Targeting SPHK1 in macrophages remodels the tumor microenvironment and enhances anti-PD-1 immunotherapy efficacy in colorectal cancer liver metastasis.

IF 20.1 1区 医学 Q1 ONCOLOGY
Yizhi Zhan, Jinsong Xu, Zhanqiao Zhang, Yating Hu, Yongsheng Li, Junying Qian, Yunyan Ling, Dehua Wu, Haijun Deng, Guoxin Li, Zhiyong Shen, Yuan Fang
{"title":"Targeting SPHK1 in macrophages remodels the tumor microenvironment and enhances anti-PD-1 immunotherapy efficacy in colorectal cancer liver metastasis.","authors":"Yizhi Zhan, Jinsong Xu, Zhanqiao Zhang, Yating Hu, Yongsheng Li, Junying Qian, Yunyan Ling, Dehua Wu, Haijun Deng, Guoxin Li, Zhiyong Shen, Yuan Fang","doi":"10.1002/cac2.70047","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer liver metastasis (CRLM) is characterized by an immunosuppressive microenvironment and a blunted response to immunotherapy. Notably, tumor-associated macrophages (TAMs) play a critical role in modulating immune responses and exhibit significant heterogeneity in CRLM. Sphingosine kinase 1 (SPHK1) serves as a pivotal kinase in maintaining the balance between ceramide and sphingosine-1-phosphate (S1P) levels. However, the effects of SPHK1 within TAMs on tumor immune evasion during CRLM remain elusive. This study aimed at investigating the role of TAM-intrinsic SPHK1 in tumor immunosuppressive microenvironment in CRLM.</p><p><strong>Methods: </strong>SPHK1 expression levels in TAMs were estimated by immunofluorescence and bioinformatics analysis. Several animal models were established to elucidate the role of SPHK1 in tumor immunity reprogramming in vivo. Flow cytometry, cytokine assay, and transwell assay were conducted to investigate the effects of SPHK1 in TAMs in cell-cell communication in vitro. RNA-sequencing, Western blotting, and quantitative real-time polymerase chain reaction were used to explore the molecular mechanism by which SPHK1 activated NLR family pyrin domain containing 3 (NLRP3) inflammasome in TAMs.</p><p><strong>Results: </strong>We found that SPHK1 was mainly expressed in TAMs and identified SPHK1<sup>+</sup> TAMs as associated with CRLM and diminished efficacy of immunotherapy in human patients. These SPHK1<sup>+</sup> TAMs exhibited strong immunosuppressive activities by inducing CD8<sup>+</sup> T cell exhaustion with high programmed cell death 1 (PD-1) expression via the interaction between TAMs and CRC cells. Mechanistically, SPHK1-produced S1P exerted an autocrine effect to activate NLRP3 inflammasome and interleukin 1 beta (IL-1β) release via nuclear factor-kappa B (NF-κB) and hypoxia inducible factor 1 subunit alpha (HIF-1α) signaling in TAMs. Paracrine IL-1β then upregulated the expression of monocyte chemoattractants and ADAM metallopeptidase domain 17 (ADAM17) sheddase in CRC cells, resulting in TAM infiltration and CD8<sup>+</sup> T cell dysfunction in the liver microenvironment. Furthermore, combining SPHK1-targeting treatments with anti-PD-1 therapy or radioimmunotherapy largely stalled liver metastasis and caused a significant extension of lifespan in preclinical mouse models.</p><p><strong>Conclusions: </strong>Our findings highlighted the role of SPHK1 of TAMs in facilitating CRLM by promoting CD8<sup>+</sup> T cell dysfunction and immunosuppressive microenvironment. Combining SPHK1 blockade with anti-PD-1 therapy may be a promising treatment regimen for patients with CRLM.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":" ","pages":""},"PeriodicalIF":20.1000,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1002/cac2.70047","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Colorectal cancer liver metastasis (CRLM) is characterized by an immunosuppressive microenvironment and a blunted response to immunotherapy. Notably, tumor-associated macrophages (TAMs) play a critical role in modulating immune responses and exhibit significant heterogeneity in CRLM. Sphingosine kinase 1 (SPHK1) serves as a pivotal kinase in maintaining the balance between ceramide and sphingosine-1-phosphate (S1P) levels. However, the effects of SPHK1 within TAMs on tumor immune evasion during CRLM remain elusive. This study aimed at investigating the role of TAM-intrinsic SPHK1 in tumor immunosuppressive microenvironment in CRLM.

