Inhibition of DDR1 potentiates carbon ion radiotherapy by promoting ferroptosis and immunogenic death in head and neck squamous cell carcinoma.

IF 7.5 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
Wei Hu, Qingting Huang, Li Chen, Shikai Geng, Haojiong Zhang, Huaiyuan Chen, Xingyu Liu, Jingqin Zhong, Fengtao Su, Chunlin Shao, Lin Kong
{"title":"Inhibition of DDR1 potentiates carbon ion radiotherapy by promoting ferroptosis and immunogenic death in head and neck squamous cell carcinoma.","authors":"Wei Hu, Qingting Huang, Li Chen, Shikai Geng, Haojiong Zhang, Huaiyuan Chen, Xingyu Liu, Jingqin Zhong, Fengtao Su, Chunlin Shao, Lin Kong","doi":"10.1186/s12967-025-07062-5","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Carbon ion radiotherapy (CIR) has emerged as a promising therapeutic modality for photon-resistant malignancies due to its unique physical depth-dose distribution and enhanced radiobiological effectiveness. Nevertheless, treatment resistance persists in certain recurrent or refractory head and neck squamous cell carcinoma (HNSCC) cases, underscoring the need for novel combinatorial strategies. Here, we demonstrated the sensitizing effect of targeting discoidin domain receptor 1 (DDR1) in HNSCC for CIR.</p><p><strong>Methods: </strong>MOC1 and and Cal27 cell lines along with tumor-bearing C57BL/6 mice were used for in vitro and in vivo studies. DDR1 was knocked down via lentivirus. Cell viability and proliferation were assessed by CCK-8 and colony formation assays. Immunogenicity and tumor-infiltrating lymphocytes were measured via flow cytometry and immunofluorescence. Tumor suppression mechanisms were investigated using RNA sequencing and bioinformatics. Ferroptosis markers (lipid peroxidation, iron, ROS) were detected using MDA, BODIPY 581/591 C11, FerroOrange, and DCFH-DA probes. Upstream ferroptosis mechanisms were analyzed by Western blot, co-immunoprecipitation, key molecule modulator administration, and SCD1 overexpression.</p><p><strong>Results: </strong>We demonstrated that targeting DDR1 potentiated CIR by triggering ferroptosis-mediated immunogenic cell death, which in turn enhanced antitumor immunity. Mechanistically, DDR1 sustained tumor cell survival by forming 14-3-3-mediated assembly of a DDR1/14-3-3/Akt ternary complex, thereby activating the Akt/mTORC1/SREBP1/SCD1 axis to promote monounsaturated fatty acid (MUFA) biosynthesis and suppress ferroptosis. Silencing DDR1 disrupted this complex, alleviating MUFA-mediated ferroptosis inhibition and subsequently increasing tumor immunogenicity. This immunogenic shift facilitated CD8 + T cell infiltration and cytotoxicity, amplifying CIR-induced tumor suppression. Furthermore, pharmacological inhibition of DDR1 using the small-molecule inhibitor 7rh recapitulated these effects, demonstrating potent anti-proliferative and ferroptosis-inducing capabilities, enhancing CIR sensitivity to better control tumor progression.</p><p><strong>Conclusions: </strong>Our findings positioned DDR1 targeting as a therapeutic strategy to potentiate CIR through immunogenic ferroptosis induction in HNSCC.</p>","PeriodicalId":17458,"journal":{"name":"Journal of Translational Medicine","volume":"23 1","pages":"1011"},"PeriodicalIF":7.5000,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12461992/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Translational Medicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12967-025-07062-5","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Carbon ion radiotherapy (CIR) has emerged as a promising therapeutic modality for photon-resistant malignancies due to its unique physical depth-dose distribution and enhanced radiobiological effectiveness. Nevertheless, treatment resistance persists in certain recurrent or refractory head and neck squamous cell carcinoma (HNSCC) cases, underscoring the need for novel combinatorial strategies. Here, we demonstrated the sensitizing effect of targeting discoidin domain receptor 1 (DDR1) in HNSCC for CIR.

