CPT1C-positive cancer-associated fibroblast facilitates immunosuppression through promoting IL-6-induced M2-like phenotype of macrophage.

IF 6.5 2区 医学 Q1 IMMUNOLOGY
Oncoimmunology Pub Date : 2024-05-10 eCollection Date: 2024-01-01 DOI:10.1080/2162402X.2024.2352179
Rongyuan Wei, Junquan Song, Hongda Pan, Xiaowen Liu, Jianpeng Gao
{"title":"CPT1C-positive cancer-associated fibroblast facilitates immunosuppression through promoting IL-6-induced M2-like phenotype of macrophage.","authors":"Rongyuan Wei, Junquan Song, Hongda Pan, Xiaowen Liu, Jianpeng Gao","doi":"10.1080/2162402X.2024.2352179","DOIUrl":null,"url":null,"abstract":"<p><p>Cancer-associated fibroblasts (CAFs) exhibit remarkable phenotypic heterogeneity, with specific subsets implicated in immunosuppression in various malignancies. However, whether and how they attenuate anti-tumor immunity in gastric cancer (GC) remains elusive. CPT1C, a unique isoform of carnitine palmitoyltransferase pivotal in regulating fatty acid oxidation, is briefly indicated as a protumoral metabolic mediator in the tumor microenvironment (TME) of GC. In the present study, we initially identified specific subsets of fibroblasts exclusively overexpressing CPT1C, hereby termed them as CPT1C<sup>+</sup>CAFs. Subsequent findings indicated that CPT1C<sup>+</sup>CAFs fostered a stroma-enriched and immunosuppressive TME as they correlated with extracellular matrix-related molecular features and enrichment of both immunosuppressive subsets, especially M2-like macrophages, and multiple immune-related pathways. Next, we identified that CPT1C<sup>+</sup>CAFs promoted the M2-like phenotype of macrophage <i>in vitro</i>. Bioinformatic analyses unveiled the robust IL-6 signaling between CPT1C<sup>+</sup>CAFs and M2-like phenotype of macrophage and identified CPT1C<sup>+</sup>CAFs as the primary source of IL-6. Meanwhile, suppressing CPT1C expression in CAFs significantly decreased IL-6 secretion <i>in vitro</i>. Lastly, we demonstrated the association of CPT1C<sup>+</sup>CAFs with therapeutic resistance. Notably, GC patients with high CPT1C<sup>+</sup>CAFs infiltration responded poorly to immunotherapy in clinical cohort. Collectively, our data not only present the novel identification of CPT1C<sup>+</sup>CAFs as immunosuppressive subsets in TME of GC, but also reveal the underlying mechanism that CPT1C<sup>+</sup>CAFs impair tumor immunity by secreting IL-6 to induce the immunosuppressive M2-like phenotype of macrophage in GC.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5000,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11093039/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Oncoimmunology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1080/2162402X.2024.2352179","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Cancer-associated fibroblasts (CAFs) exhibit remarkable phenotypic heterogeneity, with specific subsets implicated in immunosuppression in various malignancies. However, whether and how they attenuate anti-tumor immunity in gastric cancer (GC) remains elusive. CPT1C, a unique isoform of carnitine palmitoyltransferase pivotal in regulating fatty acid oxidation, is briefly indicated as a protumoral metabolic mediator in the tumor microenvironment (TME) of GC. In the present study, we initially identified specific subsets of fibroblasts exclusively overexpressing CPT1C, hereby termed them as CPT1C+CAFs. Subsequent findings indicated that CPT1C+CAFs fostered a stroma-enriched and immunosuppressive TME as they correlated with extracellular matrix-related molecular features and enrichment of both immunosuppressive subsets, especially M2-like macrophages, and multiple immune-related pathways. Next, we identified that CPT1C+CAFs promoted the M2-like phenotype of macrophage in vitro. Bioinformatic analyses unveiled the robust IL-6 signaling between CPT1C+CAFs and M2-like phenotype of macrophage and identified CPT1C+CAFs as the primary source of IL-6. Meanwhile, suppressing CPT1C expression in CAFs significantly decreased IL-6 secretion in vitro. Lastly, we demonstrated the association of CPT1C+CAFs with therapeutic resistance. Notably, GC patients with high CPT1C+CAFs infiltration responded poorly to immunotherapy in clinical cohort. Collectively, our data not only present the novel identification of CPT1C+CAFs as immunosuppressive subsets in TME of GC, but also reveal the underlying mechanism that CPT1C+CAFs impair tumor immunity by secreting IL-6 to induce the immunosuppressive M2-like phenotype of macrophage in GC.

CPT1C阳性的癌相关成纤维细胞通过促进IL-6诱导的巨噬细胞M2样表型来促进免疫抑制。
癌症相关成纤维细胞(CAFs)表现出显著的表型异质性,其特定亚群与各种恶性肿瘤的免疫抑制有关。然而,它们是否以及如何削弱胃癌(GC)的抗肿瘤免疫力仍是一个未知数。CPT1C是肉碱棕榈酰基转移酶的一种独特异构体,在调节脂肪酸氧化中起着关键作用。在本研究中,我们初步发现了专门过表达 CPT1C 的特定成纤维细胞亚群,并将其称为 CPT1C+CAF。随后的研究结果表明,CPT1C+CAFs 与细胞外基质相关的分子特征以及免疫抑制亚群(尤其是 M2 样巨噬细胞)和多种免疫相关通路的富集相关,从而促进了基质丰富和免疫抑制的 TME。接下来,我们发现CPT1C+CAFs在体外促进了巨噬细胞的M2样表型。生物信息学分析揭示了CPT1C+CAFs与巨噬细胞M2样表型之间强有力的IL-6信号传导,并确定CPT1C+CAFs是IL-6的主要来源。同时,抑制CPT1C在CAFs中的表达可显著减少体外IL-6的分泌。最后,我们证明了 CPT1C+CAFs 与治疗耐药性的关系。值得注意的是,在临床队列中,CPT1C+CAFs高浸润的GC患者对免疫疗法的反应较差。总之,我们的数据不仅新颖地发现了 CPT1C+CAFs 是 GC TME 中的免疫抑制亚群,还揭示了 CPT1C+CAFs 通过分泌 IL-6 诱导 GC 中巨噬细胞的免疫抑制 M2 样表型来损害肿瘤免疫的潜在机制。
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
Oncoimmunology
Oncoimmunology ONCOLOGYIMMUNOLOGY-IMMUNOLOGY
CiteScore
12.50
自引率
2.80%
发文量
276
审稿时长
24 weeks
期刊介绍: OncoImmunology is a dynamic, high-profile, open access journal that comprehensively covers tumor immunology and immunotherapy. As cancer immunotherapy advances, OncoImmunology is committed to publishing top-tier research encompassing all facets of basic and applied tumor immunology. The journal covers a wide range of topics, including: -Basic and translational studies in immunology of both solid and hematological malignancies -Inflammation, innate and acquired immune responses against cancer -Mechanisms of cancer immunoediting and immune evasion -Modern immunotherapies, including immunomodulators, immune checkpoint inhibitors, T-cell, NK-cell, and macrophage engagers, and CAR T cells -Immunological effects of conventional anticancer therapies.
×
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