizing receptors of the innate arm of the immune system, are vital in detecting pathogen-associated molecular patterns (PAMPs). The TLR-PAMP interaction initiates an intracellular signaling cascade, predominantly culminating in upregulation of antiviral components, including inducible nitric oxide synthase (iNOS). After activation, various TLR pathways can promote iNOS production via the myeloid differentiation primary response-88 (MyD-88) adapter protein. Subsequently, iNOS facilitates production of nitric oxide (NO), a highly reactive and potent antiviral molecule that can inhibit replication of RNA and DNA viruses. Furthermore, NO can diffuse freely across cell membranes and elicit antiviral mechanisms in various ways, including direct and indirect damage to viral genomes. This review emphasizes current knowledge of NO-mediated antiviral responses elicited after activation of TLR signaling pathways.
Collapse
Affiliation(s)
- Mohamed Sarjoon Abdul-Cader
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Center 2C58, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Aruna Amarasinghe
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Center 2C58, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Mohamed Faizal Abdul-Careem
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Center 2C58, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
34
|
Zhang L, Xiang W, Wang G, Yan Z, Zhu Z, Guo Z, Sengupta R, Chen AF, Loughran PA, Lu B, Wang Q, Billiar TR. Interferon β (IFN-β) Production during the Double-stranded RNA (dsRNA) Response in Hepatocytes Involves Coordinated and Feedforward Signaling through Toll-like Receptor 3 (TLR3), RNA-dependent Protein Kinase (PKR), Inducible Nitric Oxide Synthase (iNOS), and Src Protein. J Biol Chem 2016; 291:15093-107. [PMID: 27226571 DOI: 10.1074/jbc.m116.717942] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 12/19/2022] Open
Abstract
The sensing of double-stranded RNA (dsRNA) in the liver is important for antiviral defenses but can also contribute to sterile inflammation during liver injury. Hepatocytes are often the target of viral infection and are easily injured by inflammatory insults. Here we sought to establish the pathways involved in the production of type I interferons (IFN-I) in response to extracellular poly(I:C), a dsRNA mimetic, in hepatocytes. This was of interest because hepatocytes are long-lived and, unlike most immune cells that readily die after activation with dsRNA, are not viewed as cells with robust antimicrobial capacity. We found that poly(I:C) leads to rapid up-regulation of inducible nitric oxide synthase (iNOS), double-stranded RNA-dependent protein kinase (PKR), and Src. The production of IFN-β was dependent on iNOS, PKR, and Src and partially dependent on TLR3/Trif. iNOS and Src up-regulation was partially dependent on TLR3/Trif but entirely dependent on PKR. The phosphorylation of TLR3 on tyrosine 759 was shown to increase in parallel to IFN-β production in an iNOS- and Src-dependent manner, and Src was found to directly interact with TLR3 in the endosomal compartment of poly(I:C)-treated cells. Furthermore, we identified a robust NO/cGMP/PKG-dependent feedforward pathway for the amplification of iNOS expression. These data identify iNOS/NO as an integral component of IFN-β production in response to dsRNA in hepatocytes in a pathway that involves the coordinated activities of TLR3/Trif and PKR.
Collapse
Affiliation(s)
- Liyong Zhang
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Wenpei Xiang
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, the Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoliang Wang
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Zhengzheng Yan
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Zhaowei Zhu
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Zhong Guo
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Rajib Sengupta
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Alex F Chen
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Patricia A Loughran
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, the Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, and
| | - Ben Lu
- the Xiangya Third Hospital and Central South University School of Medicine, Changsha, China
| | - Qingde Wang
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Timothy R Billiar
- From the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213,
| |
Collapse
|
35
|
Response of Mammalian Macrophages to Challenge with the Chlorovirus Acanthocystis turfacea Chlorella Virus 1. J Virol 2015; 89:12096-107. [PMID: 26401040 DOI: 10.1128/jvi.01254-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/16/2015] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED It was recently reported that 44% of the oropharyngeal samples from the healthy humans in a study cohort had DNA sequences similar to that of the chlorovirus ATCV-1 (Acanthocystis turfacea chlorella virus 1, family Phycodnaviridae) and that these study subjects had decreases in visual processing and visual motor speed compared with individuals in whom no virus was detected. Moreover, mice inoculated orally with ATCV-1 developed immune responses to ATCV-1 proteins and had decreases in certain cognitive domains. Because heightened interleukin-6 (IL-6), nitric oxide (NO), and ERK mitogen-activated protein (MAP) kinase activation from macrophages are linked to cognitive impairments, we evaluated cellular responses and viral PFU counts in murine RAW264.7 cells and primary macrophages after exposure to ATCV-1 in vitro for up to 72 h after a virus challenge. Approximately 8% of the ATCV-1 inoculum was associated with macrophages after 1 h, and the percentage increased 2- to 3-fold over 72 h. Immunoblot assays with rabbit anti-ATCV-1 antibody detected a 55-kDa protein consistent with the viral capsid protein from 1 to 72 h and increasing de novo synthesis of a previously unidentified 17-kDa protein beginning at 24 h. Emergence of the 17-kDa protein did not occur and persistence of the 55-kDa protein declined over time when cells were exposed to heat-inactivated ATCV-1. Moreover, starting at 24 h, RAW264.7 cells exhibited cytopathic effects, annexin V staining, and cleaved caspase 3. Activation of ERK MAP kinases occurred in these cells by 30 min postchallenge, which preceded the expression of IL-6 and NO. Therefore, ATCV-1 persistence in and induction of inflammatory factors by these macrophages may contribute to declines in the cognitive abilities of mice and humans. IMPORTANCE Virus infections that persist in and stimulate inflammatory factors in macrophages contribute to pathologies in humans. A previous study showed that DNA sequences homologous to the chlorovirus ATCV-1 were found in a significant fraction of oropharyngeal samples from a healthy human cohort. We show here that ATCV-1, whose only known host is a eukaryotic green alga (Chlorella heliozoae) that is an endosymbiont of the heliozoon Acanthocystis turfacea, can unexpectedly persist within murine macrophages and trigger inflammatory responses including factors that contribute to immunopathologies. The inflammatory factors that are produced in response to ATCV-1 include IL-6 and NO, whose induction is preceded by the activation of ERK MAP kinases. Other responses of ATCV-1-challenged macrophages include an apoptotic cytopathic effect, an innate antiviral response, and a metabolic shift toward aerobic glycolysis. Therefore, mammalian encounters with chloroviruses may contribute to chronic inflammatory responses from macrophages.
Collapse
|
36
|
Barjesteh N, Brisbin JT, Behboudi S, Nagy É, Sharif S. Induction of antiviral responses against avian influenza virus in embryonated chicken eggs with toll-like receptor ligands. Viral Immunol 2015; 28:192-200. [PMID: 25874816 DOI: 10.1089/vim.2014.0145] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Early responses against viruses, such as avian influenza virus (AIV), may be induced by Toll-like receptor (TLR) pathways. In the present study, an in ovo model was employed to study the antiviral activities of TLR ligands. It was hypothesized that administration of TLR ligands in ovo at the appropriate dose and time can reduce AIV titer in embryonated chicken eggs. Moreover, the study aimed to determine the mechanisms involved in the TLR-mediated antiviral responses in the chorioallantoic membrane (CAM). Embryonated eggs (10-14 day old) were treated with TLR2, 4, 7, and 21 ligands using different doses and times pre- and post-AIV infection. The results revealed that treatment of embryonated chicken eggs with TLR ligands reduced AIV replication. Further analysis showed that TLR ligands induced interferon (IFN)-γ and IFN stimulatory genes in the CAM, which may have played a role in the reduction of the AIV titer. The timing and dose of TLR ligands administration had significant impacts on the outcome of the treated eggs. In conclusion, the present study demonstrated that the in ovo route may be employed to determine the antiviral characteristics of TLR ligands against AIV.
Collapse
Affiliation(s)
- Neda Barjesteh
- 1 Department of Pathobiology, University of Guelph , Guelph, Canada
| | | | | | | | | |
Collapse
|
37
|
Collins SE, Mossman KL. Danger, diversity and priming in innate antiviral immunity. Cytokine Growth Factor Rev 2014; 25:525-31. [PMID: 25081316 DOI: 10.1016/j.cytogfr.2014.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 07/03/2014] [Indexed: 12/24/2022]
Abstract
The prototypic response to viral infection involves the recognition of pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs), leading to the activation of transcription factors such as IRF3 and NFkB and production of type 1 IFN. While this response can lead to the induction of hundreds of IFN-stimulated genes (ISGs) and recruitment and activation of immune cells, such a comprehensive response is likely inappropriate for routine low level virus exposure. Moreover, viruses have evolved a plethora of immune evasion strategies to subvert antiviral signalling. There is emerging evidence that cells have developed very sensitive methods of detecting not only specific viral PAMPS, but also more general danger or stress signals associated with viral entry and replication. Such stress-induced cellular responses likely serve to prime cells to respond to further PAMP stimulation or allow for a rapid and localized intracellular response independent of IFN production and its potential immune sequelae. This review discusses diversity in innate antiviral players and pathways, the role of "danger" sensing, and how alternative pathways, such as the IFN-independent pathway, may serve to prime cells for further pathogen attack.
