Cargando…

Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration

The production of nitric oxide (NO) is a key feature of immunosuppressive myeloid cells, which impair T cell activation and proliferation via reversibly blocking interleukin-2 receptor signaling. NO is mainly produced from L-arginine by inducible NO synthase (iNOS). Moreover, L-arginine is an essent...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Jieyu, Luo, Yuan, Yuan, Cheng, Han, Linzhi, Wu, Qiuji, Xu, Liexi, Gao, Yuke, Sun, Yingming, Ma, Shijing, Tang, Guiliang, Li, Shuying, Sun, Wenjie, Gong, Yan, Xie, Conghua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7097922/
https://www.ncbi.nlm.nih.gov/pubmed/32226302
http://dx.doi.org/10.7150/ijbs.41653
_version_ 1783511078904266752
author Xu, Jieyu
Luo, Yuan
Yuan, Cheng
Han, Linzhi
Wu, Qiuji
Xu, Liexi
Gao, Yuke
Sun, Yingming
Ma, Shijing
Tang, Guiliang
Li, Shuying
Sun, Wenjie
Gong, Yan
Xie, Conghua
author_facet Xu, Jieyu
Luo, Yuan
Yuan, Cheng
Han, Linzhi
Wu, Qiuji
Xu, Liexi
Gao, Yuke
Sun, Yingming
Ma, Shijing
Tang, Guiliang
Li, Shuying
Sun, Wenjie
Gong, Yan
Xie, Conghua
author_sort Xu, Jieyu
collection PubMed
description The production of nitric oxide (NO) is a key feature of immunosuppressive myeloid cells, which impair T cell activation and proliferation via reversibly blocking interleukin-2 receptor signaling. NO is mainly produced from L-arginine by inducible NO synthase (iNOS). Moreover, L-arginine is an essential element for T cell proliferation and behaviors. Impaired T cell function further inhibits anti-tumor immunity and promotes tumor progression. Previous studies indicated that radiotherapy activated anti-tumor immune responses in multiple tumors. However, myeloid-derived cells in the tumor microenvironment may neutralize these responses. We hypothesized that iNOS, as an important regulator of the immunosuppressive effects in myeloid-derived cells, mediated radiation resistance of cancer cells. In this study, we used 1400W dihydrochloride, a potent small-molecule inhibitor of iNOS, to explore the regulatory roles of NO in anti-tumor immunity. Radiotherapy and iNOS inhibition by 1400W collaboratively suppressed tumor growth and increased survival time, as well as increased tumor-infiltrating CD8(+) T cells and specific inflammatory cytokine levels, in both lung and breast cancer cells in vivo. Our results also suggested that myeloid cell-mediated inhibition of T cell proliferation was effectively counteracted by radiation and 1400W-mediated NO blockade in vitro. Thus, these results demonstrated that iNOS was an important regulator of radiotherapy-induced antitumor immune responses. The combination of radiotherapy with iNOS blockade might be an effective therapy to improve the response of tumors to clinical radiation.
format Online
Article
Text
id pubmed-7097922
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-70979222020-03-28 Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration Xu, Jieyu Luo, Yuan Yuan, Cheng Han, Linzhi Wu, Qiuji Xu, Liexi Gao, Yuke Sun, Yingming Ma, Shijing Tang, Guiliang Li, Shuying Sun, Wenjie Gong, Yan Xie, Conghua Int J Biol Sci Research Paper The production of nitric oxide (NO) is a key feature of immunosuppressive myeloid cells, which impair T cell activation and proliferation via reversibly blocking interleukin-2 receptor signaling. NO is mainly produced from L-arginine by inducible NO synthase (iNOS). Moreover, L-arginine is an essential element for T cell proliferation and behaviors. Impaired T cell function further inhibits anti-tumor immunity and promotes tumor progression. Previous studies indicated that radiotherapy activated anti-tumor immune responses in multiple tumors. However, myeloid-derived cells in the tumor microenvironment may neutralize these responses. We hypothesized that iNOS, as an important regulator of the immunosuppressive effects in myeloid-derived cells, mediated radiation resistance of cancer cells. In this study, we used 1400W dihydrochloride, a potent small-molecule inhibitor of iNOS, to explore the regulatory roles of NO in anti-tumor immunity. Radiotherapy and iNOS inhibition by 1400W collaboratively suppressed tumor growth and increased survival time, as well as increased tumor-infiltrating CD8(+) T cells and specific inflammatory cytokine levels, in both lung and breast cancer cells in vivo. Our results also suggested that myeloid cell-mediated inhibition of T cell proliferation was effectively counteracted by radiation and 1400W-mediated NO blockade in vitro. Thus, these results demonstrated that iNOS was an important regulator of radiotherapy-induced antitumor immune responses. The combination of radiotherapy with iNOS blockade might be an effective therapy to improve the response of tumors to clinical radiation. Ivyspring International Publisher 2020-03-05 /pmc/articles/PMC7097922/ /pubmed/32226302 http://dx.doi.org/10.7150/ijbs.41653 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Xu, Jieyu
Luo, Yuan
Yuan, Cheng
Han, Linzhi
Wu, Qiuji
Xu, Liexi
Gao, Yuke
Sun, Yingming
Ma, Shijing
Tang, Guiliang
Li, Shuying
Sun, Wenjie
Gong, Yan
Xie, Conghua
Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration
title Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration
title_full Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration
title_fullStr Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration
title_full_unstemmed Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration
title_short Downregulation of Nitric Oxide Collaborated with Radiotherapy to Promote Anti-Tumor Immune Response via Inducing CD8+ T Cell Infiltration
title_sort downregulation of nitric oxide collaborated with radiotherapy to promote anti-tumor immune response via inducing cd8+ t cell infiltration
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7097922/
https://www.ncbi.nlm.nih.gov/pubmed/32226302
http://dx.doi.org/10.7150/ijbs.41653
work_keys_str_mv AT xujieyu downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT luoyuan downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT yuancheng downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT hanlinzhi downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT wuqiuji downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT xuliexi downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT gaoyuke downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT sunyingming downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT mashijing downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT tangguiliang downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT lishuying downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT sunwenjie downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT gongyan downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration
AT xieconghua downregulationofnitricoxidecollaboratedwithradiotherapytopromoteantitumorimmuneresponseviainducingcd8tcellinfiltration