CREB3L1 facilitates pancreatic tumor progression and reprograms intratumoral tumor-associated macrophages to shape an immunotherapy-resistance microenvironment.

IF 10.3 1区 医学 Q1 IMMUNOLOGY Journal for Immunotherapy of Cancer Pub Date : 2025-01-06 DOI:10.1136/jitc-2024-010029
Haiyan Xu, Shengbai Xue, Yang Sun, Jingyu Ma, Shumin Li, Yanling Wang, Tiebo Mao, Weiyu Ge, Ming Yue, Daiyuan Shentu, Wenxin Lu, Yongchao Wang, Jiong Hu, Jiujie Cui, Xiaofei Zhang, Li Cai, Yu Wang, Liwei Wang
{"title":"CREB3L1 facilitates pancreatic tumor progression and reprograms intratumoral tumor-associated macrophages to shape an immunotherapy-resistance microenvironment.","authors":"Haiyan Xu, Shengbai Xue, Yang Sun, Jingyu Ma, Shumin Li, Yanling Wang, Tiebo Mao, Weiyu Ge, Ming Yue, Daiyuan Shentu, Wenxin Lu, Yongchao Wang, Jiong Hu, Jiujie Cui, Xiaofei Zhang, Li Cai, Yu Wang, Liwei Wang","doi":"10.1136/jitc-2024-010029","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>To date, a growing body of evidence suggests that unfolded protein response (UPR) sensors play a vital role in carcinogenesis. However, it remains unclear whether they are involved in pancreatic ductal adenocarcinoma (PDAC) and how they relate to clinical outcomes. This study aims to investigate the biological function and mechanism of how a novel UPR sensor, CREB3L1 works in PDAC and further evaluate its clinical application prospect.</p><p><strong>Methods: </strong>We tested UPR signaling including CREB3L1 in Thapsigargin-treated PDAC cells. Subsequently, we defined CREB3L1 expression and further analyzed its expression with clinical characteristics in PDAC. Then, we established gene-modified cells to determine whether CREB3L1 functions in cell proliferation and migration capacity. Besides, we constructed subcutaneously and orthotopically transplanted mice models to verify their progrowing function and pulmonary metastasis models to prove their proinvasion role. What's more, RNAseq, qPCR, Western blotting, immunohistochemistry and multicolor flow cytometry experiments were used to explore the mechanism of how CREB3L1 worked in PDAC. Lastly, CREB3L1 expression correlation with PDAC immunotherapy outcome and immune cell signatures were explored in the patients with advanced PDAC who received PD-1 antibody therapy.</p><p><strong>Results: </strong>We first confirmed CREB3L1 could be induced by endoplasmic reticulum stressor and found its aberrant activation was associated with poorer overall survival in PDAC patients indicating the protumor function of the new UPR sensor. Functionally, we confirmed CREB3L1 contributing to PDAC malignant progression including growth and metastasis by multiple in in vitro and in vivo models. Mechanistically, CREB3L1 upregulated COL3A1 and promoted dense stroma formation for facilitating PDAC and knocking down COL3A1 disrupted CREB3L1 protumor function. Furthermore, CREB3L1-induced TAM polarization toward an M2 phenotype and reduced the infiltration of CD8<sup>+</sup> T cells. Clinically, CREB3L1 correlated with immune cell signatures as well as immune checkpoint blockade (ICB) treatment response and outcome that CREB3L1aberrant activation indicated poorer efficacy and worse prognosis than the low in PDAC which might empower clinical decision.</p><p><strong>Conclusions: </strong>Collectively, this study revealed CREB3L1 facilitated PDAC progression, shaped an immune exclude tumor microenvironment and distinguished therapy response and outcome of ICB therapy indicating CREB3L1 could be a promising novel molecular target and biomarker for PDAC treatment.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3000,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749327/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal for Immunotherapy of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1136/jitc-2024-010029","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: To date, a growing body of evidence suggests that unfolded protein response (UPR) sensors play a vital role in carcinogenesis. However, it remains unclear whether they are involved in pancreatic ductal adenocarcinoma (PDAC) and how they relate to clinical outcomes. This study aims to investigate the biological function and mechanism of how a novel UPR sensor, CREB3L1 works in PDAC and further evaluate its clinical application prospect.

Methods: We tested UPR signaling including CREB3L1 in Thapsigargin-treated PDAC cells. Subsequently, we defined CREB3L1 expression and further analyzed its expression with clinical characteristics in PDAC. Then, we established gene-modified cells to determine whether CREB3L1 functions in cell proliferation and migration capacity. Besides, we constructed subcutaneously and orthotopically transplanted mice models to verify their progrowing function and pulmonary metastasis models to prove their proinvasion role. What's more, RNAseq, qPCR, Western blotting, immunohistochemistry and multicolor flow cytometry experiments were used to explore the mechanism of how CREB3L1 worked in PDAC. Lastly, CREB3L1 expression correlation with PDAC immunotherapy outcome and immune cell signatures were explored in the patients with advanced PDAC who received PD-1 antibody therapy.

Results: We first confirmed CREB3L1 could be induced by endoplasmic reticulum stressor and found its aberrant activation was associated with poorer overall survival in PDAC patients indicating the protumor function of the new UPR sensor. Functionally, we confirmed CREB3L1 contributing to PDAC malignant progression including growth and metastasis by multiple in in vitro and in vivo models. Mechanistically, CREB3L1 upregulated COL3A1 and promoted dense stroma formation for facilitating PDAC and knocking down COL3A1 disrupted CREB3L1 protumor function. Furthermore, CREB3L1-induced TAM polarization toward an M2 phenotype and reduced the infiltration of CD8+ T cells. Clinically, CREB3L1 correlated with immune cell signatures as well as immune checkpoint blockade (ICB) treatment response and outcome that CREB3L1aberrant activation indicated poorer efficacy and worse prognosis than the low in PDAC which might empower clinical decision.

Conclusions: Collectively, this study revealed CREB3L1 facilitated PDAC progression, shaped an immune exclude tumor microenvironment and distinguished therapy response and outcome of ICB therapy indicating CREB3L1 could be a promising novel molecular target and biomarker for PDAC treatment.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
CREB3L1促进胰腺肿瘤进展并重新编程肿瘤内肿瘤相关巨噬细胞,形成免疫治疗抵抗微环境。
背景:迄今为止,越来越多的证据表明未折叠蛋白反应(UPR)传感器在癌症发生中起着至关重要的作用。然而,目前尚不清楚它们是否与胰腺导管腺癌(PDAC)有关,以及它们与临床结果的关系。本研究旨在探讨新型UPR传感器CREB3L1在PDAC中的生物学功能和作用机制,并进一步评价其临床应用前景。方法:我们在thapsigargin处理的PDAC细胞中检测包括CREB3L1在内的UPR信号。随后,我们定义了CREB3L1的表达,并进一步分析了其在PDAC中的表达与临床特征。然后,我们建立了基因修饰的细胞,以确定CREB3L1是否在细胞增殖和迁移能力中起作用。此外,我们建立小鼠皮下和原位移植模型来验证其促生长功能,并建立肺转移模型来证明其预防侵袭作用。通过RNAseq、qPCR、Western blotting、免疫组织化学、多色流式细胞术等实验探讨CREB3L1在PDAC中的作用机制。最后,在接受PD-1抗体治疗的晚期PDAC患者中,探讨CREB3L1表达与PDAC免疫治疗结果和免疫细胞特征的相关性。结果:我们首次证实了CREB3L1可以被内质网应激源诱导,并发现其异常激活与PDAC患者较差的总生存率相关,这表明了新型UPR传感器的肿瘤功能。在功能上,我们通过多种体外和体内模型证实CREB3L1参与了PDAC的恶性进展,包括生长和转移。机制上,CREB3L1上调COL3A1,促进致密间质形成,促进PDAC,敲低COL3A1破坏CREB3L1蛋白功能。此外,creb3l1诱导TAM向M2表型极化,减少CD8+ T细胞的浸润。在临床上,CREB3L1与免疫细胞特征和免疫检查点阻断(ICB)治疗反应相关,与PDAC低水平相比,CREB3L1异常激活的疗效更差,预后更差,这可能有助于临床决策。结论:总的来说,本研究揭示了CREB3L1促进PDAC进展,形成免疫排斥肿瘤微环境,并区分ICB治疗的治疗反应和结果,表明CREB3L1可能是一个有希望的PDAC治疗的新分子靶点和生物标志物。
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 去求助
来源期刊
Journal for Immunotherapy of Cancer
Journal for Immunotherapy of Cancer Biochemistry, Genetics and Molecular Biology-Molecular Medicine
CiteScore
17.70
自引率
4.60%
发文量
522
审稿时长
18 weeks
期刊介绍: The Journal for ImmunoTherapy of Cancer (JITC) is a peer-reviewed publication that promotes scientific exchange and deepens knowledge in the constantly evolving fields of tumor immunology and cancer immunotherapy. With an open access format, JITC encourages widespread access to its findings. The journal covers a wide range of topics, spanning from basic science to translational and clinical research. Key areas of interest include tumor-host interactions, the intricate tumor microenvironment, animal models, the identification of predictive and prognostic immune biomarkers, groundbreaking pharmaceutical and cellular therapies, innovative vaccines, combination immune-based treatments, and the study of immune-related toxicity.
期刊最新文献
What's in a name? Memory NK cells for cancer immunotherapy. Blocking the CD39/CD73 pathway synergizes with anti-CD20 bispecific antibody in nodal B-cell lymphoma. Arginase-1-specific T cells target and modulate tumor-associated macrophages. Rapamycin-resistant polyclonal Th1/Tc1 cell therapy (RAPA-201) safely induces disease remissions in relapsed, refractory multiple myeloma. Combined PARP14 inhibition and PD-1 blockade promotes cytotoxic T cell quiescence and modulates macrophage polarization in relapsed melanoma.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1