Glioblastoma-instructed microglia transition to heterogeneous phenotypic states with phagocytic and dendritic cell-like features in patient tumors and patient-derived orthotopic xenografts

IF 10.4 1区 生物学 Q1 GENETICS & HEREDITY
Yahaya A. Yabo, Pilar M. Moreno-Sanchez, Yolanda Pires-Afonso, Tony Kaoma, Bakhtiyor Nosirov, Andrea Scafidi, Luca Ermini, Anuja Lipsa, Anaïs Oudin, Dimitrios Kyriakis, Kamil Grzyb, Suresh K. Poovathingal, Aurélie Poli, Arnaud Muller, Reka Toth, Barbara Klink, Guy Berchem, Christophe Berthold, Frank Hertel, Michel Mittelbronn, Dieter H. Heiland, Alexander Skupin, Petr V. Nazarov, Simone P. Niclou, Alessandro Michelucci, Anna Golebiewska
{"title":"Glioblastoma-instructed microglia transition to heterogeneous phenotypic states with phagocytic and dendritic cell-like features in patient tumors and patient-derived orthotopic xenografts","authors":"Yahaya A. Yabo, Pilar M. Moreno-Sanchez, Yolanda Pires-Afonso, Tony Kaoma, Bakhtiyor Nosirov, Andrea Scafidi, Luca Ermini, Anuja Lipsa, Anaïs Oudin, Dimitrios Kyriakis, Kamil Grzyb, Suresh K. Poovathingal, Aurélie Poli, Arnaud Muller, Reka Toth, Barbara Klink, Guy Berchem, Christophe Berthold, Frank Hertel, Michel Mittelbronn, Dieter H. Heiland, Alexander Skupin, Petr V. Nazarov, Simone P. Niclou, Alessandro Michelucci, Anna Golebiewska","doi":"10.1186/s13073-024-01321-8","DOIUrl":null,"url":null,"abstract":"A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry, and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood–brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment. ","PeriodicalId":12645,"journal":{"name":"Genome Medicine","volume":null,"pages":null},"PeriodicalIF":10.4000,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Genome Medicine","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s13073-024-01321-8","RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

Abstract

A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry, and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood–brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.
胶质母细胞瘤诱导的小胶质细胞向具有吞噬细胞和树突状细胞特征的异质表型状态转变,这种表型状态出现在患者肿瘤和患者来源的正位异种移植物中
导致胶质母细胞瘤(GBM)发生和发展的一个主要因素是它能够通过创造免疫抑制环境来逃避免疫系统,在这种环境中,GBM 相关的髓样细胞(包括常驻小胶质细胞和外周单核细胞衍生巨噬细胞)发挥着关键的促肿瘤作用。然而,目前还不清楚 GBM 患者招募的髓样细胞在表型和功能上是否相同,也不清楚这种异质性是否会在患者来源的正位异种移植物(PDOXs)中重现。目前对 GBM 生态系统及其在临床前模型中的再现还缺乏透彻的了解,导致结果不准确和临床试验失败。在此,我们报告了 GBM PDOXs 和患者肿瘤中肿瘤微环境(TME)在单细胞和空间水平上的系统表征。我们应用单细胞 RNA 测序、空间转录组学、多色流式细胞术、免疫组化和功能研究来检查 GBM 细胞指示的异质性 TME。我们将代表不同肿瘤表型的 GBM PDOXs 与胶质瘤小鼠 GL261 合成模型和患者肿瘤进行了比较。我们发现,GBM 肿瘤细胞与宿主细胞相互作用,在 PDOXs 中形成了 GBM 患者特异性 TME。我们在髓系细胞中检测到了最突出的转录组适应性,脑内驻留的小胶质细胞代表了细胞肿瘤中的主要群体,而外周来源的髓系细胞则在血脑屏障破坏的部位渗入大脑。更具体地说,我们发现 GBM 教育的小胶质细胞在不同的 GBM 地貌和肿瘤壁龛中过渡到不同的表型状态。GBM教育的小胶质细胞亚群显示出类似吞噬细胞和树突状细胞的基因表达程序。此外,我们还发现了表达细胞周期程序、星形胶质细胞或内皮细胞标记的新型小胶质细胞状态。最后,我们发现替莫唑胺治疗会导致转录组的可塑性,并改变 GBM 肿瘤细胞与邻近 TME 成分之间的串扰。我们的数据为 GBM 肿瘤指示的异质性 TME 的表型适应提供了新的见解。我们显示了小胶质细胞表型状态在支持 GBM 肿瘤生长和治疗反应中的关键作用。我们的数据将 PDOXs 作为评估 TME 功能性和治疗后 GBM 生态系统变化的相关模型。
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
Genome Medicine
Genome Medicine GENETICS & HEREDITY-
CiteScore
20.80
自引率
0.80%
发文量
128
审稿时长
6-12 weeks
期刊介绍: Genome Medicine is an open access journal that publishes outstanding research applying genetics, genomics, and multi-omics to understand, diagnose, and treat disease. Bridging basic science and clinical research, it covers areas such as cancer genomics, immuno-oncology, immunogenomics, infectious disease, microbiome, neurogenomics, systems medicine, clinical genomics, gene therapies, precision medicine, and clinical trials. The journal publishes original research, methods, software, and reviews to serve authors and promote broad interest and importance in the field.
×
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:481959085
Book学术官方微信