Methods: MOC1 and and Cal27 cell lines along with tumor-bearing C57BL/6 mice were used for in vitro and in vivo studies. DDR1 was knocked down via lentivirus. Cell viability and proliferation were assessed by CCK-8 and colony formation assays. Immunogenicity and tumor-infiltrating lymphocytes were measured via flow cytometry and immunofluorescence. Tumor suppression mechanisms were investigated using RNA sequencing and bioinformatics. Ferroptosis markers (lipid peroxidation, iron, ROS) were detected using MDA, BODIPY 581/591 C11, FerroOrange, and DCFH-DA probes. Upstream ferroptosis mechanisms were analyzed by Western blot, co-immunoprecipitation, key molecule modulator administration, and SCD1 overexpression.

Results: We demonstrated that targeting DDR1 potentiated CIR by triggering ferroptosis-mediated immunogenic cell death, which in turn enhanced antitumor immunity. Mechanistically, DDR1 sustained tumor cell survival by forming 14-3-3-mediated assembly of a DDR1/14-3-3/Akt ternary complex, thereby activating the Akt/mTORC1/SREBP1/SCD1 axis to promote monounsaturated fatty acid (MUFA) biosynthesis and suppress ferroptosis. Silencing DDR1 disrupted this complex, alleviating MUFA-mediated ferroptosis inhibition and subsequently increasing tumor immunogenicity. This immunogenic shift facilitated CD8 + T cell infiltration and cytotoxicity, amplifying CIR-induced tumor suppression. Furthermore, pharmacological inhibition of DDR1 using the small-molecule inhibitor 7rh recapitulated these effects, demonstrating potent anti-proliferative and ferroptosis-inducing capabilities, enhancing CIR sensitivity to better control tumor progression.

Conclusions: Our findings positioned DDR1 targeting as a therapeutic strategy to potentiate CIR through immunogenic ferroptosis induction in HNSCC.

抑制DDR1通过促进头颈部鳞状细胞癌的铁下垂和免疫原性死亡来增强碳离子放疗。
背景:碳离子放射治疗(CIR)由于其独特的物理深度剂量分布和增强的放射生物学有效性,已成为一种有前途的治疗光子抗性恶性肿瘤的方式。然而,在某些复发或难治性头颈部鳞状细胞癌(HNSCC)病例中,治疗耐药性仍然存在,这强调了需要新的联合治疗策略。本研究以盘状蛋白结构域受体1 (disidin domain receptor 1, DDR1)为靶点,在HNSCC中对cirr有增敏作用。方法:以MOC1和Cal27细胞系以及携带肿瘤的C57BL/6小鼠为实验对象,进行体外和体内研究。DDR1通过慢病毒被敲除。采用CCK-8法和菌落形成法测定细胞活力和增殖能力。采用流式细胞术和免疫荧光法检测免疫原性和肿瘤浸润淋巴细胞。利用RNA测序和生物信息学研究肿瘤抑制机制。采用MDA、BODIPY 581/591 C11、FerroOrange和DCFH-DA探针检测铁下垂标志物(脂质过氧化、铁、ROS)。通过Western blot、共免疫沉淀、关键分子调节剂给药和SCD1过表达分析上游铁死亡机制。结果:我们证明了靶向DDR1通过触发铁中毒介导的免疫原性细胞死亡来增强CIR,从而增强抗肿瘤免疫。机制上,DDR1通过形成14-3-3介导的DDR1/14-3-3/Akt三元复合物组装,从而激活Akt/mTORC1/SREBP1/SCD1轴,促进单不饱和脂肪酸(MUFA)的生物合成,抑制铁凋亡,从而维持肿瘤细胞存活。沉默DDR1破坏了这个复合体,减轻了mufa介导的铁下沉抑制,随后增加了肿瘤的免疫原性。这种免疫原性转变促进了CD8 + T细胞浸润和细胞毒性,放大了cirr诱导的肿瘤抑制。此外,使用小分子抑制剂7rh对DDR1的药理学抑制再现了这些作用,显示出有效的抗增殖和诱导铁凋亡的能力,增强了CIR的敏感性,从而更好地控制肿瘤进展。结论:我们的研究结果将DDR1靶向定位为通过免疫原性铁下垂诱导HNSCC增强CIR的治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Journal of Translational Medicine
Journal of Translational Medicine 医学-医学:研究与实验
CiteScore
10.00
自引率
1.40%
发文量
537
审稿时长
1 months
期刊介绍: The Journal of Translational Medicine is an open-access journal that publishes articles focusing on information derived from human experimentation to enhance communication between basic and clinical science. It covers all areas of translational medicine.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信