1
|
Domínguez-de-Barros A, Sirvent-Blanco C, García-Pérez O, Gajate-Arenas M, García-Ramos A, Migliazzo C, Piñero JE, Lorenzo-Morales J, Córdoba-Lanús E. Telomere Length, Oxidative Stress Markers, and Related miRNAs in Non-Invasive Samples of Mild COVID-19 Cases. Int J Mol Sci 2025; 26:4934. [PMID: 40430074 PMCID: PMC12111949 DOI: 10.3390/ijms26104934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Oxidative stress and inflammation influence immune response and epigenetic mechanisms in infectious diseases. In mild COVID-19, host-encoded miRNA profiles remain underexplored, although they reveal mechanistic insights into disease pathogenesis. This study evaluated ageing and oxidative stress biomarkers (telomere length (TL), TBARS, 8-OHdG, and circulating related-miRNA expression) in 75 mild cases and 30 non-COVID-19 controls. TL correlated with age (R = -0.384, p = 0.005) and was shorter in cases compared to controls (rTL 1.46 ± 0.51 vs. 0.99 ± 0.37; p < 0.001), being similar between saliva and blood samples (p = 0.917). miR-138-5p was upregulated in COVID-19 cases (p = 0.026) and correlated with 8-OHdG (R = 0.403, p = 0.05), which was increased in cases (p = 0.040); miR-210-3p was downregulated in infected individuals (p = 0.008), while miR-182-5p expression correlated with TBARS (R = 0.582, p = 0.018). miR-34a-5p and miR155-5p expression was not altered in mild COVID-19. These findings suggest early systemic cellular damage in mild COVID-19 and highlight miR-138-5p and miR-182-5p as potential early biomarkers of oxidative stress.
Collapse
Affiliation(s)
- Angélica Domínguez-de-Barros
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Candela Sirvent-Blanco
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
| | - Omar García-Pérez
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Malena Gajate-Arenas
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alma García-Ramos
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
| | - Claudia Migliazzo
- Dipartmento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Universita’ Degli Studi di Palermo, 90133 Palermo, Italy;
| | - José E. Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departmento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departmento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Elizabeth Córdoba-Lanús
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (A.D.-d.-B.); (C.S.-B.); (O.G.-P.); (M.G.-A.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departmento de Bioquímica, Microbiología, Biología Celular y Genética, Área Biología Celular, Facultad de Ciencias, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
| |
Collapse
|
2
|
Shahid Z, Naseer K, Hussain I, Qazi J. Lyophilized nasal swabs for COVID-19 detection by ATR-FTIR spectroscopy: Machine learning-based approach. Biophys Chem 2025; 324:107459. [PMID: 40381422 DOI: 10.1016/j.bpc.2025.107459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
The COVID-19 pandemic continues to pose challenges for global health. The disease burden and diagnostic pressure has forced scientists to explore alternate diagnostic tools beyond the standard PCR testing. One such promising tool is the use of spectroscopy-based diagnostics. The objective of this study is to assess the potential of ATR-FTIR spectroscopy, applied to lyophilized nasal swab samples to discriminate between healthy and infected COVID-19 patients. Equal number (55 each) of positive and negative freeze-dried nasal swab samples were analyzed. After pre-processing, average mean spectra (600-4000 cm-1) showed significant variations between healthy and infected sample types. Clear spectral variations were recorded at 17 locations, of which, 13 peaks were observed in COVID-19 spectra while 4 peaks were observed in negative sample spectra. Statistical discrimination was done using principal component analysis (PCA), linear discriminant analysis (LDA) and support vector machine (SVM). The first two principal components (PCs) showed a combined variance of 76 %. Classification accuracy of 100 % were observed in the LDA graph using Quadratic kernel. Similarly, SVM model with both internal validation and external validation confirmed the robustness with a 100 % classification accuracy. These results show that lyophilized nasal swab samples are the ideal sample choice for FTIR-based analysis of COVID-19. This sample preparation method coupled with spectroscopy can serve as a robust and accessible diagnostic tool for post-covid testing.
Collapse
Affiliation(s)
- Zubia Shahid
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Khulla Naseer
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Irshad Hussain
- Department of Chemistry & Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Javaria Qazi
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
3
|
Mara G, Nini G, Cotoraci C. Chronic Obstructive Pulmonary Disease and COVID-19: The Impact of Hematological Biomarkers on Disease Severity and Outcomes. J Clin Med 2025; 14:2765. [PMID: 40283596 PMCID: PMC12027599 DOI: 10.3390/jcm14082765] [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: 03/25/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Chronic obstructive pulmonary disease (COPD) patients are at heightened risk of severe COVID-19 due to underlying respiratory impairment, systemic inflammation, and immune dysregulation. This review explores the hematological changes that occur in COPD patients with COVID-19 and their implications for disease progression, prognosis, and clinical management. Methods: We conducted a comprehensive analysis of recent peer-reviewed studies from medical databases including Clarivate Analytics, PubMed, and Google Scholar. Results: Hematological alterations, such as lymphopenia, elevated neutrophil-to-lymphocyte ratio (NLR), increased D-dimer and fibrinogen levels, inflammatory anemia, and erythrocyte dysfunction, are commonly observed in COPD patients with COVID-19. These changes are linked to immune suppression, hyperinflammation, oxidative stress, and thromboembolic complications. Conclusions: Hematological biomarkers are valuable tools for early risk assessments and guiding treatment strategies in this high-risk population. The regular monitoring of D-dimer, fibrinogen, and NLR is advisable. Prophylactic anticoagulation and immunomodulatory therapies, such as corticosteroids and IL-6 and IL-1 inhibitors, may improve clinical outcomes. Further clinical studies are needed to validate personalized approaches and explore antioxidant-based interventions.
Collapse
Affiliation(s)
- Gabriela Mara
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Pneumology Department, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Gheorghe Nini
- Pneumology Department, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Coralia Cotoraci
- Clinical Hematology Department, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| |
Collapse
|
4
|
Başaran N, Szewczyk-Roszczenko O, Roszczenko P, Vassetzky Y, Sjakste N. Genotoxic risks in patients with COVID-19. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 129:105728. [PMID: 39955016 DOI: 10.1016/j.meegid.2025.105728] [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: 12/10/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
The COVID-19 pandemic has caused numerous deaths worldwide. Despite the mitigation of infection manifestations in recent months, the possible consequences of the epidemic remain difficult to predict. Genotoxicity and subsequent development of neoplasms are possible outcomes. This review summarises the data on these questions. Studies from several countries have reported increased levels of DNA damage in nucleated blood cells of patients with severe forms of COVID-19 infection. The level of DNA damage can be used as a prognostic factor for the disease outcome. It is considered that SARS-CoV-2 spike proteins play a crucial role in DNA damage; however, the virus also inhibits the DNA repair system. Co-morbidities and use of antiviral drugs may also contribute to DNA damage in patients with COVID-19.
Collapse
Affiliation(s)
- Nurşen Başaran
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Başkent University, Ankara 06790, Türkiye
| | - Olga Szewczyk-Roszczenko
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Roszczenko
- Department of Biotechnology, Medical University of Bialystok, Bialystok, Poland
| | - Yegor Vassetzky
- Chromatin Dynamics and Metabolism in Cancer, CNRS UMR9018 Institut Gustave Roussy, Univeristé Paris Saclay, 39, rue Camille-Desmoulins, 94805 Villejuif, France; Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia.
| | - Nikolajs Sjakste
- Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas Street 1, LV1004 Riga, Latvia.
| |
Collapse
|
5
|
Abiri E, Mirzaii M, Moghbeli M, Atashi A, Harati AA. Investigating DNA damage caused by COVID-19 and influenza in post COVID-19. Mamm Genome 2025; 36:200-212. [PMID: 39537997 DOI: 10.1007/s00335-024-10082-z] [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: 04/02/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The SARS-CoV-2 virus (termed COVID-19) was responsible for over 34 million global deaths. Although the COVID-19 pandemic has subsided, infection by emerging mutant variants of SARS-CoV-2 poses a continuing threat to public health. COVID-19 infection has been associated with the development of cytokine storm syndrome, hypercoagulability, immunological dysregulation and direct viral invasion of organs, and the long-term consequences for the health of COVID-19 survivors are currently unknown. Our research focuses on the possible mutagenic aspects of infection by COVID-19 and measures their harmful effects on DNA composition. DNA damage was investigated, using the comet assay method, during two periods: in the epidemic peak of COVID-19 and during the post-COVID-19 period, both in patients infected with COVID-19 and in those with influenza. During the epidemic peak, the levels of DNA damage ranged from the highest to the lowest levels in the following groups, respectively: intubated-ICU, non-intubated-ICU, non-ICU, and influenza, with a discernible increase in DNA damage in ICU-treated patients. The levels of DNA damage in the post-COVID-19 period were significantly lower compared to those in the epidemic peak period but there was still a discernible increase in DNA damage in the ICU group. Our results indicate that levels of DNA damage may be an effective indicator in prognostic decision-making and may therefore help to reduce mortality. Given that DNA damage and impaired repair processes can contribute to chronic diseases like diabetes, cancer, and neurodegenerative conditions, it will be crucial to investigate potential similar effects in patients with COVID-19.
Collapse
Affiliation(s)
- Elaheh Abiri
- Department of Biology, Islamic Azad University Damghan, Damghan Branch, Damghan, Iran
| | - Mehdi Mirzaii
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Majid Moghbeli
- Department of Biology, Islamic Azad University Damghan, Damghan Branch, Damghan, Iran
| | - Amir Atashi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahad Ali Harati
- Department of Biology, Islamic Azad University Damghan, Damghan Branch, Damghan, Iran
| |
Collapse
|
6
|
Szewczyk-Roszczenko O, Roszczenko P, Vassetzky Y, Sjakste N. Genotoxic consequences of viral infections. NPJ VIRUSES 2025; 3:5. [PMID: 40295867 PMCID: PMC11772741 DOI: 10.1038/s44298-024-00087-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/24/2024] [Indexed: 04/30/2025]
Abstract
Viral diseases continually threaten human health as evolving pathogens introduce new risks. These infections can lead to complications across organ systems, with impacts varying by virus type, infection severity, and individual immune response. This review examines the genotoxic stress caused by viral infections and its pathological consequences in humans.
Collapse
Affiliation(s)
- Olga Szewczyk-Roszczenko
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Roszczenko
- Department of Biotechnology, Medical University of Bialystok, Bialystok, Poland
| | - Yegor Vassetzky
- Chromatin Dynamics and Metabolism in Cancer, CNRS UMR9018 Institut Gustave Roussy, Univeristé Paris Saclay, 39, rue Camille-Desmoulins, 94805, Villejuif, France.
| | - Nikolajs Sjakste
- Department of Pharmacy, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas Street 1, LV1004, Riga, Latvia.
| |
Collapse
|
7
|
de Farias ECF, do Nascimento LMPP, Pavão Junior MJC, Pavão DCA, Pinheiro APS, Pinheiro AHO, Alves MCB, Ferraro KMMM, Aires LFQ, Dias LG, Machado MMM, Serrão MJD, Gomes RR, de Moraes SMP, Pontes GCL, Carvalho RDFP, Silva CTC, Neves CMAD, dos Santos JCL, de Sousa AMB, da Silva LL, de Mello MLFMF, Carvalho PB, Braga RDB, Harada KDO, Justino MCA, Costa IB, Brasil-Costa I, Monteiro MC, Clemente G, Terreri MT. Plasma IL-17A is increased in patients with critical MIS-C and associated to in-hospital mortality. Front Immunol 2025; 15:1485009. [PMID: 39931580 PMCID: PMC11807959 DOI: 10.3389/fimmu.2024.1485009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025] Open
Abstract
Background Multisystem inflammatory syndrome in children (MIS-C) is a rare and severe post-COVID-19 complication with multiple phenotypes. Objectives The aim of this study is to study inflammatory biomarkers (cytokines and oxidative stress) in critical MIS-C patients and to observe if there is association between these biomarkers and mortality. Methods A single-center prospective study enrolled patients with MIS-C (with positive molecular test), aged between 1 month and 18 years of age. Data was collected from 20 pediatric intensive care unit (PICU)'s bed. Inflammatory biomarkers (cytokines and oxidative stress markers) were performed on day 1 and 3 after hospitalization. Survival rate was calculated, and Kaplan-Meier curves were plotted. Univariate and multivariate Cox regression analyses were conducted. The ROC (Receiver Operating Characteristic) curve analysis was performed. Results and conclusions A total of 41 patients out of 109 patients admitted at PICU with suspected MIS-C during the study period were included, of which 33 (80.5%) were male, 9 (22%) were under one year old, and 30 (73.2%) presented comorbidities. Among them, 16 (39%) did not survive. The mean survival time was shorter in patients with higher levels of IL-17A (≥ 19.71 pg/mL) on day 1 (115 vs 323 days, p = 0.004). Higher levels of IL-17A on day 1 were associated with mortality in both the crude model (HR 1.03, CI95% 1.004-1.057, p = 0.022) and the adjusted model (HR 1.043, CI95% 1.013-1.075, p = 0.012). ROC analysis revealed a cut-off value for the IL-17A of 14.32 pg/ml. The other immunological and inflammatory markers did not demonstrate an association with survival (p>0.05). Our findings suggest that patients with high levels of IL-17A are at greater risk for death.
Collapse
Affiliation(s)
- Emmerson C. F. de Farias
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Luciana M. P. P. do Nascimento
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Manoel J. C. Pavão Junior
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Dalila C. A. Pavão
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Ana P. S. Pinheiro
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Andreza H. O. Pinheiro
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Marília C. B. Alves
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Kíssila M. M. M. Ferraro
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Larisse F. Q. Aires
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Luana G. Dias
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Mayara M. M. Machado
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Michaelle J. D. Serrão
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Raphaella R. Gomes
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Sara M. P. de Moraes
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Gabriela C. L. Pontes
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Railana D. F. P. Carvalho
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Cristiane T. C. Silva
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Carla M. A. das Neves
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Joyce C. L. dos Santos
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Adriana M. B. de Sousa
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Leda L. da Silva
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Mary L. F. M. F. de Mello
- Division of Pediatric Intensive Care, Department of Pediatrics, Fundação Santa Casa de Misericórdia do Pará, Belém, Brazil
| | - Patricia B. Carvalho
- Division of Pediatric Intensive Care, Departament of Pediatrics, Fundação Hospital das Clínicas Gaspar Viana, Belém, Brazil
| | - Renata de B. Braga
- Division of Pediatric Intensive Care, Departament of Pediatrics, Fundação Hospital das Clínicas Gaspar Viana, Belém, Brazil
| | - Kathia de O. Harada
- Division of Pediatric Intensive Care, Departament of Pediatrics, Fundação Hospital das Clínicas Gaspar Viana, Belém, Brazil
| | - Maria C. A. Justino
- Clinical Research Unit, Health Surveillance Secretariat, Brazilian Ministry of Health, Instituto Evandro Chagas, Ananindeua, Brazil
| | - Iran B. Costa
- Immunology Laboratory, Virology Unit, Instituto Evandro Chagas, Ananindeua, Brazil
| | - Igor Brasil-Costa
- Immunology Laboratory, Virology Unit, Instituto Evandro Chagas, Ananindeua, Brazil
| | - Marta C. Monteiro
- Pharmaceutical Science Post-Graduation Program and Neuroscience and Cell Biology Graduate Program, Health Science Institute, Federal University of Pará/UFPA, Belém, Brazil
| | - Gleice Clemente
- Division of Pediatric Rheumatology, Department of Pediatrics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Teresa Terreri
- Division of Pediatric Rheumatology, Department of Pediatrics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Zhang YJ, Liu XY, Xu WX, Yang YP. Reevaluation of prognostic and severity indicators for COVID-19 patients in the emergency department. Ann Med 2024; 56:2417178. [PMID: 39435611 PMCID: PMC11497573 DOI: 10.1080/07853890.2024.2417178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 10/23/2024] Open
Abstract
AIMS This study aimed to re-evaluate whether the scoring systems, neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were effective in predicting prognosis and severity of COVID-19 patients in the emergency department (ED). METHODS COVID-19 patients enrolled in this retrospective study divided into the death (DEA) and survival (SUR) groups, the severe/critical (SC) and non-severe/critical (non-SC) groups. The Acute Physiology and Chronic Health Evaluation (APACHE) II, Sequential Organ Failure Assessment (SOFA), National Early Warning Score (NEWS) and CCEDRRN COVID-19 Mortality Score were calculated. The neutrophil, lymphocyte and platelet counts were extracted from the first routine blood examination, and NLR and PLR were calculated accordingly. Receiver Operating Characteristic (ROC) curve and logistic regression were performed. RESULTS All the scoring systems, as well as NLR and PLR, significantly increased in both the DEA and SC groups. The ROC curve showed that the CCEDRRN COVID-19 Mortality Score had the highest predictive value for mortality and severity (AUC 0.779, 0.850, respectively), which outperformed the APACHE II, SOFA and NEWS. NLR presented better predictive ability for severity (AUC 0.741) than death (AUC 0.702). The APACHE II, NEWS and CCEDRRN COVID-19 Mortality Score were positively correlated with both prognosis and severity, whereas NLR only with severity. CONCLUSION The NEWS and CCEDRRN COVID-19 Mortality Score were reconfirmed for early and rapid predicting the poor prognosis and severity of COVID-19 patients in ED, especially the CCEDRRN COVID-19 Mortality Score with the highest discrimination capacity, and NLR was more appropriate for predicting the severity.
Collapse
Affiliation(s)
- Yi-Jie Zhang
- Department of Emergency, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiao-Yan Liu
- Department of Infection, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wan-Xin Xu
- Department of Emergency, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ya-Peng Yang
- Department of General Surgery, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Jaiswal A, Shrivastav S, Kushwaha HR, Chaturvedi R, Singh RP. Oncogenic potential of SARS-CoV-2-targeting hallmarks of cancer pathways. Cell Commun Signal 2024; 22:447. [PMID: 39327555 PMCID: PMC11426004 DOI: 10.1186/s12964-024-01818-0] [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: 07/01/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The 2019 outbreak of SARS-CoV-2 has caused a major worldwide health crisis with high rates of morbidity and death. Interestingly, it has also been linked to cancer, which begs the issue of whether it plays a role in carcinogenesis. Recent studies have revealed various mechanisms by which SARS-CoV-2 can influence oncogenic pathways, potentially promoting cancer development. The virus encodes several proteins that alter key signaling pathways associated with cancer hallmarks. Unlike classical oncogenic viruses, which transform cells through viral oncogenes or by activating host oncogenes, SARS-CoV-2 appears to promote tumorigenesis by inhibiting tumor suppressor genes and pathways while activating survival, proliferation, and inflammation-associated signaling cascades. Bioinformatic analyses and experimental studies have identified numerous interactions between SARS-CoV-2 proteins and cellular components involved in cancer-related processes. This review explores the intricate relationship between SARS-CoV-2 infection and cancer, focusing on the regulation of key hallmarks driving initiation, promotion and progression of cancer by viral proteins. By elucidating the underlying mechanisms driving cellular transformation, the potential of SARS-CoV-2 as an oncovirus is highlighted. Comprehending these interplays is essential to enhance our understanding of COVID-19 and cancer biology and further formulating strategies to alleviate SARS-CoV-2 influence on cancer consequences.
Collapse
Affiliation(s)
- Aishwarya Jaiswal
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sanah Shrivastav
- SRM Institute of Science and Technology, Delhi-NCR Campus, Ghaziabad, Uttar Pradesh, India
| | - Hemant R Kushwaha
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
10
|
Krčmová LK, Javorská L, Matoušová K, Šmahel P, Skála M, Kopecký M, Suwanvecho C, Přívratská N, Turoňová D, Melichar B. Evaluation of inflammatory biomarkers and vitamins in hospitalized patients with SARS-CoV-2 infection and post-COVID syndrome. Clin Chem Lab Med 2024; 62:1217-1227. [PMID: 38374668 DOI: 10.1515/cclm-2023-1297] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
OBJECTIVES Concentrations of neopterin, kynurenine and kynurenine/tryptophan ratios predict prognosis and the need for oxygen therapy in patients hospitalized for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The aims of the present study were to evaluate the changes of these biomarkers early in the course of infection, the association with the prior coronavirus disease (COVID-19) vaccination and therapeutic administration of Anti-SARS-CoV-2 monoclonal antibodies, investigation of other potential biomarkers including neuropilin, 8-hydroxy-2-deoxyguanosine and 8-hydroxyguanosine in patients hospitalized with SARS-CoV-2 infection and an assessment of these biomarkers and vitamins A, E and D in patients with post-COVID syndrome. METHODS Urine and blood samples were obtained on the 1st to the 4th day and 4th to 7th day from 108 patients hospitalized with COVID-19. Chromatography tandem mass spectrometry methods were used to analyse neopterin, kynurenine, tryptophan, liposoluble vitamins, and DNA damage biomarkers. RESULTS A statistically significant decrease of neopterin, kynurenine and kynurenine/tryptophan ratios was observed on after 4th to 7th day of hospitalization, and concentrations of these biomarkers were increased in patients with poor prognosis and subsequent post-COVID syndrome. The concentrations of remaining biomarker and vitamins were not associated with outcomes, although markedly decreased concentrations of vitamin A, E and D were noted. CONCLUSIONS The concentrations of neopterin, kynurenine and kynurenine/tryptophan ratios decrease during the course of infection SARS-CoV-2 and are associated with the post-COVID syndrome. No other prognostic biomarkers were identified.
Collapse
Affiliation(s)
- Lenka Kujovská Krčmová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Lenka Javorská
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Kateřina Matoušová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Petr Šmahel
- Deparment of Infectious Diseases, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Mikuláš Skála
- Department of Pneumology, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Michal Kopecký
- Department of Pneumology, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Chaweewan Suwanvecho
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Nikola Přívratská
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Dorota Turoňová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| |
Collapse
|
11
|
Grand RJ. SARS-CoV-2 and the DNA damage response. J Gen Virol 2023; 104:001918. [PMID: 37948194 PMCID: PMC10768691 DOI: 10.1099/jgv.0.001918] [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: 09/01/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
The recent coronavirus disease 2019 (COVID-19) pandemic was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is characterized by respiratory distress, multiorgan dysfunction and, in some cases, death. The virus is also responsible for post-COVID-19 condition (commonly referred to as 'long COVID'). SARS-CoV-2 is a single-stranded, positive-sense RNA virus with a genome of approximately 30 kb, which encodes 26 proteins. It has been reported to affect multiple pathways in infected cells, resulting, in many cases, in the induction of a 'cytokine storm' and cellular senescence. Perhaps because it is an RNA virus, replicating largely in the cytoplasm, the effect of SARS-Cov-2 on genome stability and DNA damage responses (DDRs) has received relatively little attention. However, it is now becoming clear that the virus causes damage to cellular DNA, as shown by the presence of micronuclei, DNA repair foci and increased comet tails in infected cells. This review considers recent evidence indicating how SARS-CoV-2 causes genome instability, deregulates the cell cycle and targets specific components of DDR pathways. The significance of the virus's ability to cause cellular senescence is also considered, as are the implications of genome instability for patients suffering from long COVID.
Collapse
Affiliation(s)
- Roger J. Grand
- Institute for Cancer and Genomic Science, The Medical School, University of Birmingham, Birmingham, UK
| |
Collapse
|
12
|
Katarzyna Lesiów M, Witwicki M, Tan NK, Graziotto ME, New EJ. Unravelling the Mystery of COVID-19 Pathogenesis: Spike Protein and Cu Can Synergize to Trigger ROS Production. Chemistry 2023; 29:e202301530. [PMID: 37414735 DOI: 10.1002/chem.202301530] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/29/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023]
Abstract
The COVID-19 pandemic has had a devastating impact on global health, highlighting the need to understand how the SARS-CoV-2 virus damages the lungs in order to develop effective treatments. Recent research has shown that patients with COVID-19 experience severe oxidative damage to various biomolecules. We propose that the overproduction of reactive oxygen species (ROS) in SARS-CoV-2 infection involves an interaction between copper ions and the virus's spike protein. We tested two peptide fragments, Ac-ELDKYFKNH-NH2 (L1) and Ac-WSHPQFEK-NH2 (L2), derived from the spike protein of the Wuhan strain and the β variant, respectively, and found that they bind Cu(II) ions and form a three-nitrogen complexes at lung pH. Our research demonstrates that these complexes trigger the overproduction of ROS, which can break both DNA strands and transform DNA into its linear form. Using A549 cells, we demonstrated that ROS overproduction occurs in the mitochondria, not in the cytoplasm. Our findings highlight the importance of the interaction between copper ions and the virus's spike protein in the development of lung damage and may aid in the development of therapeutic procedures.
Collapse
Affiliation(s)
| | - Maciej Witwicki
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383, Wrocław, Poland
| | - Nian Kee Tan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for, Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Elizabeth Joy New
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for, Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
13
|
Bowen DR, Pathak S, Nadar RM, Parise RD, Ramesh S, Govindarajulu M, Moore A, Ren J, Moore T, Dhanasekaran M. Oxidative stress and COVID-19-associated neuronal dysfunction: mechanisms and therapeutic implications. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1153-1167. [PMID: 37357527 PMCID: PMC10465323 DOI: 10.3724/abbs.2023085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/09/2023] [Indexed: 06/27/2023] Open
Abstract
Severe acute respiratory syndrome (SARS)-CoV-2 virus causes novel coronavirus disease 2019 (COVID-19), and there is a possible role for oxidative stress in the pathophysiology of neurological diseases associated with COVID-19. Excessive oxidative stress could be responsible for the thrombosis and other neuronal dysfunctions observed in COVID-19. This review discusses the role of oxidative stress associated with SARS-CoV-2 and the mechanisms involved. Furthermore, the various therapeutics implicated in treating COVID-19 and the oxidative stress that contributes to the etiology and pathogenesis of COVID-19-induced neuronal dysfunction are discussed. Further mechanistic and clinical research to combat COVID-19 is warranted to understand the exact mechanisms, and its true clinical effects need to be investigated to minimize neurological complications from COVID-19.
Collapse
Affiliation(s)
- Dylan R. Bowen
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Suhrud Pathak
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Rishi M. Nadar
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Rachel D. Parise
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Sindhu Ramesh
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Manoj Govindarajulu
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Austin Moore
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Jun Ren
- Department of CardiologyZhongshan Hospital Fudan UniversityShanghai200032China
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWA98195USA
| | - Timothy Moore
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | | |
Collapse
|
14
|
Tepebaşı MY, İlhan İ, Temel EN, Sancer O, Öztürk Ö. Investigation of inflammation, oxidative stress, and DNA damage in COVID-19 patients. Cell Stress Chaperones 2023; 28:191-199. [PMID: 36797451 PMCID: PMC9936118 DOI: 10.1007/s12192-023-01330-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/11/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
COVID-19 disease, which spreads worldwide, is a disease characterized by widespread inflammation and affects many organs, especially the lungs. The resulting inflammation can lead to reactive oxygen radicals, leading to oxidative DNA damage. The pneumonia severity of 95 hospitalized patients with positive RT-PCR test was determined and divided into three groups: mild, moderate, and severe/critical. Inflammation markers (neutrophil-lymphocyte ratio, serum reactive protein, procalcitonin, etc.) were determined, and IL-10 and IFN-γ measurements were analyzed using the enzyme-linked immunosorbent assay method. In evaluating oxidative damage, total thiol, native thiol, disulfide, and ischemia-modified albumin (IMA) levels were determined by measuring spectrophotometrically. The comet assay method's percentage of tail DNA obtained was used to determine oxidative DNA damage. As a result, when the mild and severe/critical groups were compared, we found that total thiol, native thiol, and disulfide levels decreased significantly in the severe/critical group due to the increase in inflammation markers and cytokine levels (p < 0.05). We could not detect any significance in IMA levels between the groups (p > 0.05). At the same time, we determined an increase in the tail DNA percent level, that is, DNA damage, due to the increased oxidative effect. As a result, we determined that inflammation and oxidative stress increased in patients with severe pneumonia, and there was DNA damage in these patients.
Collapse
Affiliation(s)
| | - İlter İlhan
- Department of Biochemistry, University of Süleyman Demirel, Isparta, Turkey
| | - Esra Nurlu Temel
- Department of Infectious Diseases, University of Süleyman Demirel, Isparta, Turkey
| | - Okan Sancer
- Department of Medical Biology, University of Süleyman Demirel, Isparta, Turkey
| | - Önder Öztürk
- Department of Chest Diseases, University of Süleyman Demirel, Isparta, Turkey
| |
Collapse
|
15
|
Olsen MB, Huse C, de Sousa MML, Murphy SL, Sarno A, Obermann TS, Yang K, Holter JC, Jørgensen MJ, Christensen EE, Wang W, Ji P, Heggelund L, Hoel H, Dyrhol-Riise AM, Gregersen I, Aukrust P, Bjørås M, Halvorsen B, Dahl TB. DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients. J Inflamm Res 2022; 15:6629-6644. [PMID: 36514358 PMCID: PMC9741826 DOI: 10.2147/jir.s379331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/20/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose Reactive oxygen species (ROS) are an important part of the inflammatory response during infection but can also promote DNA damage. Due to the sustained inflammation in severe Covid-19, we hypothesized that hospitalized Covid-19 patients would be characterized by increased levels of oxidative DNA damage and dysregulation of the DNA repair machinery. Patients and Methods Levels of the oxidative DNA lesion 8-oxoG and levels of base excision repair (BER) proteins were measured in peripheral blood mononuclear cells (PBMC) from patients (8-oxoG, n = 22; BER, n = 17) and healthy controls (n = 10) (Cohort 1). Gene expression related to DNA repair was investigated in two independent cohorts of hospitalized Covid-19 patients (Cohort 1; 15 patents and 5 controls, Cohort 2; 15 patients and 6 controls), and by publicly available datasets. Results Patients and healthy controls showed comparable amounts of oxidative DNA damage as assessed by 8-oxoG while levels of several BER proteins were increased in Covid-19 patients, indicating enhanced DNA repair in acute Covid-19 disease. Furthermore, gene expression analysis demonstrated regulation of genes involved in BER and double strand break repair (DSBR) in PBMC of Covid-19 patients and expression level of several DSBR genes correlated with the degree of respiratory failure. Finally, by re-analyzing publicly available data, we found that the pathway Hallmark DNA repair was significantly more regulated in circulating immune cells during Covid-19 compared to influenza virus infection, bacterial pneumonia or acute respiratory infection due to seasonal coronavirus. Conclusion Although beneficial by protecting against DNA damage, long-term activation of the DNA repair machinery could also contribute to persistent inflammation, potentially through mechanisms such as the induction of cellular senescence. However, further studies that also include measurements of additional markers of DNA damage are required to determine the role and precise molecular mechanisms for DNA repair in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mirta Mittelstedt Leal de Sousa
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway,Proteomics and Modomics Experimental Core Facility (PROMEC), NTNU, Trondheim, Norway
| | - Sarah Louise Murphy
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Antonio Sarno
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway,Department of Fisheries and New Biomarine Industry, SINTEF Ocean, Trondheim, Norway
| | - Tobias Sebastian Obermann
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Microbiology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Marte Jøntvedt Jørgensen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Erik Egeland Christensen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Wei Wang
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ping Ji
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars Heggelund
- Department of Internal Medicine, Vestre Viken Hospital Trust, Drammen, Norway,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Hedda Hoel
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Department of Medicine, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Anne Margarita Dyrhol-Riise
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway,Department of Microbiology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Division of Critical Care and Emergencies, Oslo University Hospital, Oslo, Norway,Correspondence: Tuva Børresdatter Dahl, Division of Critical Care and Emergencies and Research Institute of Internal Medicine, Oslo University Hospital, Sognsvannsveien 20, Oslo, Norway, Tel +4723072786, Email
| |
Collapse
|
16
|
Mihaljevic O, Zivancevic-Simonovic S, Cupurdija V, Marinkovic M, Tubic Vukajlovic J, Markovic A, Stanojevic-Pirkovic M, Milosevic-Djordjevic O. DNA damage in peripheral blood lymphocytes of severely ill COVID-19 patients in relation to inflammatory markers and parameters of hemostasis. Mutagenesis 2022; 37:203-212. [PMID: 35524945 PMCID: PMC9129204 DOI: 10.1093/mutage/geac011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/11/2022] [Indexed: 01/08/2023] Open
Abstract
Bearing in the mind that a variety of agents can contribute to genome instability, including viral infections, the aim of this study was to analyze DNA damage in hospitalized COVID-19 patients and its relationship with certain laboratory parameters. The potential impact of applied therapy and chest X-rays on DNA damage was also estimated. The study population included 24 severely COVID-19 patients and 15 healthy control subjects. The level of DNA damage was measured as genetic damage index (GDI) by comet assay. The standard laboratory methods and certified enzymatic reagents for the appropriate autoanalyzers were performed for the determination of the biochemical and hematological parameters. COVID-19 patients had significantly higher level of DNA damage compared with control subjects. The absolute number of neutrophil leukocytes was statistically higher, while the absolute number of lymphocytes was statistically lower in COVID-19 patients than in healthy controls. The analysis of the relationship between DNA damage and laboratory parameters indicated that GDI was positively correlated with interleukin 6 (IL-6) concentration and negatively with platelet count in COVID-19 patients. The level of DNA damage was slightly higher in female patients, in whom it was demonstrated a positive correlation of GDI with C-reactive protein (CRP) and procalcitonin. Likewise, there was a negative relationship of GDI and platelet count, and positive relationship of GDI and activated partial thromboplastin time (aPTT) in female population. The applied therapy (antibiotics, corticosteroid, anticoagulant, and antiviral therapy) as well as chest X rays has been shown to have genotoxic potential. The level of DNA damage significantly corresponds to the inflammatory markers and parameters of hemostasis in COVID-19 patients. In conclusion, inflammation, smoking habit, applied therapy, and chest X rays contribute to a higher level of DNA damage in COVID-19 patients.
