Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages

Radiotherapy for head-and-neck cancers frequently causes long-term hypofunction of salivary glands that severely compromises quality of life and is difficult to treat. Here, we studied effects and mechanisms of Sphingosine-1-phosphate (S1P), a versatile signaling sphingolipid, in preventing irrevers...

Full description

Bibliographic Details
Main Authors: Tao Yang, Qingguo Zhao, Meijun Hu, Simin Pan, Linying Zhang, Ruoxi Zhu, Bowen Zhou, Xuanhe Feng, Zhenhua Gao, Zhao Zhu, Yu Zhang, Liang Hu, Fei Liu, Zhaochen Shan
Format: Article
Language:English
Published: MDPI AG 2022-10-01
Series:Antioxidants
Subjects:
Online Access:https://www.mdpi.com/2076-3921/11/10/2050
_version_ 1797475733902196736
author Tao Yang
Qingguo Zhao
Meijun Hu
Simin Pan
Linying Zhang
Ruoxi Zhu
Bowen Zhou
Xuanhe Feng
Zhenhua Gao
Zhao Zhu
Yu Zhang
Liang Hu
Fei Liu
Zhaochen Shan
author_facet Tao Yang
Qingguo Zhao
Meijun Hu
Simin Pan
Linying Zhang
Ruoxi Zhu
Bowen Zhou
Xuanhe Feng
Zhenhua Gao
Zhao Zhu
Yu Zhang
Liang Hu
Fei Liu
Zhaochen Shan
author_sort Tao Yang
collection DOAJ
description Radiotherapy for head-and-neck cancers frequently causes long-term hypofunction of salivary glands that severely compromises quality of life and is difficult to treat. Here, we studied effects and mechanisms of Sphingosine-1-phosphate (S1P), a versatile signaling sphingolipid, in preventing irreversible dry mouth caused by radiotherapy. Mouse submandibular glands (SMGs) were irradiated with or without intra-SMG S1P pretreatment. The saliva flow rate was measured following pilocarpine stimulation. The expression of genes related to S1P signaling and radiation damage was examined by flow cytometry, immunohistochemistry, quantitative RT-PCR, Western blotting, and/or single-cell RNA-sequencing. S1P pretreatment ameliorated irradiation-induced salivary dysfunction in mice through a decrease in irradiation-induced oxidative stress and consequent apoptosis and cellular senescence, which is related to the enhancement of Nrf2-regulated anti-oxidative response. In mouse SMGs, endothelial cells and resident macrophages are the major cells capable of producing S1P and expressing the pro-regenerative S1P receptor S1pr1. Both mouse SMGs and human endothelial cells are protected from irradiation damage by S1P pretreatment, likely through the S1pr1/Akt/eNOS axis. Moreover, intra-SMG-injected S1P did not affect the growth and radiosensitivity of head-and-neck cancer in a mouse model. These data indicate that S1P signaling pathway is a promising target for alleviating irradiation-induced salivary gland hypofunction.
first_indexed 2024-03-09T20:49:03Z
format Article
id doaj.art-ebc40163985843c995ef43d5ac66f84c
institution Directory Open Access Journal
issn 2076-3921
language English
last_indexed 2024-03-09T20:49:03Z
publishDate 2022-10-01
publisher MDPI AG
record_format Article
series Antioxidants
spelling doaj.art-ebc40163985843c995ef43d5ac66f84c2023-11-23T22:39:57ZengMDPI AGAntioxidants2076-39212022-10-011110205010.3390/antiox11102050Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident MacrophagesTao Yang0Qingguo Zhao1Meijun Hu2Simin Pan3Linying Zhang4Ruoxi Zhu5Bowen Zhou6Xuanhe Feng7Zhenhua Gao8Zhao Zhu9Yu Zhang10Liang Hu11Fei Liu12Zhaochen Shan13Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaDepartment of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USAOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaDepartment of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USADepartment of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USAOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaDepartment of Statistics, Texas A&M University, College Station, TX 77843, USAOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaDepartment of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USAOutpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing 100050, ChinaRadiotherapy for head-and-neck cancers frequently causes long-term hypofunction of salivary glands that severely compromises quality of life and is difficult to treat. Here, we studied effects and mechanisms of Sphingosine-1-phosphate (S1P), a versatile signaling sphingolipid, in preventing irreversible dry mouth caused by radiotherapy. Mouse submandibular glands (SMGs) were irradiated with or without intra-SMG S1P pretreatment. The saliva flow rate was measured following pilocarpine stimulation. The expression of genes related to S1P signaling and radiation damage was examined by flow cytometry, immunohistochemistry, quantitative RT-PCR, Western blotting, and/or single-cell RNA-sequencing. S1P pretreatment ameliorated irradiation-induced salivary dysfunction in mice through a decrease in irradiation-induced oxidative stress and consequent apoptosis and cellular senescence, which is related to the enhancement of Nrf2-regulated anti-oxidative response. In mouse SMGs, endothelial cells and resident macrophages are the major cells capable of producing S1P and expressing the pro-regenerative S1P receptor S1pr1. Both mouse SMGs and human endothelial cells are protected from irradiation damage by S1P pretreatment, likely through the S1pr1/Akt/eNOS axis. Moreover, intra-SMG-injected S1P did not affect the growth and radiosensitivity of head-and-neck cancer in a mouse model. These data indicate that S1P signaling pathway is a promising target for alleviating irradiation-induced salivary gland hypofunction.https://www.mdpi.com/2076-3921/11/10/2050sphingosine-1-phosphate signalingsalivary glandsirradiationhead and neck cancersendothelial cellsresident macrophages
spellingShingle Tao Yang
Qingguo Zhao
Meijun Hu
Simin Pan
Linying Zhang
Ruoxi Zhu
Bowen Zhou
Xuanhe Feng
Zhenhua Gao
Zhao Zhu
Yu Zhang
Liang Hu
Fei Liu
Zhaochen Shan
Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages
Antioxidants
sphingosine-1-phosphate signaling
salivary glands
irradiation
head and neck cancers
endothelial cells
resident macrophages
title Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages
title_full Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages
title_fullStr Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages
title_full_unstemmed Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages
title_short Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages
title_sort sphingosine 1 phosphate alleviates irradiation induced salivary gland hypofunction through preserving endothelial cells and resident macrophages
topic sphingosine-1-phosphate signaling
salivary glands
irradiation
head and neck cancers
endothelial cells
resident macrophages
url https://www.mdpi.com/2076-3921/11/10/2050
work_keys_str_mv AT taoyang sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT qingguozhao sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT meijunhu sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT siminpan sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT linyingzhang sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT ruoxizhu sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT bowenzhou sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT xuanhefeng sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT zhenhuagao sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT zhaozhu sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT yuzhang sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT lianghu sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT feiliu sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages
AT zhaochenshan sphingosine1phosphatealleviatesirradiationinducedsalivaryglandhypofunctionthroughpreservingendothelialcellsandresidentmacrophages