Cargando…

Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production

Low-dose radiation therapy (LDRT) can suppress intractable inflammation, such as that in rheumatoid arthritis, and is used for treating more than 10,000 rheumatoid arthritis patients annually in Europe. Several recent clinical trials have reported that LDRT can effectively reduce the severity of cor...

Descripción completa

Detalles Bibliográficos
Autores principales: Song, Ha-Yeon, Chen, Fengjia, Park, Hae Ran, Han, Jeong Moo, Ji, Hyun Jung, Byun, Eui-Baek, Kwon, Yeongkag, Kim, Min-Kyu, Ahn, Ki Bum, Seo, Ho Seong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10248130/
https://www.ncbi.nlm.nih.gov/pubmed/37304302
http://dx.doi.org/10.3389/fimmu.2023.1182927
_version_ 1785055306033659904
author Song, Ha-Yeon
Chen, Fengjia
Park, Hae Ran
Han, Jeong Moo
Ji, Hyun Jung
Byun, Eui-Baek
Kwon, Yeongkag
Kim, Min-Kyu
Ahn, Ki Bum
Seo, Ho Seong
author_facet Song, Ha-Yeon
Chen, Fengjia
Park, Hae Ran
Han, Jeong Moo
Ji, Hyun Jung
Byun, Eui-Baek
Kwon, Yeongkag
Kim, Min-Kyu
Ahn, Ki Bum
Seo, Ho Seong
author_sort Song, Ha-Yeon
collection PubMed
description Low-dose radiation therapy (LDRT) can suppress intractable inflammation, such as that in rheumatoid arthritis, and is used for treating more than 10,000 rheumatoid arthritis patients annually in Europe. Several recent clinical trials have reported that LDRT can effectively reduce the severity of coronavirus disease (COVID-19) and other cases of viral pneumonia. However, the therapeutic mechanism of LDRT remains unelucidated. Therefore, in the current study, we aimed to investigate the molecular mechanism underlying immunological alterations in influenza pneumonia after LDRT. Mice were irradiated to the whole lung 1 day post-infection. The changes in levels of inflammatory mediators (cytokines and chemokines) and immune cell populations in the bronchoalveolar lavage (BALF), lungs, and serum were examined. LDRT-treated mice displayed markedly increased survival rates and reduced lung edema and airway and vascular inflammation in the lung; however, the viral titers in the lungs were unaffected. Levels of primary inflammatory cytokines were reduced after LDRT, and transforming growth factor-β (TGF-β) levels increased significantly on day 1 following LDRT. Levels of chemokines increased from day 3 following LDRT. Additionally, M2 macrophage polarization or recruitment was increased following LDRT. We found that LDRT-induced TGF-β reduced the levels of cytokines and polarized M2 cells and blocked immune cell infiltration, including neutrophils, in BALF. LDRT-induced early TGF-β production was shown to be a key regulator involved in broad-spectrum anti-inflammatory activity in virus-infected lungs. Therefore, LDRT or TGF-β may be an alternative therapy for viral pneumonia.
format Online
Article
Text
id pubmed-10248130
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-102481302023-06-09 Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production Song, Ha-Yeon Chen, Fengjia Park, Hae Ran Han, Jeong Moo Ji, Hyun Jung Byun, Eui-Baek Kwon, Yeongkag Kim, Min-Kyu Ahn, Ki Bum Seo, Ho Seong Front Immunol Immunology Low-dose radiation therapy (LDRT) can suppress intractable inflammation, such as that in rheumatoid arthritis, and is used for treating more than 10,000 rheumatoid arthritis patients annually in Europe. Several recent clinical trials have reported that LDRT can effectively reduce the severity of coronavirus disease (COVID-19) and other cases of viral pneumonia. However, the therapeutic mechanism of LDRT remains unelucidated. Therefore, in the current study, we aimed to investigate the molecular mechanism underlying immunological alterations in influenza pneumonia after LDRT. Mice were irradiated to the whole lung 1 day post-infection. The changes in levels of inflammatory mediators (cytokines and chemokines) and immune cell populations in the bronchoalveolar lavage (BALF), lungs, and serum were examined. LDRT-treated mice displayed markedly increased survival rates and reduced lung edema and airway and vascular inflammation in the lung; however, the viral titers in the lungs were unaffected. Levels of primary inflammatory cytokines were reduced after LDRT, and transforming growth factor-β (TGF-β) levels increased significantly on day 1 following LDRT. Levels of chemokines increased from day 3 following LDRT. Additionally, M2 macrophage polarization or recruitment was increased following LDRT. We found that LDRT-induced TGF-β reduced the levels of cytokines and polarized M2 cells and blocked immune cell infiltration, including neutrophils, in BALF. LDRT-induced early TGF-β production was shown to be a key regulator involved in broad-spectrum anti-inflammatory activity in virus-infected lungs. Therefore, LDRT or TGF-β may be an alternative therapy for viral pneumonia. Frontiers Media S.A. 2023-05-25 /pmc/articles/PMC10248130/ /pubmed/37304302 http://dx.doi.org/10.3389/fimmu.2023.1182927 Text en Copyright © 2023 Song, Chen, Park, Han, Ji, Byun, Kwon, Kim, Ahn and Seo https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Song, Ha-Yeon
Chen, Fengjia
Park, Hae Ran
Han, Jeong Moo
Ji, Hyun Jung
Byun, Eui-Baek
Kwon, Yeongkag
Kim, Min-Kyu
Ahn, Ki Bum
Seo, Ho Seong
Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
title Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
title_full Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
title_fullStr Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
title_full_unstemmed Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
title_short Low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
title_sort low-dose radiation therapy suppresses viral pneumonia by enhancing broad-spectrum anti-inflammatory responses via transforming growth factor-β production
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10248130/
https://www.ncbi.nlm.nih.gov/pubmed/37304302
http://dx.doi.org/10.3389/fimmu.2023.1182927
work_keys_str_mv AT songhayeon lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT chenfengjia lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT parkhaeran lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT hanjeongmoo lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT jihyunjung lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT byuneuibaek lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT kwonyeongkag lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT kimminkyu lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT ahnkibum lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction
AT seohoseong lowdoseradiationtherapysuppressesviralpneumoniabyenhancingbroadspectrumantiinflammatoryresponsesviatransforminggrowthfactorbproduction