Collapse
Affiliation(s)
- Olgica Mihaljevic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Vojislav Cupurdija
- Department of Internal Medicine Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Internal medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Milos Marinkovic
- Department of Internal medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| | | | - Aleksandra Markovic
- Department of Biology, Faculty of Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Olivera Milosevic-Djordjevic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Biology, Faculty of Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
17
|
Lorkiewicz P, Waszkiewicz N. Is SARS-CoV-2 a Risk Factor of Bipolar Disorder?-A Narrative Review. J Clin Med 2022; 11:6060. [PMID: 36294388 PMCID: PMC9604904 DOI: 10.3390/jcm11206060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
For 2.5 years we have been facing the coronavirus disease (COVID-19) and its health, social and economic effects. One of its known consequences is the development of neuropsychiatric diseases such as anxiety and depression. However, reports of manic episodes related to COVID-19 have emerged. Mania is an integral part of the debilitating illness-bipolar disorder (BD). Due to its devastating effects, it is therefore important to establish whether SARS-CoV-2 infection is a causative agent of this severe mental disorder. In this narrative review, we discuss the similarities between the disorders caused by SARS-CoV-2 and those found in patients with BD, and we also try to answer the question of whether SARS-CoV-2 infection may be a risk factor for the development of this affective disorder. Our observation shows that disorders in COVID-19 showing the greatest similarity to those in BD are cytokine disorders, tryptophan metabolism, sleep disorders and structural changes in the central nervous system (CNS). These changes, especially intensified in severe infections, may be a trigger for the development of BD in particularly vulnerable people, e.g., with family history, or cause an acute episode in patients with a pre-existing BD.
Collapse
Affiliation(s)
- Piotr Lorkiewicz
- Department of Psychiatry, Medical University of Bialystok, Wołodyjowskiego 2, 15-272 Białystok, Poland
| | | |
Collapse
|
18
|
GÜLBAY G, SAVRUN A. COVID-19 vakalarında DNA hasarı ve enflamasyon. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Purpose: The aim of this study is to see oxidative DNA damage (8-OHdG), its relationship with inflammatory mediators (IL6 and TNFA), and its reflections on laboratory findings in patients who had COVID-19 infection at different intensities.
Materials and Methods: Serum interleukin-6 (IL6), tumor necrosis factor-alpha (TNFA), and 8-hydroxy-2′-deoxyguanosine (8-OHdG) levels were measured using kits based on the enzyme-linked immunosorbent assay (ELISA) principle.
Results: In COVID-19 positive patients treated in intensive care 8-OHdG marker level is at the highest level and statistically significant. In patients receiving inpatient treatment in the hospitalized, the 8-OHdG marker level is higher than the control and outpatient groups. IL6 values were at the highest level in the patient group treated in the intensive care unit and were higher than the outpatient and control groups. There was no statistically significant difference between the control and patient groups in terms of TNFA values. Neutrophil-to-lymphocyte ratio (NLR) was lower in the control group than in all patient groups. C-reactive protein (CRP) is higher in hospitalized patients than in the control group. Lactate dehydrogenase (LDH) was found to be statistically significantly higher in hospitalized patients than outpatients.
Conclusion: As the severity of COVID-19 increases, serum 8-OHdG and IL6 levels also increase. These parameters can guide the diagnosis of COVID-19 patients in the early stages of the disease course.
Collapse
|
19
|
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
|
20
|
Li A, Yang J, Qian J, Shao X, Liao J, Lu X, Fan X. Tracing the cell-type-specific modules of immune responses during COVID-19 progression using scDisProcema. Comput Struct Biotechnol J 2022; 20:3545-3555. [PMID: 35811838 PMCID: PMC9250167 DOI: 10.1016/j.csbj.2022.06.066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 12/16/2022] Open
Abstract
COVID-19 has caused severe threats to lives and damage to property worldwide. The immunopathology of the disease is of particular concern. Currently, researchers have used gene co-expression networks (GCNs) to deepen the study of molecular mechanisms of immune responses to COVID-19. However, most efforts have not fully explored dynamic changes of cell-type-specific molecular networks in the disease process. This study proposes a GCN construction pipeline named single-cell Disease Progression cellular module analysis (scDisProcema), which can trace dynamic changes of immune system response during disease progression using single-cell data. Here, scDisProcema considers changes in cell fate and expression patterns during disease development, identifying gene modules responsible for different immune cells. The hub genes are screened for each module by the specific expression level and the intercellular connectivity of modules. Based on functional items enriched by each gene module, we elucidate the biological processes of different cells involved in disease development and explain the molecular mechanisms underlying the process of cell depletion or proliferation caused by disease. Compared with traditional WGCNA methods, scDisProcema can make more convenient use of the heterogeneity information provided by scRNA-seq data and has great potential in exploring molecular changes during disease progression and organ development.
