首页 > 最新文献

Molecular Cancer最新文献

英文 中文
Correction: Circular RNA ACVR2A suppresses bladder cancer cells proliferation and metastasis through miR-626/EYA4 axis. 更正:环状RNA ACVR2A通过miR-626/EYA4轴抑制膀胱癌细胞增殖和转移。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-22 DOI: 10.1186/s12943-025-02528-y
Wei Dong, Junming Bi, Hongwei Liu, Dong Yan, Qingqing He, Qianghua Zhou, Qiong Wang, Ruihui Xie, Yinjie Su, Meihua Yang, Tianxin Lin, Jian Huang
{"title":"Correction: Circular RNA ACVR2A suppresses bladder cancer cells proliferation and metastasis through miR-626/EYA4 axis.","authors":"Wei Dong, Junming Bi, Hongwei Liu, Dong Yan, Qingqing He, