1
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Effects of oxidative stress on viral infections: an overview. NPJ VIRUSES 2025; 3:27. [PMID: 40295852 PMCID: PMC11993764 DOI: 10.1038/s44298-025-00110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
Viral infections can trigger increased reactive oxygen species (ROS) production and a reduced antioxidant response in the host, leading to redox stress, inflammation, apoptosis, and ultimately, cell and tissue damage, which contribute to disease development. A better understanding of how ROS contributes to viral pathogenesis is critical for the development of novel therapeutic interventions. In this review, we discuss the current knowledge on ROS production and its effects across various viral infections, including severe acute respiratory syndrome-coronavirus-2, influenza A virus, dengue virus, Zika virus, hepatitis B virus, hepatitis C virus, and human immunodeficiency virus infections, to improve future therapeutic and preventive strategies for these infections.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan.
| |
Collapse
|
2
|
Adams TJ, Schuliga M, Pearce N, Bartlett NW, Liang M. Targeting respiratory virus-induced reactive oxygen species in airways diseases. Eur Respir Rev 2025; 34:240169. [PMID: 40240057 PMCID: PMC12000908 DOI: 10.1183/16000617.0169-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/02/2025] [Indexed: 04/18/2025] Open
Abstract
The immune response to virus infection in the respiratory tract must be carefully balanced to achieve pathogen clearance without excessive immunopathology. For chronic respiratory diseases where there is ongoing inflammation, such as in asthma and COPD, airway immune balance is perturbed, and viral infection frequently worsens (exacerbates) these conditions. Reactive oxygen species (ROS) are critical to the induction and propagation of inflammation, and when appropriately regulated, ROS are vital cell signalling molecules and contribute to innate immunity. However, extended periods of high ROS concentration can cause excessive cellular damage that dysregulates antiviral immunity and promotes inflammation. Traditional antioxidant therapeutics have had limited success treating inflammatory diseases such as viral exacerbations of asthma or COPD, owing to nonspecific pharmacology and poorly understood pharmacokinetic properties. These drawbacks could be addressed with novel drug delivery technologies and pharmacological agents. This review summarises current research on ROS imbalances during virus infection, discusses the commercially available mitochondrial antioxidant drugs that have progressed to clinical trial and assesses novel drug delivery approaches for antioxidant delivery to the airways. Additionally, it provides a perspective on future research into pharmacological targeting of ROS for the treatment of respiratory virus infection and disease.
Collapse
Affiliation(s)
- Thomas J Adams
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Nyoaki Pearce
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Nathan W Bartlett
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| |
Collapse
|
3
|
Walter T, Matteo F, Marta DA, Carolina S, Leonardo S, Elena P, Maria Elena M, Fabio M, Enrica M, Raffaella N, Laura P, Anna Teresa P, Guido A, Alessandra P, Lucia N. NRF2 Antioxidant Response and Interferon-Stimulated Genes Are Differentially Expressed in SARS-CoV-2-Positive Young Subjects. Immun Inflamm Dis 2025; 13:e70109. [PMID: 39810451 PMCID: PMC11733084 DOI: 10.1002/iid3.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Several respiratory viruses, including Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2), suppress nuclear factor-E2-related factor-2 (NRF2) antioxidant response, generating oxidative stress conditions to its advantage. NRF2 has also been reported to regulate the innate immune response through the inhibition of the interferon (IFN) pathway. However, its modulation in younger individuals and its correlation with the IFN response remain to be elucidated. METHODS The NRF2 and redox-related genes expression was examined in nasopharyngeal swabs from children attending the pediatric hospital for SARS-CoV-2 molecular testing. Expression levels were analyzed by stratifying the population according to the SARS-CoV-2 positivity, age, or the presence of symptoms. The results were correlated with Types I and III IFN genes and IFN-stimulated genes (ISGs). RESULTS We found that NRF2 expression was markedly diminished in positive patients compared to negative. Moreover, it correlated with higher expression of IFNα2 and IFNλ3, as well as ISG15 and ISG56. Interestingly, symptomatic patients with anosmia/ageusia showed pronounced expression of apurinic/apyrimidinic endonuclease1/redox factor 1 (APE1), together with Type I IFNs, ISG56, and the inflammasome component NLRP3. CONCLUSION The results indicate an interdependence between NRF2 antioxidant pathway and IFN-mediated response during SARS-CoV-2 infection in young subjects.
Collapse
Affiliation(s)
- Toscanelli Walter
- Laboratory Affiliated to Istituto Pasteur Italia‐Fondazione Cenci Bolognetti, Department of Public Health and Infectious DiseasesSapienza UniversityRomeItaly
| | - Fracella Matteo
- Laboratory of Virology, Department of Molecular MedicineSapienza UniversityRomeItaly
| | - De Angelis Marta
- Laboratory Affiliated to Istituto Pasteur Italia‐Fondazione Cenci Bolognetti, Department of Public Health and Infectious DiseasesSapienza UniversityRomeItaly
- Laboratory of Virology, Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Scagnolari Carolina
- Laboratory of Virology, Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Sorrentino Leonardo
- Laboratory of Virology, Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Piselli Elena
- Laboratory Affiliated to Istituto Pasteur Italia‐Fondazione Cenci Bolognetti, Department of Public Health and Infectious DiseasesSapienza UniversityRomeItaly
| | - Marcocci Maria Elena
- Laboratory Affiliated to Istituto Pasteur Italia‐Fondazione Cenci Bolognetti, Department of Public Health and Infectious DiseasesSapienza UniversityRomeItaly
| | - Midulla Fabio
- Department of Maternal Infantile and Urological SciencesSapienza UniversityRomeItaly
| | - Mancino Enrica
- Department of Maternal Infantile and Urological SciencesSapienza UniversityRomeItaly
| | - Nenna Raffaella
- Department of Maternal Infantile and Urological SciencesSapienza UniversityRomeItaly
| | - Petrarca Laura
- Department of Maternal Infantile and Urological SciencesSapienza UniversityRomeItaly
| | - Palamara Anna Teresa
- Laboratory Affiliated to Istituto Pasteur Italia‐Fondazione Cenci Bolognetti, Department of Public Health and Infectious DiseasesSapienza UniversityRomeItaly
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Antonelli Guido
- Laboratory of Virology, Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Pierangeli Alessandra
- Laboratory of Virology, Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Nencioni Lucia
- Laboratory Affiliated to Istituto Pasteur Italia‐Fondazione Cenci Bolognetti, Department of Public Health and Infectious DiseasesSapienza UniversityRomeItaly
| |
Collapse
|
4
|
Nakhaie M, Rukerd MRZ, Shahpar A, Pardeshenas M, Khoshnazar SM, Khazaeli M, Bashash D, Nezhad NZ, Charostad J. A Closer Look at the Avian Influenza Virus H7N9: A Calm before the Storm? J Med Virol 2024; 96:e70090. [PMID: 39601174 DOI: 10.1002/jmv.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The avian influenza A (H7N9) virus, which circulates in wild birds and poultry, has been a major concern for public health since it was first discovered in China in 2013 due to its demonstrated ability to infect humans, causing severe respiratory illness with high mortality rates. According to the World Health Organization (WHO), a total of 1568 human infections with 616 fatal cases caused by novel H7N9 viruses have been reported in China from early 2013 to January 2024. This manuscript provides a comprehensive review of the virology, evolutionary patterns, and pandemic potential of H7N9. The H7N9 virus exhibits a complex reassortment history, receiving genes from H9N2 and other avian influenza viruses. The presence of certain molecular markers, such as mutations in the hemagglutinin and polymerase basic protein 2, enhances the virus's adaptability to human hosts. The virus activates innate immune responses through pattern recognition receptors, leading to cytokine production and inflammation. Clinical manifestations range from mild to severe, with complications including pneumonia, acute respiratory distress syndrome, and multiorgan failure. Diagnosis relies on molecular assays such as reverse transcription-polymerase chain reaction. The increasing frequency of human infections, along with the virus's ability to bind to human receptors and cause severe disease, highlights its pandemic potential. Continued surveillance, vaccine development, and public health measures are crucial to limit the risk posed by H7N9. Understanding the virus's ecology, transmission dynamics, and pathogenesis is essential for developing effective prevention and control strategies.
Collapse
Affiliation(s)
- Mohsen Nakhaie
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Rezaei Zadeh Rukerd
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amirhossein Shahpar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Pardeshenas
- Department of Microbiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mana Khazaeli
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Zeinali Nezhad
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Javad Charostad
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
5
|
Dou B, Wu X, He Y, Xu G, Zhang H, Huang Q, Chen X, Duan N, Zhou L, Zhang W, An H, Zheng Y. Fei-Yan-Qing-Hua decoction attenuates influenza virus infection by enhancing host antiviral response through microbiota-derived acetate. Front Pharmacol 2024; 15:1446749. [PMID: 39449967 PMCID: PMC11499185 DOI: 10.3389/fphar.2024.1446749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Background Fei-Yan-Qing-Hua decoction (FYQHD) is derived from the well-known Ma Xing Shi Gan decoction, which was documented in Zhang Zhong Jing's "Treatise on Exogenous Febrile Disease" during the Han Dynasty. Although FYQHD has been used in the treatment of pneumonia and has demonstrated clinical efficacy for decades, the underlying mechanism by which FYQHD protects against influenza virus infection through modulation of gut flora remains unclear. Here, we examined the regulatory impacts of FYQHD on an influenza virus-infected mouse model and explored the mechanisms involved. Methods An infectious mouse model was created by intranasal instillation of influenza A virus (IAV). The effectiveness of FYQHD was assessed through various measures, including weight loss, lung wet/dry ratio, oxidative stress levels, viral load in lung tissues, and intestinal injuries. Changes in gut microbiota and SCFA production were also examined. Results The results showed that FYQHD significantly reduced viral load, increased the production of type I interferon (IFN-I), and restored the integrity of the intestinal barrier following IAV challenge. Additionally, FYQHD significantly corrected the dysbiosis of gut microbiota induced by influenza virus infection, enhancing the abundance of SCFA-producing bacteria and acetate production. However, the depletion of gut microbiota significantly attenuated the protective effects of FYQHD against influenza virus infection. In vitro, the antiviral effect of acetate was demonstrated through the upregulation of concentrations of IFN-β. Conclusion FYQHD attenuates influenza virus-induced lung and intestinal injuries by boosting the host antiviral response through increasing the abundance of Lachnospiraceae_NK4A136 and Roseburia, along with elevated acetate levels. The study advances our understanding of the therapeutic mechanisms of FYQHD and provides a theoretical basis for the application of FYQHD in the treatment of influenza.
Collapse
Affiliation(s)
- Biao Dou
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Medical Laboratory, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Xiao Wu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yurong He
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huan Zhang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qilin Huang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Naifan Duan
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linqiong Zhou
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huazhang An
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Zhou C, Wang Q, Cao H, Jiang J, Gao L. Nanozybiotics: Advancing Antimicrobial Strategies Through Biomimetic Mechanisms. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403362. [PMID: 38874860 DOI: 10.1002/adma.202403362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Infectious diseases caused by bacterial, viral, and fungal pathogens present significant global health challenges. The rapid emergence of antimicrobial resistance exacerbates this issue, leading to a scenario where effective antibiotics are increasingly scarce. Traditional antibiotic development strategies are proving inadequate against the swift evolution of microbial resistance. Therefore, there is an urgent need to develop novel antimicrobial strategies with mechanisms distinct from those of existing antibiotics. Nanozybiotics, which are nanozyme-based antimicrobials, mimic the catalytic action of lysosomal enzymes in innate immune cells to kill infectious pathogens. This review reinforces the concept of nanozymes and provides a comprehensive summary of recent research advancements on potential antimicrobial candidates. Initially, nanozybiotics are categorized based on their activities, mimicking either oxidoreductase-like or hydrolase-like functions, thereby highlighting their superior mechanisms in combating antimicrobial resistance. The review then discusses the progress of nanozybiotics in treating bacterial, viral, and fungal infections, confirming their potential as novel antimicrobial candidates. The translational potential of nanozybiotic-based products, including hydrogels, nanorobots, sprays, bandages, masks, and protective clothing, is also considered. Finally, the current challenges and future prospects of nanozybiotic-related products are explored, emphasizing the design and antimicrobial capabilities of nanozybiotics for future applications.
Collapse
Affiliation(s)
- Caiyu Zhou
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Haidian, Beijing, 100049, China
| | - Qian Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Haidian, Beijing, 100049, China
| | - Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Haidian, Beijing, 100049, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
| |
Collapse
|
7
|
Zhang Y, Zhu Y, Jiang G, Chen K, Zhang G, Chen K, Ye T, Zhou Y, Li G. ROS Induced by Aphrocallistes vastus Lectin Enhance Oncolytic Vaccinia Virus Replication and Induce Apoptosis in Hepatocellular Carcinoma Cells. Mar Drugs 2024; 22:307. [PMID: 39057416 PMCID: PMC11278381 DOI: 10.3390/md22070307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Oncolytic virotherapy is expected to provide a new treatment strategy for cancer. Aphrocallistes vastus lectin (AVL) is a Ca2+-dependent lectin receptor containing the conserved domain of C-type lectin and the hydrophobic N-terminal region, which can bind to the bird's nest glycoprotein and D-galactose. Our previous studies suggested that the oncolytic vaccinia virus (oncoVV) armed with the AVL gene exerted remarkable replication and antitumor effects in vitro and in vivo. In this study, we found that oncoVV-AVL may reprogram the metabolism of hepatocellular carcinoma cells to promote ROS, and elevated ROS subsequently promoted viral replication and induced apoptosis. This study will provide a new theoretical basis for the application of oncoVV-AVL in liver cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yanrong Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Gongchu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
8
|
Zhang J, Xia Y, Li X, He R, Xie X. Case report: A case of Acute Macular Neuroretinopathy secondary to Influenza A virus during Long COVID. Front Immunol 2024; 14:1302504. [PMID: 38288123 PMCID: PMC10822910 DOI: 10.3389/fimmu.2023.1302504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/18/2023] [Indexed: 01/31/2024] Open
Abstract
Ocular abnormalities have been reported in association with viral infections, including Long COVID, a debilitating illness caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This report presents a case of a female patient diagnosed with Acute Macular Neuroretinopathy (AMN) following an Influenza A virus infection during Long COVID who experienced severe inflammation symptoms and ocular complications. We hypothesize that the rare occurrence of AMN in this patient could be associated with the immune storm secondary to the viral infection during Long COVID.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yihao Xia
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaodong Li
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Runxi He
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuejun Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Guerrero M, Hernández J, Gomez L, Guerrero C. Oxidative stress enhances rotavirus oncolysis in breast cancer and leukemia, except in melanoma with abundant matrix. Virus Res 2024; 339:199285. [PMID: 38013142 PMCID: PMC10711233 DOI: 10.1016/j.virusres.2023.199285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVES This study aimed to explore the impact of oxidative stress and extracellular matrix integrity on rotavirus infection in various cancer cells, including breast cancer, acute lymphoblastic leukemia, and melanoma. METHODS We induced oxidative stress using ROS-inducing drugs (cisplatin, metronidazole, melatonin, valproic acid, doxorubicin, losartan, nitrofurantoin, and DHA) and investigated the effects on viral infection in MCF-7, Reh, A375, B16-F1, and SK-MEL-28 cells and the generation of virions from infected cells by harvesting the supernatants every two hours, reinfecting other cells, and analyzing cell viability and DNA fragmentation. FINDINGS In MCF-7 and Reh cells, rotavirus Wt1-5 infection led to increased ROS generation, virion production, membrane permeability, mitochondrial dysfunction, DNA damage, and cell death. These effects were amplified by ROS-inducing drugs. Conversely, melanoma cells (SK-MEL-28 and A375) with a robust extracellular matrix network showed limited sensitivity to the drugs. Notably, losartan, which modulates the extracellular matrix, enhanced viral infection in melanoma cells (99 %). CONCLUSIONS Oxidative stress promotes oncolytic rotavirus infection in breast cancer and acute lymphoblastic leukemia cells, suggesting potential utility in combination with radiotherapy or chemotherapy due to their shared induction of intracellular oxidative stress.
Collapse
Affiliation(s)
- Marvi Guerrero
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad Nacional de Colombia, DC, Bogotá, Colombi
| | - Juan Hernández
- Grupo de Fisiología Molecular del Instituto Nacional de Salud. A. A. 80080. Av. Calle 26 No. 51-20 DC, Bogotá, Colombia
| | - Luis Gomez
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad Nacional de Colombia, DC, Bogotá, Colombi; Grupo de Fisiología Molecular del Instituto Nacional de Salud. A. A. 80080. Av. Calle 26 No. 51-20 DC, Bogotá, Colombia
| | - Carlos Guerrero
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad Nacional de Colombia, DC, Bogotá, Colombi.
| |
Collapse
|
10
|
Sutton SS, Magagnoli J, Cummings T, Hardin JW. Serum Bilirubin Levels and Risk of Venous Thromboembolism among Influenza Patients: A Cohort Study. Clin Appl Thromb Hemost 2024; 30:10760296241275138. [PMID: 39215507 PMCID: PMC11367695 DOI: 10.1177/10760296241275138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the associations between total serum bilirubin levels and the incidence of venous thromboembolism (VTE) among patients with influenza infection. METHODS A retrospective cohort study was conducted among outpatients with laboratory-confirmed influenza using data from the Veterans Affairs Informatics and Computing Infrastructure (VINCI). Propensity score weighting was applied to balance study groups across baseline covariates. Cox proportional hazards models assessed VTE risk by total bilirubin levels, adjusting for important covariates including age, sex, race, comorbidity index, BMI, and smoking status. RESULTS A total of 487 patients with total bilirubin levels <0.3 mg/dL, 8608 patients with levels between 0.3-1 mg/dL, and 1148 patients with levels >1 mg/dL were included. Patients with bilirubin <0.3 mg/dL exhibited a 6-fold higher risk of VTE compared to those with levels 0.3-1 mg/dL within 30 days of infection (HR = 6.2, 95% CI = 1.46-26.42). Elevated risks were noted through 90 days post infection (HR = 4.71, 95% CI = (1.42-15.67)). CONCLUSIONS Serum bilirubin levels, particularly below 0.3 mg/dL, were significantly associated with an increased risk of VTE among individuals with influenza. These findings suggest that lower bilirubin levels may contribute to heightened inflammatory responses and subsequent thromboembolic events in patients with influenza. The underlying mechanisms and potential therapeutic implications for VTE prevention among patients with acute respiratory infection warrants further consideration.
Collapse
Affiliation(s)
- S. Scott Sutton
- Dorn Research Institute, Columbia VA Health Care System, Columbia, South Carolina; Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Joseph Magagnoli
- Dorn Research Institute, Columbia VA Health Care System, Columbia, South Carolina; Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Tammy Cummings
- Dorn Research Institute, Columbia VA Health Care System, Columbia, South Carolina; Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - James W. Hardin
- Dorn Research Institute, Columbia VA Health Care System, Columbia, South Carolina; Department of Epidemiology & Biostatistics, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
11
|
De Angelis M, Anichini G, Palamara AT, Nencioni L, Gori Savellini G. Dysregulation of intracellular redox homeostasis by the SARS-CoV-2 ORF6 protein. Virol J 2023; 20:239. [PMID: 37853388 PMCID: PMC10585933 DOI: 10.1186/s12985-023-02208-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
SARS-CoV-2 has evolved several strategies to overcome host cell defenses by inducing cell injury to favour its replication. Many viruses have been reported to modulate the intracellular redox balance, affecting the Nuclear factor erythroid 2-Related Factor 2 (NRF2) signaling pathway. Although antioxidant modulation by SARS-CoV-2 infection has already been described, the viral factors involved in modulating the NRF2 pathway are still elusive. Given the antagonistic activity of ORF6 on several cellular pathways, we investigated the role of the viral protein towards NRF2-mediated antioxidant response. The ectopic expression of the wt-ORF6 protein negatively impacts redox cell homeostasis, leading to an increase in ROS production, along with a decrease in NRF2 protein and its downstream controlled genes. Moreover, when investigating the Δ61 mutant, previously described as an inactive nucleopore proteins binding mutant, we prove that the oxidative stress induced by ORF6 is substantially related to its C-terminal domain, speculating that ORF6 mechanism of action is associated with the inhibition of nuclear mRNA export processes. In addition, activation by phosphorylation of the serine residue at position 40 of NRF2 is increased in the cytoplasm of wt-ORF6-expressing cells, supporting the presence of an altered redox state, although NRF2 nuclear translocation is hindered by the viral protein to fully antagonize the cell response. Furthermore, wt-ORF6 leads to phosphorylation of a stress-activated serine/threonine protein kinase, p38 MAPK, suggesting a role of the viral protein in regulating p38 activation. These findings strengthen the important role of oxidative stress in the pathogenesis of SARS-CoV-2 and identify ORF6 as an important viral accessory protein hypothetically involved in modulating the antioxidant response during viral infection.
Collapse
Affiliation(s)
- Marta De Angelis
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy.
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy.
| | - Gabriele Anichini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Department of Infectious Diseases, Istituto Superiore Di Sanità, Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | | |
Collapse
|
12
|
Percaccio E, De Angelis M, Acquaviva A, Nicotra G, Ferrante C, Mazzanti G, Di Giacomo S, Nencioni L, Di Sotto A. ECHOPvir: A Mixture of Echinacea and Hop Extracts Endowed with Cytoprotective, Immunomodulatory and Antiviral Properties. Nutrients 2023; 15:4380. [PMID: 37892456 PMCID: PMC10609862 DOI: 10.3390/nu15204380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Respiratory viral infections continue to pose significant challenges, particularly for more susceptible and immunocompromised individuals. Nutraceutical strategies have been proposed as promising strategies to mitigate their impact and improve public health. In the present study, we developed a mixture of two hydroalcoholic extracts from the aerial parts of Echinacea purpurea (L.) Moench (ECP) and the cones of Humulus lupulus L. (HOP) that can be harnessed in the prevention and treatment of viral respiratory diseases. The ECP/HOP mixture (named ECHOPvir) was characterized for the antioxidant and cytoprotective properties in airway cells. Moreover, the immunomodulating properties of the mixture in murine macrophages against antioxidant and inflammatory stimuli and its antiviral efficacy against the PR8/H1N1 influenza virus were assayed. The modulation of the Nrf2 was also investigated as a mechanistic hypothesis. The ECP/HOP mixture showed a promising multitarget bioactivity profile, with combined cytoprotective, antioxidant, immunomodulating and antiviral activities, likely due to the peculiar phytocomplexes of both ECP and HOP, and often potentiated the effect of the single extracts. The Nrf2 activation seemed to trigger these cytoprotective properties and suggest a possible usefulness in counteracting the damage caused by different stressors, including viral infection. Further studies may strengthen the interest in this product and underpin its future nutraceutical applications.
Collapse
Affiliation(s)
- Ester Percaccio
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (E.P.); (G.M.)
| | - Marta De Angelis
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.D.A.); (L.N.)
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandra Acquaviva
- Department of Pharmacy, Botanic Garden “Giardino dei Semplici”, Università degli Studi “Gabriele d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy; (A.A.); (C.F.)
| | | | - Claudio Ferrante
- Department of Pharmacy, Botanic Garden “Giardino dei Semplici”, Università degli Studi “Gabriele d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy; (A.A.); (C.F.)
| | - Gabriela Mazzanti
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (E.P.); (G.M.)
| | - Silvia Di Giacomo
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (E.P.); (G.M.)
- Unit of Human Nutrition and Health, Department of Food Safety, Nutrition and Veterinary Public Health, National Institute of Health, 00161 Rome, Italy;
| | - Lucia Nencioni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.D.A.); (L.N.)
| | - Antonella Di Sotto
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (E.P.); (G.M.)
| |
Collapse
|
13
|
Hernandez-Morfa M, Olivero NB, Zappia VE, Piñas GE, Reinoso-Vizcaino NM, Cian MB, Nuñez-Fernandez M, Cortes PR, Echenique J. The oxidative stress response of Streptococcus pneumoniae: its contribution to both extracellular and intracellular survival. Front Microbiol 2023; 14:1269843. [PMID: 37789846 PMCID: PMC10543277 DOI: 10.3389/fmicb.2023.1269843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Streptococcus pneumoniae is a gram-positive, aerotolerant bacterium that naturally colonizes the human nasopharynx, but also causes invasive infections and is a major cause of morbidity and mortality worldwide. This pathogen produces high levels of H2O2 to eliminate other microorganisms that belong to the microbiota of the respiratory tract. However, it also induces an oxidative stress response to survive under this stressful condition. Furthermore, this self-defense mechanism is advantageous in tolerating oxidative stress imposed by the host's immune response. This review provides a comprehensive overview of the strategies employed by the pneumococcus to survive oxidative stress. These strategies encompass the utilization of H2O2 scavengers and thioredoxins, the adaptive response to antimicrobial host oxidants, the regulation of manganese and iron homeostasis, and the intricate regulatory networks that control the stress response. Here, we have also summarized less explored aspects such as the involvement of reparation systems and polyamine metabolism. A particular emphasis is put on the role of the oxidative stress response during the transient intracellular life of Streptococcus pneumoniae, including coinfection with influenza A and the induction of antibiotic persistence in host cells.
Collapse
Affiliation(s)
- Mirelys Hernandez-Morfa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nadia B. Olivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria E. Zappia
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - German E. Piñas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolas M. Reinoso-Vizcaino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Melina B. Cian
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Nuñez-Fernandez
- Centro de Química Aplicada, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paulo R. Cortes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jose Echenique
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
14
|
Guo M, Chen D, Zhao M, Xu T, Zhang Y, Xiao M, Li Y, Zhu B. 5-Nitrobenzo[c][1, 2, 5]selenadiazole as therapeutic agents in the regulation of oxidative stress and inflammation induced by influenza A(H1N1)pdm09 in vitro and in vivo. J Med Virol 2023; 95:e28920. [PMID: 37386905 DOI: 10.1002/jmv.28920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 05/23/2023] [Accepted: 06/11/2023] [Indexed: 07/01/2023]
Abstract
Currently, various problems are being faced in the treatment of influenza, so the development of new safe and effective drugs is crucial. Selenadiazole, an important component of selenium heterocyclic compounds, has received wide attention for its biological activity. This study aimed to verify the antiviral activity of 5-nitrobenzo[c][1,2,5]selenadiazole (SeD-3) in vivo and in vitro. The cell counting kit-8 assay and observation of cytopathic effect verified that SeD-3 could improve the survival of influenza A(H1N1)pdm09-infected Madin-Darby canine kidney cells. Polymerase chain reaction quantification and neuraminidase assay showed that SeD-3 could inhibit the proliferation of H1N1 virus. The time of addition assay demonstrated that SeD-3 may have a direct effect on virus particles and block some stages of H1N1 life cycle after virus adsorption. Cell cycle, JC-1, Annexin V, and terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling-4',6-diamidino-2-phenylindole (TUNEL-DAPI) assays showed that SeD-3 inhibited H1N1 infection-induced apoptosis. Cytokine detection demonstrated SeD-3 inhibited the production of proinflammatory factors after infection, including tumor necrosis factor-α (TNF-α), TNF-β, interferon-γ, interleukin 12 (IL-12), and IL-17F. In vivo experiments suggested that the pathological damage in the lungs was significantly alleviated after treatment with SeD-3 by hematoxylin and eosin staining. The TUNEL assay of lung tissues indicated that SeD-3 inhibited DNA damage during H1N1 infection. Immunohistochemical assays were performed to further explore the mechanism that SeD-3 inhibited H1N1-induced apoptosis via reactive oxygen species-mediated MAPK, AKT, and P53 signaling pathways. In conclusion, SeD-3 may become a new potential anti-H1N1 influenza virus drug due to its antiviral and anti-inflammatory activity.
Collapse
Affiliation(s)
- Min Guo
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Danyang Chen
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mingqi Zhao
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Tiantian Xu
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yingying Zhang
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Misi Xiao
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yinghua Li
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Bing Zhu
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Marsili G, Acchioni C, Remoli AL, Amatore D, Sgarbanti R, De Angelis M, Orsatti R, Acchioni M, Astolfi A, Iraci N, Puzelli S, Facchini M, Perrotti E, Cecchetti V, Sabatini S, Superti F, Agamennone M, Barreca ML, Hiscott J, Nencioni L, Sgarbanti M. Identification of Anti-Influenza A Compounds Inhibiting the Viral Non-Structural Protein 1 (NS1) Using a Type I Interferon-Driven Screening Strategy. Int J Mol Sci 2023; 24:10495. [PMID: 37445672 DOI: 10.3390/ijms241310495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
There is an urgent need to identify efficient antiviral compounds to combat existing and emerging RNA virus infections, particularly those related to seasonal and pandemic influenza outbreaks. While inhibitors of the influenza viral integral membrane proton channel protein (M2), neuraminidase (NA), and cap-dependent endonuclease are available, circulating influenza viruses acquire resistance over time. Thus, the need for the development of additional anti-influenza drugs with novel mechanisms of action exists. In the present study, a cell-based screening assay and a small molecule library were used to screen for activities that antagonized influenza A non-structural protein 1 (NS1), a highly conserved, multifunctional accessory protein that inhibits the type I interferon response against influenza. Two potential anti-influenza agents, compounds 157 and 164, were identified with anti-NS1 activity, resulting in the reduction of A/PR/8/34(H1N1) influenza A virus replication and the restoration of IFN-β expression in human lung epithelial A549 cells. A 3D pharmacophore modeling study of the active compounds provided a glimpse of the structural motifs that may contribute to anti-influenza virus activity. This screening approach is amenable to a broader analysis of small molecule compounds to inhibit other viral targets.
Collapse
Affiliation(s)
- Giulia Marsili
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Chiara Acchioni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Anna Lisa Remoli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Donatella Amatore
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Rossella Sgarbanti
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Roberto Orsatti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marta Acchioni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Andrea Astolfi
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Nunzio Iraci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Simona Puzelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marzia Facchini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Edvige Perrotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Fabiana Superti
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Mariangela Agamennone
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Maria Letizia Barreca
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - John Hiscott
- Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Marco Sgarbanti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
16
|
Gopal J, Muthu M, Sivanesan I. A Comprehensive Survey on the Expediated Anti-COVID-19 Options Enabled by Metal Complexes-Tasks and Trials. Molecules 2023; 28:molecules28083354. [PMID: 37110587 PMCID: PMC10143858 DOI: 10.3390/molecules28083354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Contemporary pharmacology dating back to the late 19th/early 20th centuries has benefitted largely from the incorporation of metal complexes. Various biological attributes have been successfully realized using metal/metal complex-based drugs. Among anticancer, antimicrobial, and antiviral applications, anticancer applications have extracted the maximum benefit from the metal complex, Cisplatin. The following review has compiled the various antiviral benefits harnessed through inputs from metal complexes. As a result of exploiting the pharmacological aspects of metal complexes, the anti-COVID-19 deliverables have been summarized. The challenges ahead, the gaps in this research area, the need to improvise incorporating nanoaspects in metal complexes, and the need to test metal complex-based drugs in clinical trials have been discussed and deliberated. The pandemic shook the entire world and claimed quite a percentage of the global population. Metal complex-based drugs are already established for their antiviral property with respect to enveloped viruses and extrapolating them for COVID-19 can be an effective way to manipulate drug resistance and mutant issues that the current anti-COVID-19 drugs are facing.
Collapse
Affiliation(s)
- Judy Gopal
- Department of Research and Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Manikandan Muthu
- Department of Research and Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Iyyakkannu Sivanesan
- Department of Bioresources and Food Science, Institute of Natural Science and Agriculture, Konkuk University, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
17
|
How the Competition for Cysteine May Promote Infection of SARS-CoV-2 by Triggering Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12020483. [PMID: 36830041 PMCID: PMC9952211 DOI: 10.3390/antiox12020483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
SARS-CoV-2 induces a broad range of clinical manifestations. Besides the main receptor, ACE2, other putative receptors and co-receptors have been described and could become genuinely relevant to explain the different tropism manifested by new variants. In this study, we propose a biochemical model envisaging the competition for cysteine as a key mechanism promoting the infection and the selection of host receptors. The SARS-CoV-2 infection produces ROS and triggers a massive biosynthesis of proteins rich in cysteine; if this amino acid becomes limiting, glutathione levels are depleted and cannot control oxidative stress. Hence, infection succeeds. A receptor should be recognized as a marker of suitable intracellular conditions, namely the full availability of amino acids except for low cysteine. First, we carried out a comparative investigation of SARS-CoV-2 proteins and human ACE2. Then, using hierarchical cluster protein analysis, we searched for similarities between all human proteins and spike produced by the latest variant, Omicron BA.1. We found 32 human proteins very close to spike in terms of amino acid content. Most of these potential SARS-CoV-2 receptors have less cysteine than spike. We suggest that these proteins could signal an intracellular shortage of cysteine, predicting a burst of oxidative stress when used as viral entry mediators.
Collapse
|
18
|
Checconi P, Coni C, Limongi D, Baldelli S, Ciccarone F, De Angelis M, Mengozzi M, Ghezzi P, Ciriolo MR, Nencioni L, Palamara AT. Influenza virus replication is affected by glutaredoxin1-mediated protein deglutathionylation. FASEB J 2023; 37:e22729. [PMID: 36583688 DOI: 10.1096/fj.202201239rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022]
Abstract
Several redox modifications have been described during viral infection, including influenza virus infection, but little is known about glutathionylation and this respiratory virus. Glutathionylation is a reversible, post-translational modification, in which protein cysteine forms transient disulfides with glutathione (GSH), catalyzed by cellular oxidoreductases and in particular by glutaredoxin (Grx). We show here that (i) influenza virus infection induces protein glutathionylation, including that of viral proteins such as hemagglutinin (HA); (ii) Grx1-mediated deglutathionylation is important for the viral life cycle, as its inhibition, either with an inhibitor of its enzymatic activity or by siRNA, decreases viral replication. Overall these data contribute to the characterization of the complex picture of redox regulation of the influenza virus replication cycle and could help to identify new targets to control respiratory viral infection.
Collapse
Affiliation(s)
- Paola Checconi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy.,Laboratory of Microbiology of Chronic-Neurodegenerative Diseases, IRCCS San Raffaele Roma, Rome, Italy
| | - Cristiana Coni
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy.,Laboratory of Microbiology of Chronic-Neurodegenerative Diseases, IRCCS San Raffaele Roma, Rome, Italy
| | - Dolores Limongi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy.,Laboratory of Microbiology of Chronic-Neurodegenerative Diseases, IRCCS San Raffaele Roma, Rome, Italy
| | - Sara Baldelli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy.,Laboratory of Biochemistry of Aging, IRCCS San Raffaele Roma, Rome, Italy
| | - Fabio Ciccarone
- Laboratory of Biochemistry of Aging, IRCCS San Raffaele Roma, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Marta De Angelis
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Manuela Mengozzi
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Brighton, UK
| | - Pietro Ghezzi
- Department of Biomolecular Sciences, University of Urbino, Urbino, Italy
| | - Maria Rosa Ciriolo
- Laboratory of Biochemistry of Aging, IRCCS San Raffaele Roma, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Lucia Nencioni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Anna Teresa Palamara
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.,Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
19
|
Shrivastava AK, Sahu PK, Cecchi T, Shrestha L, Shah SK, Gupta A, Palikhey A, Joshi B, Gupta PP, Upadhyaya J, Paudel M, Koirala N. An emerging natural antioxidant therapy for COVID‐19 infection patients: Current and future directions. FOOD FRONTIERS 2023. [DOI: 10.1002/fft2.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Amit Kumar Shrivastava
- Department of Pharmacology Universal College of Medical Sciences Bhairahawa Rupandehi Nepal
| | - Prafulla Kumar Sahu
- School of Pharmacy Centurion University of Technology and Management Bhubaneswar Odisha India
| | | | - Laxmi Shrestha
- Department of Pharmacology Universal College of Medical Sciences Bhairahawa Rupandehi Nepal
| | - Sanjay Kumar Shah
- Department of Reproductive MedicineJoint Inter‐national Research Laboratory of Reproduction and DevelopmentChongquing Medical University ChongqingPeople's Republic of China
| | - Anamika Gupta
- Sharjah Institute for Medical Sciences University of Sharjah Sharjah United Arab Emirates
| | - Anjan Palikhey
- Department of Pharmacology Universal College of Medical Sciences Bhairahawa Rupandehi Nepal
| | - Bishal Joshi
- Department of Physiology, Universal College of Medical Sciences Bhairahawa Rupandehi Nepal
| | - Pramodkumar P. Gupta
- School of Biotechnology and Bioinformatics D. Y. Patil Deemed to be University, CBD Belapur Navi Mumbai India
| | - Jitendra Upadhyaya
- Institute of Agriculture and Animal Science Tribhuvan University Chitwan Nepal
| | - Mahendra Paudel
- Department of Agri‐Botany and Ecology Institute of Agriculture and Animal Science Tribhuvan University Mahendranagar Nepal
| | - Niranjan Koirala
- Natural Products Research FacilityGandaki Province Academy of Science and Technology Pokhara, Gandaki Province Nepal
| |
Collapse
|
20
|
NOX2 oxidase inhibitor GSK2795039 possess antiviral activity against H1N1 influenza A virus in vitro and vivo. Microb Pathog 2023; 174:105942. [PMID: 36502994 DOI: 10.1016/j.micpath.2022.105942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
The continuous zoonotic circulation and reassortment potential of influenza A viruses (IAV) in nature represents an enormous public health threat to humans. Beside vaccination antivirals are needed to efficiently control spreading of the disease. The previous research has shown that NOX2 involved in IAV replication, but the detailed mechanism has not been reported. In the present study we investigated the roles of NOX2 in host inflammatory response and IAV replication using a novel inhibitor GSK2795039. The drug significantly reduced H1N1 virus induced NOX2 activity and ROS release in human lung epithelial cells. The results of time course experiments suggested that GSK2795039 inhibited an early post-entry step of viral infection. Concomitantly, there was a decreased expression of pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interferon (IFN)-β and interleukin (IL)-6) in NOX2 suppressed cells. In vivo, compared with control groups, suppression of NOX2 improved the survival rate of mice infected with H1N1 virus (42.9% in GSK2795039 treated mice versus >0% of control mice) and viral burden also decreased in the GSK2795039 treated group. Thus, our data demonstrated a critical role for NOX2 in the establishment of H1N1 infection and subsequent inflammatory reactions, which suggest that GSK2795039 may be a potential therapeutic drug for IAV infection.
Collapse
|
21
|
Ristovski JT, Matin MM, Kong R, Kusturica MP, Zhang H. In vitro testing and computational analysis of specific phytochemicals with antiviral activities considering their possible applications against COVID-19. SOUTH AFRICAN JOURNAL OF BOTANY : OFFICIAL JOURNAL OF THE SOUTH AFRICAN ASSOCIATION OF BOTANISTS = SUID-AFRIKAANSE TYDSKRIF VIR PLANTKUNDE : AMPTELIKE TYDSKRIF VAN DIE SUID-AFRIKAANSE GENOOTSKAP VAN PLANTKUNDIGES 2022; 151:248-258. [PMID: 35165493 PMCID: PMC8828436 DOI: 10.1016/j.sajb.2022.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/17/2021] [Accepted: 02/05/2022] [Indexed: 06/13/2023]
Abstract
The purpose of this study was to investigate the reservoir of natural products against the SARS-CoV-2 virus and to identify suitable candidates in order to recommend appropriate phytotherapy. Adequately prepared 65 molecules from traditional Chinese medicine with proven antiviral properties were subjected to docking analysis using AutoDock Vina 4 software with the aim to investigate binding affinity and interactions of compounds with Mpro from the SARS-CoV-2 virus. Biflavonoids and tannins show best docking scores with -9,80 kcal/mol for biflavonoids and -9,00 kcal/mol for tannins. Biflavonoids: amentoflavone, agathistaflavone, robustaflavone, hinokiflavone and rhusflavanone were tested for their radical scavenging activity. Partition coefficients were examined by RP-HPLC. Evaluation of drug-likeness properties of investigated biflavonoids suggested rhusflavanone as a molecule with the best ADMET characteristics. Anti-inflammatory activity of rhusflavanone was investigated in LPS stimulated RAW264.7 macrophages. Tested biflavonoids exibit beneficial effects against inflammation by scavenging free radicals and by suppressing the production of proinflammatory mediators by macrophages. Both predictions of affinity spectra for substances (PASS) and in vitro testing showed promising biological activity of investigated biflavonoids. A Quantum chemical study was performed in order to calculate the thermodynamic, molecular orbital, and electrostatic potential of selected molecules and to compare their biological and chemical features. Our results highlighted antioxidant, anti-inflammatory and antiviral properties of investigated compounds, emphasizing the significance of biflavonoid moiety to selected characteristics, which encourage further investigational strategies against COVID-19.
Collapse
Affiliation(s)
| | - Mohammed Mahbubul Matin
- Bioorganic and Medicinal Chemistry Laboratory, Department of Chemistry, Faculty of Science, University of Chittagong, Chattogram 4331, Bangladesh
| | - Ren Kong
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Milica Paut Kusturica
- University of Novi Sad, Faculty of Medicine, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Hao Zhang
- Graduate School of Hebei, Medical University, Shijiazhuang 050017, China
| |
Collapse
|
22
|
Hernandez AM, Mossman JA, Toapanta FR, Previte DM, Ross TM, Nau GJ. Altered transcriptional responses in the lungs of aged mice after influenza infection. Immun Ageing 2022; 19:27. [PMID: 35650631 PMCID: PMC9158162 DOI: 10.1186/s12979-022-00286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 05/25/2022] [Indexed: 12/02/2022]
Abstract
Background Influenza causes a serious infection in older individuals who are at the highest risk for mortality from this virus. Changes in the immune system with age are well known. This study used transcriptomic analysis to evaluate how aging specifically affects the functional host response to influenza in the lung. Adult (12–16 weeks) and aged (72–76 weeks) mice were infected with influenza and lungs were processed for RNA analysis. Results Older mice demonstrated a delayed anti-viral response on the level of transcription compared to adults, similar to the immunologic responses measured in prior work. The transcriptional differences, however, were evident days before observable differences in the protein responses described previously. The transcriptome response to influenza in aged mice was dominated by immunoglobulin genes and B cell markers compared to adult animals, suggesting immune dysregulation. Despite these differences, both groups of mice had highly similar transcriptional responses involving non-immune genes one day after inoculation and T cell genes during resolution. Conclusions These results define a delayed and dysregulated immune response in the lungs of aged mice infected with influenza. The findings implicate B cells and immunoglobulins as markers or mechanisms of immune aging. In addition to discovering new therapeutic targets, the findings underscore the value of transcription studies and network analysis to characterize complex biological processes, and serve as a model to analyze the susceptibility of the elderly to infectious agents. Supplementary information The online version contains supplementary material available at 10.1186/s12979-022-00286-9.
Collapse
|
23
|
Yang S, Wang L, Pan X, Liang Y, Zhang Y, Li J, Zhou B. 5-Methoxyflavone-induced AMPKα activation inhibits NF-κB and P38 MAPK signaling to attenuate influenza A virus-mediated inflammation and lung injury in vitro and in vivo. Cell Mol Biol Lett 2022; 27:82. [PMID: 36180831 PMCID: PMC9524045 DOI: 10.1186/s11658-022-00381-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/02/2022] [Indexed: 11/23/2022] Open
Abstract
Influenza-related acute lung injury (ALI) is a life-threatening condition that results mostly from uncontrolled replication of influenza virus (IV) and severe proinflammatory responses. The methoxy flavonoid compound 5-methoxyflavone (5-MF) is believed to have superior biological activity in the treatment of cancer. However, the effects and underlying mechanism of 5-MF on IV-mediated ALI are still unclear. Here, we showed that 5-MF significantly improved the survival of mice with lethal IV infection and ameliorated IV-mediated lung edema, lung histological changes, and inflammatory cell lung recruitment. We found that 5-MF has antiviral activity against influenza A virus (IAV), which was probably associated with increased expression of radical S-adenosyl methionine domain containing 2 (RSAD2) and suppression of endosomal acidification. Moreover, IV-infected A549 cells with 5-MF treatment markedly reduced proinflammatory mediator expression (IL-6, CXCL8, TNF-α, CXCL10, CCL2, CCL3, CCL4, GM-CSF, COX-2, and PGE2) and prevented P-IKBα, P-P65, and P-P38 activation. Interestingly, we demonstrated that 5-MF treatment could trigger activation of AMP-activated protein kinase (AMPK)α in IV-infected A549 cells, as evidenced by activation of the AMPKα downstream molecule P53. Importantly, the addition of AMPKα blocker compound C dramatically abolished 5-MF-mediated increased levels of RSAD2, the inhibitory effects on H1N1 virus-elicited endosomal acidification, and the suppression expression of proinflammatory mediators (IL-6, TNF-α, CXCL10, COX-2 and PGE2), as well as the inactivation of P-IKBα, P-P65, and P-P38 MAPK signaling pathways. Furthermore, inhibition of AMPKα abrogated the protective effects of 5-MF on H1N1 virus-mediated lung injury and excessive inflammation in vivo. Taken together, these results indicate that 5-MF alleviated IV-mediated ALI and suppressed excessive inflammatory responses through activation of AMPKα signaling.
Collapse
Affiliation(s)
- Sushan Yang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | | | | | - Yueyun Liang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Yuehan Zhang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China. .,Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou, 525200, China.
| |
Collapse
|
24
|
Žarković N, Jastrząb A, Jarocka-Karpowicz I, Orehovec B, Baršić B, Tarle M, Kmet M, Lukšić I, Łuczaj W, Skrzydlewska E. The Impact of Severe COVID-19 on Plasma Antioxidants. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165323. [PMID: 36014561 PMCID: PMC9416063 DOI: 10.3390/molecules27165323] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022]
Abstract
Several studies suggested the association of COVID-19 with systemic oxidative stress, in particular with lipid peroxidation and vascular stress. Therefore, this study aimed to evaluate the antioxidant signaling in the plasma of eighty-eight patients upon admission to the Clinical Hospital Dubrava in Zagreb, of which twenty-two died within a week, while the other recovered. The differences between the deceased and the survivors were found, especially in the reduction of superoxide dismutases (SOD-1 and SOD-2) activity, which was accompanied by the alteration in glutathione-dependent system and the intensification of the thioredoxin-dependent system. Reduced levels of non-enzymatic antioxidants, especially tocopherol, were also observed, which correlated with enhanced lipid peroxidation (determined by 4-hydroxynonenal (4-HNE) and neuroprostane levels) and oxidative modifications of proteins assessed as 4-HNE-protein adducts and carbonyl groups. These findings confirm the onset of systemic oxidative stress in patients with severe SARS-CoV-2, especially those who died from COVID-19, as manifested by strongly reduced tocopherol level and SOD activity associated with lipid peroxidation. Therefore, we propose that preventive and/or supplementary use of antioxidants, especially of lipophilic nature, could be beneficial for the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Neven Žarković
- Laboratory for Oxidative Stress (LabOS), Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
- Correspondence:
| | - Anna Jastrząb
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Iwona Jarocka-Karpowicz
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Biserka Orehovec
- Clinical Department of Laboratory Diagnostics, Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia
| | - Bruno Baršić
- Department of Internal Medicine, Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia
| | - Marko Tarle
- Department of Maxillofacial Surgery, Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia
| | - Marta Kmet
- Clinical Department of Laboratory Diagnostics, Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia
| | - Ivica Lukšić
- Department of Maxillofacial Surgery, Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, HR-10000 Zagreb, Croatia
| | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
25
|
Yegiazaryan A, Abnousian A, Alexander LJ, Badaoui A, Flaig B, Sheren N, Aghazarian A, Alsaigh D, Amin A, Mundra A, Nazaryan A, Guilford FT, Venketaraman V. Recent Developments in the Understanding of Immunity, Pathogenesis and Management of COVID-19. Int J Mol Sci 2022; 23:9297. [PMID: 36012562 PMCID: PMC9409103 DOI: 10.3390/ijms23169297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 02/03/2023] Open
Abstract
Coronaviruses represent a diverse family of enveloped positive-sense single stranded RNA viruses. COVID-19, caused by Severe Acute Respiratory Syndrome Coronavirus-2, is a highly contagious respiratory disease transmissible mainly via close contact and respiratory droplets which can result in severe, life-threatening respiratory pathologies. It is understood that glutathione, a naturally occurring antioxidant known for its role in immune response and cellular detoxification, is the target of various proinflammatory cytokines and transcription factors resulting in the infection, replication, and production of reactive oxygen species. This leads to more severe symptoms of COVID-19 and increased susceptibility to other illnesses such as tuberculosis. The emergence of vaccines against COVID-19, usage of monoclonal antibodies as treatments for infection, and implementation of pharmaceutical drugs have been effective methods for preventing and treating symptoms. However, with the mutating nature of the virus, other treatment modalities have been in research. With its role in antiviral defense and immune response, glutathione has been heavily explored in regard to COVID-19. Glutathione has demonstrated protective effects on inflammation and downregulation of reactive oxygen species, thereby resulting in less severe symptoms of COVID-19 infection and warranting the discussion of glutathione as a treatment mechanism.
Collapse
Affiliation(s)
- Aram Yegiazaryan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Arbi Abnousian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Logan J. Alexander
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Ali Badaoui
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Brandon Flaig
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Nisar Sheren
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Armin Aghazarian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Dijla Alsaigh
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Arman Amin
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Akaash Mundra
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Anthony Nazaryan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
26
|
Deng H, Li Y, Li J, Shen W, Chen Q, Weng S, He J, Xu X. Neomycin inhibits Megalocytivirus infection in fish by antagonizing the increase of intracellular reduced glutathione. FISH & SHELLFISH IMMUNOLOGY 2022; 127:148-154. [PMID: 35714896 DOI: 10.1016/j.fsi.2022.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/04/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Infectious spleen and kidney necrosis virus (ISKNV) is the type species of the Megalocytivirus genus that infects a number of marine and freshwater fishes, causing huge economic losses in aquaculture. The ISKNV infection leads to increase of reducing power in cells. As the antibiotic neomycin can promote the production of reactive oxygen species (ROS) in animal cells, in the current study, the potential therapeutic effect of neomycin on ISKNV infection was explored. We showed that neomycin could decrease the reducing power in cultured MFF-1 cells and inhibit ISKNV infection by antagonizing the shift of the cellular redox balance toward reduction. In vivo experiments further demonstrated that neomycin treatment significantly suppresses ISKNV infection in mandarin fish. Expression of the major capsid protein (MCP) and the proportion of infected cells in tissues were down-regulated after neomycin treatment. Furthermore, neomycin showed complex effects on expression of a set of antiviral related genes of the host. Taking together, the current study suggested that the viral-induced redox imbalance in the infected cells could be used as a target for suppressing ISKNV infection. Neomycin can be potentially utilized for therapeutic treatment of Megalocytivirus diseases by antagonizing intracellular redox changes.
Collapse
Affiliation(s)
- Hengwei Deng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
| | - Yeyu Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Jinling Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Wenjie Shen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Qiankang Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
27
|
Abstract
The first appearance of SARS-CoV-2 is dated back to 2019. This new member of the coronavirus family has caused more than 5 million deaths worldwide up until the end of January 2022. At the moment, and after intensive vaccination programmes throughout the world, the pandemic is still active, whilst new mutations constantly appear. Researchers are working intensively to discover antiviral drugs to combat the severe cases in intensive care units, giving the overloaded hospital units a breather. Alongside various research projects focusing on developing small pharmaceutical molecules, a significant proportion of the research community has shifted towards paying attention to metal drugs. In this small review, we make brief reference to the use of metal drugs in therapeutics and provide some examples of metal drugs that are of extreme interest in the current pandemic. At the same time, we will also examine some of their promising mechanisms of action and possible effectiveness against COVID-19.
Collapse
Affiliation(s)
- Kyriacos Ioannou
- Department of Life and Health Sciences, University of Nicosia, 2417, Nicosia, Cyprus
| | - Manos C Vlasiou
- Department of Life and Health Sciences, University of Nicosia, 2417, Nicosia, Cyprus.
| |
Collapse
|
28
|
Wang L, Cao Z, Wang Z, Guo J, Wen J. Reactive oxygen species associated immunoregulation post influenza virus infection. Front Immunol 2022; 13:927593. [PMID: 35967412 PMCID: PMC9373727 DOI: 10.3389/fimmu.2022.927593] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
An appropriate level of reactive oxygen species (ROS) is necessary for cell proliferation, signaling transduction, and apoptosis due to their highly reactive character. ROS are generated through multiple metabolic pathways under a fine-tuned control between oxidant and antioxidant signaling. A growing number of evidence has proved their highly relevant role in modulating inflammation during influenza virus infection. As a network of biological process for protecting organism from invasion of pathogens, immune system can react and fight back through either innate immune system or adaptive immune system, or both. Herein, we provide a review about the mechanisms of ROS generation when encounter influenza virus infection, and how the imbalanced level of ROS influences the replication of virus. We also summarize the pathways used by both the innate and adaptive immune system to sense and attack the invaded virus and abnormal levels of ROS. We further review the limitation of current strategies and discuss the direction of future work.
Collapse
Affiliation(s)
- Lan Wang
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States,UCLA Acquired Immune Deficiency Syndrome (AIDS) Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Zheng Cao
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Zi Wang
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Jimin Guo
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States,UCLA Acquired Immune Deficiency Syndrome (AIDS) Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States,UCLA Acquired Immune Deficiency Syndrome (AIDS) Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States,*Correspondence: Jing Wen,
| |
Collapse
|
29
|
Keskinidou C, Vassiliou AG, Dimopoulou I, Kotanidou A, Orfanos SE. Mechanistic Understanding of Lung Inflammation: Recent Advances and Emerging Techniques. J Inflamm Res 2022; 15:3501-3546. [PMID: 35734098 PMCID: PMC9207257 DOI: 10.2147/jir.s282695] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury characterized by an acute inflammatory response in the lung parenchyma. Hence, it is considered as the most appropriate clinical syndrome to study pathogenic mechanisms of lung inflammation. ARDS is associated with increased morbidity and mortality in the intensive care unit (ICU), while no effective pharmacological treatment exists. It is very important therefore to fully characterize the underlying pathobiology and the related mechanisms, in order to develop novel therapeutic approaches. In vivo and in vitro models are important pre-clinical tools in biological and medical research in the mechanistic and pathological understanding of the majority of diseases. In this review, we will present data from selected experimental models of lung injury/acute lung inflammation, which have been based on clinical disorders that can lead to the development of ARDS and related inflammatory lung processes in humans, including ventilation-induced lung injury (VILI), sepsis, ischemia/reperfusion, smoke, acid aspiration, radiation, transfusion-related acute lung injury (TRALI), influenza, Streptococcus (S.) pneumoniae and coronaviruses infection. Data from the corresponding clinical conditions will also be presented. The mechanisms related to lung inflammation that will be covered are oxidative stress, neutrophil extracellular traps, mitogen-activated protein kinase (MAPK) pathways, surfactant, and water and ion channels. Finally, we will present a brief overview of emerging techniques in the field of omics research that have been applied to ARDS research, encompassing genomics, transcriptomics, proteomics, and metabolomics, which may recognize factors to help stratify ICU patients at risk, predict their prognosis, and possibly, serve as more specific therapeutic targets.
Collapse
Affiliation(s)
- Chrysi Keskinidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Alice G Vassiliou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Stylianos E Orfanos
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| |
Collapse
|
30
|
El-Lateef HMA, El-Dabea T, Khalaf MM, Abu-Dief AM. Development of Metal Complexes for Treatment of Coronaviruses. Int J Mol Sci 2022; 23:6418. [PMID: 35742870 PMCID: PMC9223400 DOI: 10.3390/ijms23126418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/29/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Coronavirus disease (SARS-CoV-2) is a global epidemic. This pandemic, which has been linked to high rates of death, has forced some countries throughout the world to implement complete lockdowns in order to contain the spread of infection. Because of the advent of new coronavirus variants, it is critical to find effective treatments and vaccines to prevent the virus's rapid spread over the world. In this regard, metal complexes have attained immense interest as antibody modifiers and antiviral therapies, and they have a lot of promise towards SARS-CoV-2 and their suggested mechanisms of action are discussed, i.e., a new series of metal complexes' medicinal vital role in treatment of specific proteins or SARS-CoV-2 are described. The structures of the obtained metal complexes were fully elucidated by different analytical and spectroscopic techniques also. Molecular docking and pharmacophore studies presented that most of complexes studied influenced good binding affinity to the main protease SARS-CoV-2, which also was attained as from the RCSB pdb (Protein Data Bank) data PDB ID: 6 W41, to expect the action of metal complexes in contradiction of COVID-19. Experimental research is required to determine the pharmacokinetics of most of the complexes analyzed for the treatment of SARS-CoV-2-related disease. Finally, the toxicity of a metal-containing inorganic complex will thus be discussed by its capability to transfer metals which may bind with targeted site.
Collapse
Affiliation(s)
- Hany M. Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Tarek El-Dabea
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Mai M. Khalaf
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Ahmed M. Abu-Dief
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
- Chemistry Department, College of Science, Taibah University, Madinah 344, Saudi Arabia
| |
Collapse
|
31
|
Kumova OK, Galani IE, Rao A, Johnson H, Triantafyllia V, Matt SM, Pascasio J, Gaskill PJ, Andreakos E, Katsikis PD, Carey AJ. Severity of neonatal influenza infection is driven by type I interferon and oxidative stress. Mucosal Immunol 2022; 15:1309-1320. [PMID: 36352099 PMCID: PMC9724789 DOI: 10.1038/s41385-022-00576-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 09/26/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
Neonates exhibit increased susceptibility to respiratory viral infections, attributed to inflammation at the developing pulmonary air-blood interface. IFN I are antiviral cytokines critical to control viral replication, but also promote inflammation. Previously, we established a neonatal murine influenza virus (IV) model, which demonstrates increased mortality. Here, we sought to determine the role of IFN I in this increased mortality. We found that three-day-old IFNAR-deficient mice are highly protected from IV-induced mortality. In addition, exposure to IFNβ 24 h post IV infection accelerated death in WT neonatal animals but did not impact adult mortality. In contrast, IFN IIIs are protective to neonatal mice. IFNβ induced an oxidative stress imbalance specifically in primary neonatal IV-infected pulmonary type II epithelial cells (TIIEC), not in adult TIIECs. Moreover, neonates did not have an infection-induced increase in antioxidants, including a key antioxidant, superoxide dismutase 3, as compared to adults. Importantly, antioxidant treatment rescued IV-infected neonatal mice, but had no impact on adult morbidity. We propose that IFN I exacerbate an oxidative stress imbalance in the neonate because of IFN I-induced pulmonary TIIEC ROS production coupled with developmentally regulated, defective antioxidant production in response to IV infection. This age-specific imbalance contributes to mortality after respiratory infections in this vulnerable population.
Collapse
Affiliation(s)
- Ogan K. Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ioanna-Evdokia Galani
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Abhishek Rao
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Hannah Johnson
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Stephanie M. Matt
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Judy Pascasio
- Pathology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Peter J. Gaskill
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Peter D. Katsikis
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alison J. Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States.,Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
32
|
Salmin VV, Morgun AV, Olovyannikova RY, Kutyakov VA, Lychkovskaya EV, Brusina EB, Salmina AB. Atmospheric Reactive Oxygen Species and Some Aspects of the Antiviral Protection at the Respiratory Epithelium. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2022; 16:79-90. [PMID: 35601461 PMCID: PMC9113385 DOI: 10.1134/s1990750822020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022]
Affiliation(s)
- V. V. Salmin
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, ul. Partizana Zheleznyaka 1, 660022 Krasnoyarsk, Russia
| | - A. V. Morgun
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, ul. Partizana Zheleznyaka 1, 660022 Krasnoyarsk, Russia
| | - R. Ya. Olovyannikova
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, ul. Partizana Zheleznyaka 1, 660022 Krasnoyarsk, Russia
| | - V. A. Kutyakov
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, ul. Partizana Zheleznyaka 1, 660022 Krasnoyarsk, Russia
| | - E. V. Lychkovskaya
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, ul. Partizana Zheleznyaka 1, 660022 Krasnoyarsk, Russia
| | - E. B. Brusina
- Kemerovo State Medical University, ul. Voroshilova 22A, 650056 Kemerovo, Russia
| | - A. B. Salmina
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, ul. Partizana Zheleznyaka 1, 660022 Krasnoyarsk, Russia
- Research Center of Neurology, Volokolamskoe shosse 80, 125367 Moscow, Russia
| |
Collapse
|
33
|
Hendricks KS, To EE, Luong R, Liong F, Erlich JR, Shah AM, Liong S, O’Leary JJ, Brooks DA, Vlahos R, Selemidis S. Endothelial NOX4 Oxidase Negatively Regulates Inflammation and Improves Morbidity During Influenza A Virus Lung Infection in Mice. Front Cell Infect Microbiol 2022; 12:883448. [PMID: 35601109 PMCID: PMC9115386 DOI: 10.3389/fcimb.2022.883448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/07/2022] [Indexed: 12/02/2022] Open
Abstract
Endosomal NOX2 oxidase-dependent ROS production promotes influenza pathogenicity, but the role of NOX4 oxidase, which is highly expressed in the lung endothelium, is largely unknown. The aim of this study was to determine if endothelial NOX4 expression can influence viral pathology in vivo, using a mouse model of influenza infection. WT and transgenic endothelial NOX4 overexpressing mice (NOX4 TG) were infected intranasally with the Hong Kong H3N2 X-31 influenza A virus (104 PFU; HK x-31) or PBS control. Mice were culled at either 3 or 7 days post-infection to analyse: airway inflammation by bronchoalveolar lavage fluid (BALF) cell counts; NOX4, as well as inflammatory cytokine and chemokine gene expression by QPCR; and ROS production by an L-012-enhanced chemiluminescence assay. Influenza A virus infection of WT mice resulted in a significant reduction in lung NOX4 mRNA at day 3, which persisted until day 7, when compared to uninfected mice. Influenza A virus infection of NOX4 TG mice resulted in significantly less weight loss than that of WT mice at 3-days post infection. Viral titres were decreased in infected NOX4 TG mice compared to the infected WT mice, at both 3- and 7-days post infection and there was significantly less lung alveolitis, peri-bronchial inflammation and neutrophil infiltration. The oxidative burst from BALF inflammatory cells extracted from infected NOX4 TG mice was significantly less than that in the WT mice. Expression of macrophage and neutrophil chemoattractants CXCL10, CCL3, CXCL1 and CXCL2 in the lung tissue were significantly lower in NOX4 TG mice compared to the WT mice at 3-days post infection. We conclude that endothelial NOX4 oxidase is protective against influenza morbidity and is a potential target for limiting influenza A virus-induced lung inflammation.
Collapse
Affiliation(s)
- Keshia S. Hendricks
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Eunice E. To
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Raymond Luong
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Felicia Liong
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Jonathan R. Erlich
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Ajay M. Shah
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Stella Liong
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, St. James’s Hospital Dublin, Dublin, Ireland
| | - Doug A. Brooks
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| |
Collapse
|
34
|
Metabolic Response to Tick-Borne Encephalitis Virus Infection and Bacterial Co-Infections. Pathogens 2022; 11:pathogens11040384. [PMID: 35456059 PMCID: PMC9030592 DOI: 10.3390/pathogens11040384] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
Ticks are vectors of various pathogens, including tick-borne encephalitis virus and bacteria such as B. burgdorferi and A. phagocytophilum, causing infections/co-infections, which are still a diagnostic and therapeutic problem. Therefore, the aim of this study was to compare the effects of TBEV infection/bacterial co-infection on metabolic changes in the blood of patients before and after treatment. It was found that those infections promote plasma ROS enhanced generation and antioxidant defence reduction, especially in relation to glutathione and thioredoxin systems, despite the increased effectiveness of Nrf2 transcription factor in granulocytes. Observed oxidative stress promotes the oxidative modifications of phospholipids containing polyunsaturated fatty acids (LA, AA, EPA) with increased lipid peroxidation (estimated as 8-isoPGF2α, 4-HNE). It is accompanied by protein modifications measured as 4-HNE-protein adducts, carbonyl groups, dityrosine increase, and tryptophan level decrease, which promote structural and functional modification of the following transcription factors: Nrf2 and NFkB inhibitors. The lower level of 8-iso-PGF2α in co-infections indicates an impairment of the body’s ability to intensify inflammation and fight co-infections, while an increased level of Trx after therapy may contribute to the intensification of the inflammatory process. The obtained results indicate the potential possibility of using the assessed metabolic parameters to introduce targeted pharmacotherapy in cases of TBEV infections/bacterial co-infections.
Collapse
|
35
|
Allen CNS, Arjona SP, Santerre M, Sawaya BE. Hallmarks of Metabolic Reprogramming and Their Role in Viral Pathogenesis. Viruses 2022; 14:602. [PMID: 35337009 PMCID: PMC8955778 DOI: 10.3390/v14030602] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer and has proven to be critical in viral infections. Metabolic reprogramming provides the cell with energy and biomass for large-scale biosynthesis. Based on studies of the cellular changes that contribute to metabolic reprogramming, seven main hallmarks can be identified: (1) increased glycolysis and lactic acid, (2) increased glutaminolysis, (3) increased pentose phosphate pathway, (4) mitochondrial changes, (5) increased lipid metabolism, (6) changes in amino acid metabolism, and (7) changes in other biosynthetic and bioenergetic pathways. Viruses depend on metabolic reprogramming to increase biomass to fuel viral genome replication and production of new virions. Viruses take advantage of the non-metabolic effects of metabolic reprogramming, creating an anti-apoptotic environment and evading the immune system. Other non-metabolic effects can negatively affect cellular function. Understanding the role metabolic reprogramming plays in viral pathogenesis may provide better therapeutic targets for antivirals.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
- Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
36
|
Implication of COVID-19 on Erythrocytes Functionality: Red Blood Cell Biochemical Implications and Morpho-Functional Aspects. Int J Mol Sci 2022; 23:ijms23042171. [PMID: 35216286 PMCID: PMC8878454 DOI: 10.3390/ijms23042171] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Several diseases (such as diabetes, cancer, and neurodegenerative disorders) affect the morpho-functional aspects of red blood cells, sometimes altering their normal metabolism. In this review, the hematological changes are evaluated, with particular focus on the morphology and metabolic aspects of erythrocytes. Changes in the functionality of such cells may, in fact, help provide important information about disease severity and progression. The viral infection causes significant damage to the blood cells that are altered in size, rigidity, and distribution width. Lower levels of hemoglobin and anemia have been reported in several studies, and an alteration in the concentration of antioxidant enzymes has been shown to promote a dangerous state of oxidative stress in red blood cells. Patients with severe COVID-19 showed an increase in hematological changes, indicating a progressive worsening as COVID-19 severity progressed. Therefore, monitored hematological alterations in patients with COVID-19 may play an important role in the management of the disease and prevent the risk of a severe course of the disease. Finally, monitored changes in erythrocytes and blood, in general, may be one of the causes of the condition known as Long COVID.
Collapse
|
37
|
De Angelis M, Amatore D, Checconi P, Zevini A, Fraternale A, Magnani M, Hiscott J, De Chiara G, Palamara AT, Nencioni L. Influenza Virus Down-Modulates G6PD Expression and Activity to Induce Oxidative Stress and Promote Its Replication. Front Cell Infect Microbiol 2022; 11:804976. [PMID: 35071051 PMCID: PMC8770543 DOI: 10.3389/fcimb.2021.804976] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022] Open
Abstract
Influenza virus infection induces oxidative stress in host cells by decreasing the intracellular content of glutathione (GSH) and increasing reactive oxygen species (ROS) level. Glucose-6-phosphate dehydrogenase (G6PD) is responsible for the production of reducing equivalents of nicotinamide adenine dinucleotide phosphate (NADPH) that is used to regenerate the reduced form of GSH, thus restoring redox homeostasis. Cells deficient in G6PD display elevated levels of ROS and an increased susceptibility to viral infection, although the consequences of G6PD modulation during viral infection remain to be elucidated. In this study, we demonstrated that influenza virus infection decreases G6PD expression and activity, resulting in an increase in oxidative stress and virus replication. Moreover, the down regulation of G6PD correlated with a decrease in the expression of nuclear factor erythroid 2-related factor 2 (NRF2), a key transcription factor that regulates the expression of the antioxidant response gene network. Also down-regulated in influenza virus infected cells was sirtuin 2 (SIRT2), a NADPH-dependent deacetylase involved in the regulation of G6PD activity. Acetylation of G6PD increased during influenza virus infection in a manner that was strictly dependent on SIRT2 expression. Furthermore, the use of a pharmacological activator of SIRT2 rescued GSH production and NRF2 expression, leading to decreased influenza virus replication. Overall, these data identify a novel strategy used by influenza virus to induce oxidative stress and to favor its replication in host cells. These observations furthermore suggest that manipulation of metabolic and oxidative stress pathways could define new therapeutic strategies to interfere with influenza virus infection.
Collapse
Affiliation(s)
- Marta De Angelis
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Donatella Amatore
- Scientific Department, Army Medical Center, Via di Santo Stefano Rotondo, Rome, Italy
| | - Paola Checconi
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, IRCCS San Raffaele Roma, Rome, Italy
| | - Alessandra Zevini
- Pasteur Laboratory, Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | | | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - John Hiscott
- Pasteur Laboratory, Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Anna Teresa Palamara
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.,Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Nencioni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
38
|
Abstract
Abstract
Viruses completely rely on the energy and metabolic systems of host cells for life activities. Viral infections usually lead to cytopathic effects and host diseases. To date, there are still no specific clinical vaccines or drugs against most viral infections. Therefore, understanding the molecular and cellular mechanisms of viral infections is of great significance to prevent and treat viral diseases. A variety of viral infections are related to the p38 MAPK signalling pathway, and p38 is an important host factor in virus-infected cells. Here, we introduce the different signalling pathways of p38 activation and then summarise how different viruses induce p38 phosphorylation. Finally, we provide a general summary of the effect of p38 activation on virus replication. Our review provides integrated data on p38 activation and viral infections and describes the potential application of targeting p38 as an antiviral strategy.
Collapse
|
39
|
Bellanti F, Lo Buglio A, Vendemiale G. Redox Homeostasis and Immune Alterations in Coronavirus Disease-19. BIOLOGY 2022; 11:159. [PMID: 35205026 PMCID: PMC8869285 DOI: 10.3390/biology11020159] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023]
Abstract
The global Coronavirus Disease 2019 (COVID-19) pandemic is characterized by a wide variety of clinical features, from no or moderate symptoms to severe illness. COVID-19 is caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) that first affects the respiratory tract. Other than being limited to lungs, SARS-CoV-2 may lead to a multisystem disease that can even be durable (long COVID). The clinical spectrum of COVID-19 depends on variability in the immune regulation. Indeed, disease progression is consequent to failure in the immune regulation, characterized by an intensification of the pro-inflammatory response. Disturbance of systemic and organ-related redox balance may be a further mechanism underlying variability in COVID-19 severity. Other than being determinant for SARS-CoV-2 entry and fusion to the host cell, reactive species and redox signaling are deeply involved in the immune response. This review sums up the present knowledge on the role of redox balance in the regulation of susceptibility to SARS-CoV-2 infection and related immune response, debating the effectiveness of antioxidant compounds in the management of COVID-19.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (A.L.B.); (G.V.)
| | | | | |
Collapse
|
40
|
Mahdavinezhad F, Farmani AR, Pakniat H, Taghavi S, Gharaei R, Valipour J, Amidi F. COVID-19 and varicocele: the possible overlap factors and the common therapeutic approaches. Am J Reprod Immunol 2021; 87:e13518. [PMID: 34967487 DOI: 10.1111/aji.13518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/02/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Varicocele is recognized as one of the main attributable causes of male infertility which can affect spermatogenesis by various pathophysiological mechanisms. Recent studies have identified oxidative stress and reduction in antioxidant, hyperthermia, hypoxia, hormonal dysfunction, and inflammatory conditions as major factors in the pathophysiology of varicocele, all of which have known direct associations with the coronavirus disease 2019 (COVID-19) and can significantly increase the risk of detrimental COVID-19-related outcomes. Emerging data have shown an association between COVID-19 and inflammation, overproduction of cytokine, and other pathophysiological processes. The present review, summarizes the current understanding of the pathophysiology of varicocele and investigates the potential correlation between the severity of COVID-19 and the varicocele disease. In addition, various possible treatments which can be effective in both diseases were examined. Despite numerous challenges associated with the prevalence of COVID-19 in healthcare systems in infected countries, special attention should be given to maintaining a high level of care for complex patients with a pre-existing disease such as varicocele and providing appropriate practical advice for optimal control of the COVID-19 disease. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Forough Mahdavinezhad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Farmani
- Department of Tissue Engineering, Faculty of Advanced Technologies, Tehran University of Medical Sciences, Tehran, Iran.,Tissue Engineering Department, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamideh Pakniat
- Department of Obstetrics and Gynecology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Saeed Taghavi
- Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Roghaye Gharaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamal Valipour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Vicenti I, Martina MG, Boccuto A, De Angelis M, Giavarini G, Dragoni F, Marchi S, Trombetta CM, Crespan E, Maga G, Eydoux C, Decroly E, Montomoli E, Nencioni L, Zazzi M, Radi M. System-oriented optimization of multi-target 2,6-diaminopurine derivatives: Easily accessible broad-spectrum antivirals active against flaviviruses, influenza virus and SARS-CoV-2. Eur J Med Chem 2021; 224:113683. [PMID: 34273661 PMCID: PMC8255191 DOI: 10.1016/j.ejmech.2021.113683] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/16/2021] [Accepted: 07/01/2021] [Indexed: 11/29/2022]
Abstract
The worldwide circulation of different viruses coupled with the increased frequency and diversity of new outbreaks, strongly highlight the need for new antiviral drugs to quickly react against potential pandemic pathogens. Broad-spectrum antiviral agents (BSAAs) represent the ideal option for a prompt response against multiple viruses, new and re-emerging. Starting from previously identified anti-flavivirus hits, we report herein the identification of promising BSAAs by submitting the multi-target 2,6-diaminopurine chemotype to a system-oriented optimization based on phenotypic screening on cell cultures infected with different viruses. Among the synthesized compounds, 6i showed low micromolar potency against Dengue, Zika, West Nile and Influenza A viruses (IC50 = 0.5-5.3 μM) with high selectivity index. Interestingly, 6i also inhibited SARS-CoV-2 replication in different cell lines, with higher potency on Calu-3 cells that better mimic the SARS-CoV-2 infection in vivo (IC50 = 0.5 μM, SI = 240). The multi-target effect of 6i on flavivirus replication was also analyzed in whole cell studies (in vitro selection and immunofluorescence) and against isolated host/viral targets.