Collapse
Affiliation(s)
- Susan E Collins
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Institute for Infectious Disease Research, McMaster University, Hamilton, Canada L8S 4K1
| | - Karen L Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Institute for Infectious Disease Research, McMaster University, Hamilton, Canada L8S 4K1.
| |
Collapse
|
38
|
Argininosuccinate synthetase 1 depletion produces a metabolic state conducive to herpes simplex virus 1 infection. Proc Natl Acad Sci U S A 2013; 110:E5006-15. [PMID: 24297925 DOI: 10.1073/pnas.1321305110] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) infection triggers specific metabolic changes in its host cell. To explore the interactions between cellular metabolism and HSV-1 infection, we performed an siRNA screen of cellular metabolic genes, measuring their effect on viral replication. The screen identified multiple enzymes predicted to influence HSV-1 replication, including argininosuccinate synthetase 1 (AS1), which consumes aspartate as part of de novo arginine synthesis. Knockdown of AS1 robustly enhanced viral genome replication and the production of infectious virus. Using high-resolution liquid chromatography-mass spectrometry, we found that the metabolic phenotype induced by knockdown of AS1 in human fibroblasts mimicked multiple aspects of the metabolic program observed during HSV-1 infection, including an increase in multiple nucleotides and their precursors. Together with the observation that AS1 protein and mRNA levels decrease during wild-type infection, this work suggests that reduced AS1 activity is partially responsible for the metabolic program induced by infection.
Collapse
|
39
|
Moore TC, Petro TM. IRF3 and ERK MAP-kinases control nitric oxide production from macrophages in response to poly-I:C. FEBS Lett 2013; 587:3014-20. [PMID: 23892079 DOI: 10.1016/j.febslet.2013.07.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/20/2013] [Accepted: 07/13/2013] [Indexed: 11/18/2022]
Abstract
Understanding nitric oxide (NO) in innate anti-viral immunity and immune-mediated pathology is hampered by incomplete details of its transcriptional and signaling factors. We found in macrophages that IRF3, ERK MAP-kinases, and PKR are essential to NO production in response to RNA-virus mimic, poly I:C, a TLR3 agonist. ERK's role in NO induction may be through phosphorylation of serine-171 of IRF3 and expression of NO-inducing cytokines, IL-6 and IFN-β. However, these cytokines induced less NO in IRF3 knockout or knockdown macrophages. These findings show that ERK and IRF3 coordinate induction of NO by macrophages in response to stimulation of TLR3.
Collapse
Affiliation(s)
- Tyler C Moore
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | |
Collapse
|
40
|
Poon DCH, Ho YS, Chiu K, Chang RCC. Cytokines: how important are they in mediating sickness? Neurosci Biobehav Rev 2012; 37:1-10. [PMID: 23153795 DOI: 10.1016/j.neubiorev.2012.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 09/28/2012] [Accepted: 11/04/2012] [Indexed: 01/20/2023]
Abstract
Sickness refers to a set of coordinated physiological and behavioral changes in response to systemic inflammation. It is characterized by fever, malaise, social withdrawal, fatigue, and anorexia. While these responses collectively represent a protective mechanism against infection and injury, increasing lines of evidence indicate that over-exaggerated or persistent sickness can damage the brain, and could possibly raise the risk to developing delirium. Therefore, a clear understanding in sickness will be beneficial. It has long been believed that sickness results from increased systemic cytokines occurring during systemic inflammation. However, in recent years more and more conflicting data have suggested that development of sickness following peripheral immune challenge could be independent of cytokines. Hence, it is confusing as to whether cytokines really do act as primary mediators of sickness, or if they are secondary to alternative inducing factor(s). In this review, we will (1) introduce the relationships between systemic inflammation, cytokines, sickness, and delirium, and (2) attempt to interpret the recent controversies.
Collapse
Affiliation(s)
- David Chun-Hei Poon
- Laboratory of Neurodegenerative Diseases, Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | |
Collapse
|