Methods: SPHK1 expression levels in TAMs were estimated by immunofluorescence and bioinformatics analysis. Several animal models were established to elucidate the role of SPHK1 in tumor immunity reprogramming in vivo. Flow cytometry, cytokine assay, and transwell assay were conducted to investigate the effects of SPHK1 in TAMs in cell-cell communication in vitro. RNA-sequencing, Western blotting, and quantitative real-time polymerase chain reaction were used to explore the molecular mechanism by which SPHK1 activated NLR family pyrin domain containing 3 (NLRP3) inflammasome in TAMs.

Results: We found that SPHK1 was mainly expressed in TAMs and identified SPHK1+ TAMs as associated with CRLM and diminished efficacy of immunotherapy in human patients. These SPHK1+ TAMs exhibited strong immunosuppressive activities by inducing CD8+ T cell exhaustion with high programmed cell death 1 (PD-1) expression via the interaction between TAMs and CRC cells. Mechanistically, SPHK1-produced S1P exerted an autocrine effect to activate NLRP3 inflammasome and interleukin 1 beta (IL-1β) release via nuclear factor-kappa B (NF-κB) and hypoxia inducible factor 1 subunit alpha (HIF-1α) signaling in TAMs. Paracrine IL-1β then upregulated the expression of monocyte chemoattractants and ADAM metallopeptidase domain 17 (ADAM17) sheddase in CRC cells, resulting in TAM infiltration and CD8+ T cell dysfunction in the liver microenvironment. Furthermore, combining SPHK1-targeting treatments with anti-PD-1 therapy or radioimmunotherapy largely stalled liver metastasis and caused a significant extension of lifespan in preclinical mouse models.

Conclusions: Our findings highlighted the role of SPHK1 of TAMs in facilitating CRLM by promoting CD8+ T cell dysfunction and immunosuppressive microenvironment. Combining SPHK1 blockade with anti-PD-1 therapy may be a promising treatment regimen for patients with CRLM.

巨噬细胞靶向SPHK1可重塑肿瘤微环境,增强抗pd -1免疫治疗结直肠癌肝转移的疗效。
背景:结直肠癌肝转移(CRLM)的特点是免疫抑制微环境和对免疫治疗的迟钝反应。值得注意的是,肿瘤相关巨噬细胞(tam)在调节免疫应答中发挥关键作用,并在CRLM中表现出显著的异质性。鞘氨醇激酶1 (SPHK1)是维持神经酰胺和鞘氨醇-1-磷酸(S1P)水平平衡的关键激酶。然而,tam中SPHK1在CRLM期间对肿瘤免疫逃避的影响尚不清楚。本研究旨在探讨TAM-intrinsic SPHK1在CRLM肿瘤免疫抑制微环境中的作用。方法:采用免疫荧光和生物信息学分析方法检测SPHK1在tam中的表达水平。为了阐明SPHK1在体内肿瘤免疫重编程中的作用,我们建立了几种动物模型。通过流式细胞术、细胞因子实验和transwell实验研究SPHK1在tam细胞-细胞通讯中的作用。采用rna测序、Western blotting、实时定量聚合酶链反应等方法探讨SPHK1激活tam中NLR家族pyrin domain containing 3 (NLRP3)炎性体的分子机制。结果:我们发现SPHK1主要在tam中表达,并发现SPHK1+ tam与人类患者的CRLM和免疫治疗效果降低有关。这些SPHK1+ tam通过与CRC细胞的相互作用诱导CD8+ T细胞衰竭并高表达程序性细胞死亡1 (PD-1),表现出很强的免疫抑制活性。机制上,sphk1产生的S1P发挥自分泌作用,通过核因子κB (NF-κB)和缺氧诱导因子1亚单位α (HIF-1α)信号通路激活NLRP3炎性体和白细胞介素1β (IL-1β)释放。分泌旁分泌IL-1β上调CRC细胞单核细胞趋化剂和ADAM金属肽酶结构域17 (ADAM17)脱落酶的表达,导致肝微环境中TAM浸润和CD8+ T细胞功能障碍。此外,在临床前小鼠模型中,sphk1靶向治疗与抗pd -1治疗或放射免疫治疗相结合,在很大程度上阻止了肝转移,并显著延长了寿命。结论:我们的研究结果强调了tam的SPHK1通过促进CD8+ T细胞功能障碍和免疫抑制微环境来促进CRLM的作用。联合SPHK1阻断与抗pd -1治疗可能是一种很有希望的治疗方案。
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
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
×
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