Collapse
Affiliation(s)
- Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Adele Boccuto
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Giorgia Giavarini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Filippo Dragoni
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Emmanuele Crespan
- Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Giovanni Maga
- Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Cecilia Eydoux
- AFMB, CNRS, Université Aix-Marseille, UMR 7257, Marseille, France
| | - Etienne Decroly
- AFMB, CNRS, Université Aix-Marseille, UMR 7257, Marseille, France
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; VisMederi s.r.l, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy.
| |
Collapse
|
42
|
Martín-Fernández M, Aller R, Heredia-Rodríguez M, Gómez-Sánchez E, Martínez-Paz P, Gonzalo-Benito H, Sánchez-de Prada L, Gorgojo Ó, Carnicero-Frutos I, Tamayo E, Tamayo-Velasco Á. Lipid peroxidation as a hallmark of severity in COVID-19 patients. Redox Biol 2021; 48:102181. [PMID: 34768063 PMCID: PMC8572041 DOI: 10.1016/j.redox.2021.102181] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Oxidative stress may be a key player in COVID-19 pathogenesis due to its significant role in response to infections. A defective redox balance has been related to viral pathogenesis developing a massive induction of cell death provoked by oxidative stress. The aim of this study is to perform a complete oxidative stress profile evaluation regarding antioxidant enzymes, total antioxidant capacity and oxidative cell damage in order to characterize its role in diagnosis and severity of this disease. METHODS Blood samples were obtained from 108 COVID-19 patients and 28 controls and metabolites representative of oxidative stress were assessed. The association between lipid peroxidation and 28-day intubation/death risk was evaluated by multivariable regression analysis. Probability of intubation/death to day-28 was analyzed by using Kaplan-Meier curves and tested with the log-rank test. RESULTS Antioxidant enzymes (Superoxide dismutase (SOD) and Catalase) and oxidative cell damage (Carbonyl and Lipid peroxidation (LPO)) levels were significantly higher in COVID-19 patients while total antioxidant capacity (ABTS and FRAP) levels were lower in these patients. The comparison of oxidative stress molecules' levels across COVID-19 severity revealed that only LPO was statistically different between mild and intubated/death COVID-19 patients. COX multivariate regression analysis identified LPO levels over the OOP (LPO>1948.17 μM) as an independent risk factor for 28-day intubation/death in COVID-19 patients [OR: 2.57; 95% CI: 1.10-5.99; p = 0.029]. Furthermore, Kaplan-Meier curve analysis revealed that COVID-19 patients showing LPO levels above 1948.17 μM were intubated or died 8.4 days earlier on average (mean survival time 15.4 vs 23.8 days) when assessing 28-day intubation/death risk (p < 0.001). CONCLUSION These findings deepen our knowledge of oxidative stress status in SARS-CoV-2 infection, supporting its important role in COVID-19. In fact, higher lipid peroxidation levels are independently associated to a higher risk of intubation or death at 28 days in COVID-19 patients.
Collapse
Affiliation(s)
- Marta Martín-Fernández
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Department of Medicine, Dermatology and Toxicology, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Rocío Aller
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Department of Medicine, Dermatology and Toxicology, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain; Gastroenterology Department, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - María Heredia-Rodríguez
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain; Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Salamanca, 37007 Salamanca, Spain.
| | - Esther Gómez-Sánchez
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain; Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain.
| | - Pedro Martínez-Paz
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Hugo Gonzalo-Benito
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Institute of Health Sciences of Castile and Leon (IECSCYL), 42002 Soria, Spain
| | - Laura Sánchez-de Prada
- Department of Microbiology, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Óscar Gorgojo
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Institute of Health Sciences of Castile and Leon (IECSCYL), 42002 Soria, Spain
| | - Irene Carnicero-Frutos
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Institute of Health Sciences of Castile and Leon (IECSCYL), 42002 Soria, Spain
| | - Eduardo Tamayo
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain; Anesthesiology and Critical Care Department, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Álvaro Tamayo-Velasco
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain; Haematology and Hemotherapy Department, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| |
Collapse
|
43
|
De Santis R, Luca V, Näslund J, Ehmann RK, De Angelis M, Lundmark E, Nencioni L, Faggioni G, Fillo S, Amatore D, Regalbuto E, Molinari F, Petralito G, Wölfel R, Stefanelli P, Rezza G, Palamara AT, Antwerpen M, Forsman M, Lista F. Rapid inactivation of SARS-CoV-2 with LED irradiation of visible spectrum wavelengths. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2021; 8:100082. [PMID: 34729540 PMCID: PMC8552801 DOI: 10.1016/j.jpap.2021.100082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/29/2022] Open
Abstract
Difficulty in controlling SARS-CoV-2 transmission made the ability to inactivate viruses in aerosols and fomites to be an important and attractive risk reduction measure. Evidence that light frequencies have the ability to inhibit microorganisms has already been reported by many studies which, however, focused on ultraviolet (UV) wavelengths, which are known to induce potential injury in humans. In the present study, the effect on suspensions of SARS-CoV-2 of a Light Emitting Diode (LED) device capable of radiating frequencies in the non-hazardous visible light spectrum (VIS) was investigated. In order to evaluate the efficiency of viral inactivation, plaque assay and western blot of viral proteins were performed. The observed results showed a significant reduction in infectious particles that had been exposed to the LED irradiation of visible light. Furthermore, the analysis of the intracellular expression of viral proteins confirmed the inactivating effect of this irradiation technology. This in vitro study revealed for the first time the inactivation of SARS-CoV-2 through LED irradiation with multiple wavelengths of the visible spectrum. However additional and more in-depth studies can aim to demonstrate the data obtained during these experiments in different matrices, in mutable environmental conditions and on other respiratory viruses such as the influenza virus. The type of LED technology can decisively contribute on reducing virus transmission through the continuous sanitation of common environments without risks for humans and animals.
Collapse
Affiliation(s)
| | - Vincenzo Luca
- Scientific Department, Army Medical Center, Rome, Italy.,7th CBRN Defence Regiment "Cremona", Civitavecchia, Italy
| | - Jonas Näslund
- Department of CBRN Protection and Security, Swedish Defence Research Agency (FOI), Umeå, Sweden
| | - Rosina K Ehmann
- Section Viral and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, Italy
| | - Eva Lundmark
- Department of CBRN Protection and Security, Swedish Defence Research Agency (FOI), Umeå, Sweden
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, Italy
| | | | - Silvia Fillo
- Scientific Department, Army Medical Center, Rome, Italy
| | | | | | | | | | - Roman Wölfel
- Section Viral and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
| | - Paola Stefanelli
- Department of Infectious Disease, National Institute of Health, Rome, Italy
| | - Gianni Rezza
- Department of Infectious Disease, National Institute of Health, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, Italy
| | - Markus Antwerpen
- Section Viral and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
| | - Mats Forsman
- Department of CBRN Protection and Security, Swedish Defence Research Agency (FOI), Umeå, Sweden
| | | |
Collapse
|
44
|
Salmin VV, Morgun AV, Olovyannikova RY, Kutyakov VA, Lychkovskaya EV, Brusina EB, Salmina AB. [Atmospheric reactive oxygen species and some aspects of the antiviral protection of the respiratory epithelium]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:383-393. [PMID: 34730551 DOI: 10.18097/pbmc20216705383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review focuses on molecular and biochemical mechanisms of nonspecific protection of respiratory epithelium. The authors provide a comprehensive analysis of up-to-date data on the activity of the lactoperoxidase system expressed on the surface of the respiratory epithelium which provides the generation of hypothiocyanate and hypoiodite in the presence of locally produced or inhaled hydrogen peroxide. Molecular mechanisms of production of active compounds with antiviral and antibacterial effects, expression profiles of enzymes, transporters and ion channels involved in the generation of hypothiocyanite and hypoiodate in the mucous membrane of the respiratory system in physiological and pathological conditions (inflammation) are discussed. In the context of antibacterial and antiviral defense special attention is paid to recent data confirming the effects of atmospheric air composition on the efficiency of hypothiocyanite and hypoiodate synthesis in the respiratory epithelium. The causes and outcomes of lactoperoxidase system impairment due to the action of atmospheric factors are discussed in the context of controlling the sensitivity of the epithelium to the action of bacterial agents and viruses. Restoration of the lactoperoxidase system activity can be achieved by application of pharmacological agents aimed to compensate for the lack of halides in tissues, and by the control of chemical composition of the inhaled air.
Collapse
Affiliation(s)
- V V Salmin
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - A V Morgun
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - R Ya Olovyannikova
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - V A Kutyakov
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - E V Lychkovskaya
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - E B Brusina
- Kemerovo State Medical University, Kemerovo, Russia
| | - A B Salmina
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia; Research Center of Neurology, Moscow, Russia
| |
Collapse
|
45
|
Khatchadourian C, Sisliyan C, Nguyen K, Poladian N, Tian Q, Tamjidi F, Luong B, Singh M, Robison J, Venketaraman V. Hyperlipidemia and Obesity's Role in Immune Dysregulation Underlying the Severity of COVID-19 Infection. Clin Pract 2021; 11:694-707. [PMID: 34698139 PMCID: PMC8544571 DOI: 10.3390/clinpract11040085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Obesity and hyperlipidemia are known to be risk factors for various pathological disorders, including various forms of infectious respiratory disease, including the current Coronavirus outbreak termed Coronavirus Disease 19 (COVID-19). This review studies the effects of hyperlipidemia and obesity on enhancing the inflammatory response seen in COVID-19 and potential therapeutic pathways related to these processes. In order to better understand the underlying processes of cytokine and chemokine-induced inflammation, we must further investigate the immunomodulatory effects of agents such as Vitamin D and the reduced form of glutathione as adjunctive therapies for COVID-19 disease.
Collapse
Affiliation(s)
- Christopher Khatchadourian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Christina Sisliyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Kevin Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Nicole Poladian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Qi Tian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Faraaz Tamjidi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Bao Luong
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
| | - Manpreet Singh
- Department of Emergency Medicine, St. Barnabas Hospital Health System, Bronx, NY 10457, USA; (M.S.); (J.R.)
| | - Jeremiah Robison
- Department of Emergency Medicine, St. Barnabas Hospital Health System, Bronx, NY 10457, USA; (M.S.); (J.R.)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (C.K.); (C.S.); (K.N.); (N.P.); (Q.T.); (F.T.); (B.L.)
