SPP1+ macrophages polarized by lactate confer the progression of hypoxic adaptive tumor cells in brain.

IF 13.4 1区 医学 Q1 CLINICAL NEUROLOGY
Jianlei Zhang, Zhihui Li, Jiang Yin, Weina Fan, Hongfan Liao, Jing Dong, Xianfeng Yu, Yabing Cao, Qiong Zhang, Guopei Zheng, Qianming Yao
{"title":"SPP1+ macrophages polarized by lactate confer the progression of hypoxic adaptive tumor cells in brain.","authors":"Jianlei Zhang, Zhihui Li, Jiang Yin, Weina Fan, Hongfan Liao, Jing Dong, Xianfeng Yu, Yabing Cao, Qiong Zhang, Guopei Zheng, Qianming Yao","doi":"10.1093/neuonc/noaf208","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Brain malignancies originating from the central nervous system and metastasizing from extracerebral tumors remain incurable, while the underlying mechanisms remain unclear. In this study, we comprehensively investigated the pan-brain tumor microenvironment.</p><p><strong>Methods: </strong>We employed transgenic mice, stereotactic brain injections, flow cytometry, CRISPR/Cas9 gene editing, immunohistochemistry, immunofluorescence, quantitative reverse transcription-polymerase chain reaction, western blotting, co-immunoprecipitation, DNA pulldown assays, and chromatin immunoprecipitation.</p><p><strong>Results: </strong>We constructed single-cell RNA sequencing and spatial transcriptome profiles of pan-brain tumors and identified the enhanced hypoxia-inducible factor 1 (HIF-1) signaling in the intracerebral metastases compared with extracerebral parts, as well as in mesenchymal-subtype glioblastomas. Hypoxic adaptability mediated by HIF-1 signaling confers a tumor growth advantage in the brain. Integrated analysis and experimental models revealed the co-localization and mutual dependence between brain tumor hypoxic adaptability and macrophage infiltration. Hypoxic adaptive tumor cells recruit macrophages via galectin 1 (LGALS1) and induce differentiation toward the secreted phosphoprotein 1 (SPP1) + subpopulation via lactate mediated histone lactylation. SPP1 directly activates mitogen-activated protein kinase (MAPK) signaling in tumor cells to promote tumor growth and inhibits the cytotoxic activity of CD8+ T cells. Genetic SPP1 deficiency in macrophages delays hypoxic adaptive tumor growth in the brain and enhances the tumor response to anti-programmed cell death-1 (anti-PD-1) therapy. Preclinically, targeting lactate dehydrogenase A (LDHA) by stiripentol with blood-brain barrier permeability impedes brain tumor progression and synergizes with anti-PD-1 therapy.</p><p><strong>Conclusions: </strong>The interrelationship between hypoxic adaptive tumor cells and macrophages in the brain highlights the possibility of SPP1+ macrophage-based microenvironment remodeling in brain tumor therapy.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4000,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Neuro-oncology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1093/neuonc/noaf208","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CLINICAL NEUROLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Brain malignancies originating from the central nervous system and metastasizing from extracerebral tumors remain incurable, while the underlying mechanisms remain unclear. In this study, we comprehensively investigated the pan-brain tumor microenvironment.

Methods: We employed transgenic mice, stereotactic brain injections, flow cytometry, CRISPR/Cas9 gene editing, immunohistochemistry, immunofluorescence, quantitative reverse transcription-polymerase chain reaction, western blotting, co-immunoprecipitation, DNA pulldown assays, and chromatin immunoprecipitation.

Results: We constructed single-cell RNA sequencing and spatial transcriptome profiles of pan-brain tumors and identified the enhanced hypoxia-inducible factor 1 (HIF-1) signaling in the intracerebral metastases compared with extracerebral parts, as well as in mesenchymal-subtype glioblastomas. Hypoxic adaptability mediated by HIF-1 signaling confers a tumor growth advantage in the brain. Integrated analysis and experimental models revealed the co-localization and mutual dependence between brain tumor hypoxic adaptability and macrophage infiltration. Hypoxic adaptive tumor cells recruit macrophages via galectin 1 (LGALS1) and induce differentiation toward the secreted phosphoprotein 1 (SPP1) + subpopulation via lactate mediated histone lactylation. SPP1 directly activates mitogen-activated protein kinase (MAPK) signaling in tumor cells to promote tumor growth and inhibits the cytotoxic activity of CD8+ T cells. Genetic SPP1 deficiency in macrophages delays hypoxic adaptive tumor growth in the brain and enhances the tumor response to anti-programmed cell death-1 (anti-PD-1) therapy. Preclinically, targeting lactate dehydrogenase A (LDHA) by stiripentol with blood-brain barrier permeability impedes brain tumor progression and synergizes with anti-PD-1 therapy.

Conclusions: The interrelationship between hypoxic adaptive tumor cells and macrophages in the brain highlights the possibility of SPP1+ macrophage-based microenvironment remodeling in brain tumor therapy.

SPP1+巨噬细胞被乳酸极化与脑缺氧适应性肿瘤细胞的进展有关。
背景:起源于中枢神经系统并转移到脑外肿瘤的脑恶性肿瘤是无法治愈的,其潜在机制尚不清楚。在本研究中,我们全面研究了泛脑肿瘤微环境。方法:采用转基因小鼠、立体定向脑注射、流式细胞术、CRISPR/Cas9基因编辑、免疫组织化学、免疫荧光、定量逆转录聚合酶链反应、western blotting、共免疫沉淀、DNA pull - down测定、染色质免疫沉淀等方法。结果:我们构建了泛脑肿瘤的单细胞RNA测序和空间转录组图谱,并在脑内转移瘤和间充质亚型胶质母细胞瘤中发现了与脑外转移瘤相比,低氧诱导因子1 (HIF-1)信号的增强。HIF-1信号介导的缺氧适应性赋予肿瘤在大脑中的生长优势。综合分析和实验模型揭示了脑肿瘤缺氧适应性与巨噬细胞浸润的共定位和相互依赖关系。低氧适应性肿瘤细胞通过凝集素1 (LGALS1)招募巨噬细胞,并通过乳酸介导的组蛋白乳酸化诱导向分泌磷酸化蛋白1 (SPP1) +亚群分化。SPP1直接激活肿瘤细胞中的丝裂原活化蛋白激酶(MAPK)信号,促进肿瘤生长,抑制CD8+ T细胞的细胞毒活性。巨噬细胞中SPP1基因的缺失延迟了大脑中低氧适应性肿瘤的生长,并增强了肿瘤对抗程序性细胞死亡-1 (anti-程序性细胞死亡-1)治疗的反应。临床前研究表明,施替利妥醇具有血脑屏障通透性,靶向乳酸脱氢酶A (LDHA)可阻碍脑肿瘤进展,并与抗pd -1治疗协同作用。结论:脑内低氧适应性肿瘤细胞与巨噬细胞之间的相互关系提示了SPP1+巨噬细胞为基础的微环境重塑在脑肿瘤治疗中的可能性。
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
Neuro-oncology
Neuro-oncology 医学-临床神经学
CiteScore
27.20
自引率
6.30%
发文量
1434
审稿时长
3-8 weeks
期刊介绍: Neuro-Oncology, the official journal of the Society for Neuro-Oncology, has been published monthly since January 2010. Affiliated with the Japan Society for Neuro-Oncology and the European Association of Neuro-Oncology, it is a global leader in the field. The journal is committed to swiftly disseminating high-quality information across all areas of neuro-oncology. It features peer-reviewed articles, reviews, symposia on various topics, abstracts from annual meetings, and updates from neuro-oncology societies worldwide.
×
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