Cargando…

DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients

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 oxidativ...

Descripción completa

Detalles Bibliográficos
Autores principales: Olsen, Maria Belland, Huse, Camilla, de Sousa, Mirta Mittelstedt Leal, Murphy, Sarah Louise, Sarno, Antonio, Obermann, Tobias Sebastian, Yang, Kuan, Holter, Jan Cato, Jørgensen, Marte Jøntvedt, Christensen, Erik Egeland, Wang, Wei, Ji, Ping, Heggelund, Lars, Hoel, Hedda, Dyrhol-Riise, Anne Margarita, Gregersen, Ida, Aukrust, Pål, Bjørås, Magnar, Halvorsen, Bente, Dahl, Tuva Børresdatter
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9741826/
https://www.ncbi.nlm.nih.gov/pubmed/36514358
http://dx.doi.org/10.2147/JIR.S379331
_version_ 1784848399290335232
author Olsen, Maria Belland
Huse, Camilla
de Sousa, Mirta Mittelstedt Leal
Murphy, Sarah Louise
Sarno, Antonio
Obermann, Tobias Sebastian
Yang, Kuan
Holter, Jan Cato
Jørgensen, Marte Jøntvedt
Christensen, Erik Egeland
Wang, Wei
Ji, Ping
Heggelund, Lars
Hoel, Hedda
Dyrhol-Riise, Anne Margarita
Gregersen, Ida
Aukrust, Pål
Bjørås, Magnar
Halvorsen, Bente
Dahl, Tuva Børresdatter
author_facet Olsen, Maria Belland
Huse, Camilla
de Sousa, Mirta Mittelstedt Leal
Murphy, Sarah Louise
Sarno, Antonio
Obermann, Tobias Sebastian
Yang, Kuan
Holter, Jan Cato
Jørgensen, Marte Jøntvedt
Christensen, Erik Egeland
Wang, Wei
Ji, Ping
Heggelund, Lars
Hoel, Hedda
Dyrhol-Riise, Anne Margarita
Gregersen, Ida
Aukrust, Pål
Bjørås, Magnar
Halvorsen, Bente
Dahl, Tuva Børresdatter
author_sort Olsen, Maria Belland
collection PubMed
description 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.
format Online
Article
Text
id pubmed-9741826
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-97418262022-12-12 DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients Olsen, Maria Belland Huse, Camilla de Sousa, Mirta Mittelstedt Leal Murphy, Sarah Louise Sarno, Antonio Obermann, Tobias Sebastian Yang, Kuan Holter, Jan Cato Jørgensen, Marte Jøntvedt Christensen, Erik Egeland Wang, Wei Ji, Ping Heggelund, Lars Hoel, Hedda Dyrhol-Riise, Anne Margarita Gregersen, Ida Aukrust, Pål Bjørås, Magnar Halvorsen, Bente Dahl, Tuva Børresdatter J Inflamm Res Original Research 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. Dove 2022-12-07 /pmc/articles/PMC9741826/ /pubmed/36514358 http://dx.doi.org/10.2147/JIR.S379331 Text en © 2022 Olsen et al. https://creativecommons.org/licenses/by-nc/3.0/This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/ (https://creativecommons.org/licenses/by-nc/3.0/) ). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
spellingShingle Original Research
Olsen, Maria Belland
Huse, Camilla
de Sousa, Mirta Mittelstedt Leal
Murphy, Sarah Louise
Sarno, Antonio
Obermann, Tobias Sebastian
Yang, Kuan
Holter, Jan Cato
Jørgensen, Marte Jøntvedt
Christensen, Erik Egeland
Wang, Wei
Ji, Ping
Heggelund, Lars
Hoel, Hedda
Dyrhol-Riise, Anne Margarita
Gregersen, Ida
Aukrust, Pål
Bjørås, Magnar
Halvorsen, Bente
Dahl, Tuva Børresdatter
DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients
title DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients
title_full DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients
title_fullStr DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients
title_full_unstemmed DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients
title_short DNA Repair Mechanisms are Activated in Circulating Lymphocytes of Hospitalized Covid-19 Patients
title_sort dna repair mechanisms are activated in circulating lymphocytes of hospitalized covid-19 patients
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9741826/
https://www.ncbi.nlm.nih.gov/pubmed/36514358
http://dx.doi.org/10.2147/JIR.S379331
work_keys_str_mv AT olsenmariabelland dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT husecamilla dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT desousamirtamittelstedtleal dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT murphysarahlouise dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT sarnoantonio dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT obermanntobiassebastian dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT yangkuan dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT holterjancato dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT jørgensenmartejøntvedt dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT christensenerikegeland dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT wangwei dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT jiping dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT heggelundlars dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT hoelhedda dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT dyrholriiseannemargarita dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT gregersenida dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT aukrustpal dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT bjørasmagnar dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT halvorsenbente dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients
AT dahltuvabørresdatter dnarepairmechanismsareactivatedincirculatinglymphocytesofhospitalizedcovid19patients