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E Second Street, Pomona, CA 91766, USA
| |
Collapse
|
46
|
Dutta M, Tareq AM, Rakib A, Mahmud S, Sami SA, Mallick J, Islam MN, Majumder M, Uddin MZ, Alsubaie A, Almalki ASA, Khandaker MU, Bradley D, Rana MS, Emran TB. Phytochemicals from Leucas zeylanica Targeting Main Protease of SARS-CoV-2: Chemical Profiles, Molecular Docking, and Molecular Dynamics Simulations. BIOLOGY 2021; 10:789. [PMID: 34440024 PMCID: PMC8389631 DOI: 10.3390/biology10080789] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/05/2021] [Accepted: 08/15/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a contemporary coronavirus, has impacted global economic activity and has a high transmission rate. As a result of the virus's severe medical effects, developing effective vaccinations is vital. Plant-derived metabolites have been discovered as potential SARS-CoV-2 inhibitors. The SARS-CoV-2 main protease (Mpro) is a target for therapeutic research because of its highly conserved protein sequence. Gas chromatography-mass spectrometry (GC-MS) and molecular docking were used to screen 34 compounds identified from Leucas zeylanica for potential inhibitory activity against the SARS-CoV-2 Mpro. In addition, prime molecular mechanics-generalized Born surface area (MM-GBSA) was used to screen the compound dataset using a molecular dynamics simulation. From molecular docking analysis, 26 compounds were capable of interaction with the SARS-CoV-2 Mpro, while three compounds, namely 11-oxa-dispiro[4.0.4.1]undecan-1-ol (-5.755 kcal/mol), azetidin-2-one 3,3-dimethyl-4-(1-aminoethyl) (-5.39 kcal/mol), and lorazepam, 2TMS derivative (-5.246 kcal/mol), exhibited the highest docking scores. These three ligands were assessed by MM-GBSA, which revealed that they bind with the necessary Mpro amino acids in the catalytic groove to cause protein inhibition, including Ser144, Cys145, and His41. The molecular dynamics simulation confirmed the complex rigidity and stability of the docked ligand-Mpro complexes based on the analysis of mean radical variations, root-mean-square fluctuations, solvent-accessible surface area, radius of gyration, and hydrogen bond formation. The study of the postmolecular dynamics confirmation also confirmed that lorazepam, 11-oxa-dispiro[4.0.4.1]undecan-1-ol, and azetidin-2-one-3, 3-dimethyl-4-(1-aminoethyl) interact with similar Mpro binding pockets. The results of our computerized drug design approach may assist in the fight against SARS-CoV-2.
Collapse
Affiliation(s)
- Mycal Dutta
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh; (M.D.); (J.M.); (M.Z.U.)
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Abu Montakim Tareq
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (A.M.T.); (M.N.I.)
| | - Ahmed Rakib
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (A.R.); (S.A.S.)
| | - Shafi Mahmud
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Saad Ahmed Sami
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh; (A.R.); (S.A.S.)
| | - Jewel Mallick
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh; (M.D.); (J.M.); (M.Z.U.)
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (A.M.T.); (M.N.I.)
| | - Mohuya Majumder
- Drug Discovery, GUSTO A Research Group, Chittagong 4203, Bangladesh;
| | - Md. Zia Uddin
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh; (M.D.); (J.M.); (M.Z.U.)
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Abdullah Alsubaie
- Department of Physics, College of Khurma, Taif University, Taif 21944, Saudi Arabia;
| | | | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Malaysia; (M.U.K.); (D.A.B.)
| | - D.A. Bradley
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Malaysia; (M.U.K.); (D.A.B.)
- Department of Physics, University of Surrey, Guilford GU2 7XH, UK
| | - Md. Sohel Rana
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh; (M.D.); (J.M.); (M.Z.U.)
| |
Collapse
|
47
|
Wang X, Jiang Z, Yue N, Jin X, Zhang X, Li Z, Zhang Y, Wang X, Han C, Yu J, Li D. Barley stripe mosaic virus γb protein disrupts chloroplast antioxidant defenses to optimize viral replication. EMBO J 2021; 40:e107660. [PMID: 34254679 PMCID: PMC8365260 DOI: 10.15252/embj.2021107660] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 01/21/2023] Open
Abstract
The plant antioxidant system plays important roles in response to diverse abiotic and biotic stresses. However, the effects of virus infection on host redox homeostasis and how antioxidant defense pathway is manipulated by viruses remain poorly understood. We previously demonstrated that the Barley stripe mosaic virus (BSMV) γb protein is recruited to the chloroplast by the viral αa replicase to enhance viral replication. Here, we show that BSMV infection induces chloroplast oxidative stress. The versatile γb protein interacts directly with NADPH-dependent thioredoxin reductase C (NTRC), a core component of chloroplast antioxidant systems. Overexpression of NbNTRC significantly impairs BSMV replication in Nicotiana benthamiana plants, whereas disruption of NbNTRC expression leads to increased viral accumulation and infection severity. To counter NTRC-mediated defenses, BSMV employs the γb protein to competitively interfere with NbNTRC binding to 2-Cys Prx. Altogether, this study indicates that beyond acting as a helicase enhancer, γb also subverts NTRC-mediated chloroplast antioxidant defenses to create an oxidative microenvironment conducive to viral replication.
Collapse
Affiliation(s)
- Xueting Wang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Zhihao Jiang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Ning Yue
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Xuejiao Jin
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Xuan Zhang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Zhaolei Li
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Yongliang Zhang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Xian‐Bing Wang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Chenggui Han
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Jialin Yu
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Dawei Li
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil MicrobiologyCollege of Biological SciencesChina Agricultural UniversityBeijingChina
| |
Collapse
|
48
|
Emanuele S, Celesia A, D’Anneo A, Lauricella M, Carlisi D, De Blasio A, Giuliano M. The Good and Bad of Nrf2: An Update in Cancer and New Perspectives in COVID-19. Int J Mol Sci 2021; 22:7963. [PMID: 34360732 PMCID: PMC8348506 DOI: 10.3390/ijms22157963] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 01/08/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a well-known transcription factor best recognised as one of the main regulators of the oxidative stress response. Beyond playing a crucial role in cell defence by transactivating cytoprotective genes encoding antioxidant and detoxifying enzymes, Nrf2 is also implicated in a wide network regulating anti-inflammatory response and metabolic reprogramming. Such a broad spectrum of actions renders the factor a key regulator of cell fate and a strategic player in the control of cell transformation and response to viral infections. The Nrf2 protective roles in normal cells account for its anti-tumour and anti-viral functions. However, Nrf2 overstimulation often occurs in tumour cells and a complex correlation of Nrf2 with cancer initiation and progression has been widely described. Therefore, if on one hand, Nrf2 has a dual role in cancer, on the other hand, the factor seems to display a univocal function in preventing inflammation and cytokine storm that occur under viral infections, specifically in coronavirus disease 19 (COVID-19). In such a variegate context, the present review aims to dissect the roles of Nrf2 in both cancer and COVID-19, two widespread diseases that represent a cause of major concern today. In particular, the review describes the molecular aspects of Nrf2 signalling in both pathological situations and the most recent findings about the advantages of Nrf2 inhibition or activation as possible strategies for cancer and COVID-19 treatment respectively.
Collapse
Affiliation(s)
- Sonia Emanuele
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.C.); (M.L.); (D.C.)
| | - Adriana Celesia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.C.); (M.L.); (D.C.)
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Biochemistry Building, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.D.); (A.D.B.); (M.G.)
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.C.); (M.L.); (D.C.)
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.C.); (M.L.); (D.C.)
| | - Anna De Blasio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Biochemistry Building, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.D.); (A.D.B.); (M.G.)
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Biochemistry Building, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (A.D.); (A.D.B.); (M.G.)
| |
Collapse
|
49
|
Kosanovic T, Sagic D, Djukic V, Pljesa-Ercegovac M, Savic-Radojevic A, Bukumiric Z, Lalosevic M, Djordjevic M, Coric V, Simic T. Time Course of Redox Biomarkers in COVID-19 Pneumonia: Relation with Inflammatory, Multiorgan Impairment Biomarkers and CT Findings. Antioxidants (Basel) 2021; 10:antiox10071126. [PMID: 34356359 PMCID: PMC8301049 DOI: 10.3390/antiox10071126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
Although the original data on systemic oxidative stress in COVID-19 patients have recently started to emerge, we are still far from a complete profile of changes in patients' redox homeostasis. We aimed to assess the extent of oxidative damage of proteins, lipids and DNA during the course of acute disease, as well as their association with CT pulmonary patterns. In order to obtain more insight into the origin of the systemic oxidative stress, the observed parameters were correlated with inflammatory biomarkers and biomarkers of multiorgan impairment. In this prospective study, we included 58 patients admitted between July and October 2020 with COVID-19 pneumonia. Significant changes in malondialdehyde, 8-hydroxy-2'-deoxyguanosine and advanced oxidation protein products levels exist during the course of COVID-19. Special emphasis should be placed on the fact that the pattern of changes differs between non-hospitalized and hospitalized individuals. Our results point to the time-dependent relation of oxidative stress parameters with inflammatory and multiorgan impairment biomarkers, as well as pulmonary patterns in COVID-19 pneumonia patients. Correlation between redox biomarkers and immunological or multiorgan impairment biomarkers, as well as pulmonary CT pattern, confirms the suggested involvement of neutrophils networks, IL-6 production, along with different organ/tissue involvement in systemic oxidative stress in COVID-19.
Collapse
Affiliation(s)
- Tijana Kosanovic
- Radiology Department, The University Hospital ‘Dr. Dragisa Misovic- Dedinje’, 11000 Belgrade, Serbia; (T.K.); (V.D.); (M.L.); (M.D.)
| | - Dragan Sagic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia;
| | - Vladimir Djukic
- Radiology Department, The University Hospital ‘Dr. Dragisa Misovic- Dedinje’, 11000 Belgrade, Serbia; (T.K.); (V.D.); (M.L.); (M.D.)
| | - Marija Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.P.-E.); (A.S.-R.)
| | - Ana Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.P.-E.); (A.S.-R.)
| | - Zoran Bukumiric
- Institute of Medical Statistics and Informatics, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Miodrag Lalosevic
- Radiology Department, The University Hospital ‘Dr. Dragisa Misovic- Dedinje’, 11000 Belgrade, Serbia; (T.K.); (V.D.); (M.L.); (M.D.)
| | - Marjana Djordjevic
- Radiology Department, The University Hospital ‘Dr. Dragisa Misovic- Dedinje’, 11000 Belgrade, Serbia; (T.K.); (V.D.); (M.L.); (M.D.)
| | - Vesna Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.P.-E.); (A.S.-R.)
- Correspondence: (V.C.); (T.S.); Tel.: +381-113643273 (V.C.); +381-113643250 (T.S.)
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.P.-E.); (A.S.-R.)
- Serbian Academy of Science and Arts, 11000 Belgrade, Serbia
- Correspondence: (V.C.); (T.S.); Tel.: +381-113643273 (V.C.); +381-113643250 (T.S.)
| |
Collapse
|
50
|
Yildiz H, Alp HH, Ekin S, Arisoy A, Gunbatar H, Asker S, Cilingir BM, Sunnetcioglu A, Celikel M, Esen N, Bedirhanoglu S, Baykal ND, Haylu M. Analysis of endogenous oxidative damage markers and association with pulmonary involvement severity in patients with SARS-CoV-2 pneumonia. Infect Dis Now 2021; 51:429-434. [PMID: 34146758 PMCID: PMC8236077 DOI: 10.1016/j.idnow.2021.06.302] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 01/05/2023]
Abstract
Introduction The SARS-CoV-2 virus affects many organs, especially the lungs, with widespread inflammation. We aimed to compare the endogenous oxidative damage markers of coenzyme Q10, nicotinamide dinucleotide oxidase 4, malondialdehyde, and ischemia-modified albumin levels in patients with pneumonia caused by SARS-CoV-2 and in an healthy control group. We also aimed to compare these parameters between patients with severe and non-severe pulmonary involvement. Methods The study included 58 adult patients with SARS-CoV-2 pneumonia and 30 healthy volunteers. CoQ10 and MDA levels were determined by high-pressure liquid chromatography. NOX4 and IMA levels were determined by ELISA assay and colorimetric method. Results Higher levels of CoQ10, MDA, NOX4, and IMA and lower levels of COQ10H were observed in patients with SARS-CoV-2 pneumonia than in the control group. MDA, IMA, NOX4, and CoQ10 levels were significantly higher in patients with severe pulmonary involvement than in patients with non-severe pulmonary involvement, but no significant difference was observed in CoQ10H levels. CoQ10 levels were significantly and positively correlated with both ferritin and CRP levels. Conclusion SARS-CoV-2 pneumonia is significantly associated with increased endogenous oxidative damage. Oxidative damage seems to be associated with pulmonary involvement severity.
Collapse
Affiliation(s)
- Hanifi Yildiz
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey.
| | - Hamit Hakan Alp
- Van Yüzüncü Yıl University, Faculty of Medicine, Department of Biochemistry, Van, Turkey
| | - Selami Ekin
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Ahmet Arisoy
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Hulya Gunbatar
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Selvi Asker
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Buket Mermit Cilingir
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Aysel Sunnetcioglu
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Masuk Celikel
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Nevzat Esen
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Serhat Bedirhanoglu
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Nuruban Delal Baykal
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| | - Mine Haylu
- Van Yüzüncü Yil University, Faculty of Medicine, Department of Chest Diseases, Van, Turkey
| |
Collapse
|