Collapse
Affiliation(s)
- Anyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Jihong Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou 310058, China
- Zhang Boli Intelligent Health Innovation Lab, Hangzhou 311100, China
| | - Jingyang Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Jie Liao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Xiaoyan Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou 310058, China
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou 310058, China
- Corresponding author at: College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
21
|
Yang L, Xiong H, Li X, Li Y, Zhou H, Lin X, Chan TF, Li R, Lai KP, Chen X. Network Pharmacology and Comparative Transcriptome Reveals Biotargets and Mechanisms of Curcumol Treating Lung Adenocarcinoma Patients With COVID-19. Front Nutr 2022; 9:870370. [PMID: 35520289 PMCID: PMC9063984 DOI: 10.3389/fnut.2022.870370] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/16/2022] [Indexed: 12/31/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has led to 4,255,892 deaths worldwide. Although COVID-19 vaccines are available, mutant forms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have reduced the effectiveness of vaccines. Patients with cancer are more vulnerable to COVID-19 than patients without cancer. Identification of new drugs to treat COVID-19 could reduce mortality rate, and traditional Chinese Medicine(TCM) has shown potential in COVID-19 treatment. In this study, we focused on lung adenocarcinoma (LUAD) patients with COVID-19. We aimed to investigate the use of curcumol, a TCM, to treat LUAD patients with COVID-19, using network pharmacology and systematic bioinformatics analysis. The results showed that LUAD and patients with COVID-19 share a cluster of common deregulated targets. The network pharmacology analysis identified seven core targets (namely, AURKA, CDK1, CCNB1, CCNB2, CCNE1, CCNE2, and TTK) of curcumol in patients with COVID-19 and LUAD. Clinicopathological analysis of these targets demonstrated that the expression of these targets is associated with poor patient survival rates. The bioinformatics analysis further highlighted the involvement of this target cluster in DNA damage response, chromosome stability, and pathogenesis of LUAD. More importantly, these targets influence cell-signaling associated with the Warburg effect, which supports SARS-CoV-2 replication and inflammatory response. Comparative transcriptomic analysis on in vitro LUAD cell further validated the effect of curcumol for treating LUAD through the control of cell cycle and DNA damage response. This study supports the earlier findings that curcumol is a potential treatment for patients with LUAD and COVID-19.
Collapse
Affiliation(s)
- Lu Yang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Hao Xiong
- Guilin Center for Disease Control and Prevention, Guilin, China
| | - Xin Li
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Yu Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Huanhuan Zhou
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Xiao Lin
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ting Fung Chan
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Rong Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- *Correspondence: Rong Li
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- Keng Po Lai
| | - Xu Chen
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Xu Chen ;
| |
Collapse
|
22
|
Analysis of 19 urinary biomarkers of oxidative stress, nitrative stress, metabolic disorders, and inflammation using liquid chromatography–tandem mass spectrometry. Anal Bioanal Chem 2022; 414:2103-2116. [PMID: 35013809 PMCID: PMC8747998 DOI: 10.1007/s00216-021-03844-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/23/2021] [Accepted: 12/09/2021] [Indexed: 01/23/2023]
Abstract
Environmental chemical exposures have been associated with cancer, diabetes, hormonal and immunological disorders, and cardiovascular diseases. Some direct effects of chemical exposure that are precursors to adverse health outcomes, including oxidative stress, nitrative stress, hormonal imbalance, neutrophilia, and eosinophilia, can be assessed through the analysis of biomarkers in urine. In this study, we describe a novel methodology for the determination of 19 biomarkers of health effects: malondialdehyde (MDA), 8-isoprostaglandin-F2α (8-PGF2α), 11-β-prostaglandin-F2α (11-PGF2α), 15-prostaglandin-F2α (15-PGF2α), 8-iso-15-prostaglandin-F2α (8,15-PGF2α), 8-hydroxy-2′-deoxyguanosine (8-OHdG), 8-hydroxyguanosine (8-HdG), 8-hydroxyguanine (8-HG), dityrosine (diY), allantoin (Alla), and two metabolic products of 4-hydroxynonenal (HNE), namely 4-hydroxy-2-nonenal glutathione (HNE-GSH) and 4-hydroxy-2-nonenal mercapturic acid (HNE-MA) (in total, 12 oxidative stress biomarkers, OSBs); 8-nitroguanosine (8-NdG), 8-nitroguanine (8-NG), and 3-nitrotyrosine (NY) (3 nitrative stress biomarkers, NSBs); chlorotyrosine (CY) and bromotyrosine (BY) (2 inflammatory biomarkers); and the advanced glycation end-products (AGEs) Nε-carboxymethyllysine (CML) and Nε-carboxyethyllysine (CEL) (2 metabolic disorder biomarkers). Since these biomarkers are trigged by a variety of environmental insults and produced by different biomolecular pathways, their selective and sensitive determination in urine would help broadly elucidate the pathogenesis of diseases mediated by environmental factors.
Collapse
|
23
|
The potential role of COVID-19 in the induction of DNA damage. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 789:108411. [PMID: 35690420 PMCID: PMC8767986 DOI: 10.1016/j.mrrev.2022.108411] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/30/2021] [Accepted: 01/17/2022] [Indexed: 01/07/2023]
Abstract
The coronavirus disease-2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is challenging global health and economic systems. In some individuals, COVID-19 can cause a wide array of symptoms, affecting several organs, such as the lungs, heart, bowels, kidneys and brain, causing multiorgan failure, sepsis and death. These effects are related in part to direct viral infection of these organs, immunological deregulation, a hypercoagulatory state and the potential for development of cytokine storm syndrome. Since the appearance of COVID-19 is recent, the long-term effects on the health of recovered patients remain unknown. In this review, we focused on current evidence of the mechanisms of DNA damage mediated by coronaviruses. Data supports that these viruses can induce DNA damage, genomic instability, and cell cycle deregulation during their replication in mammalian cells. Since the induction of DNA damage and aberrant DNA repair mechanisms are related to the development of chronic diseases such as cancer, diabetes, neurodegenerative disorders, and atherosclerosis, it will be important to address similar effects and outcomes in recovered COVID-19 patients.
Collapse
|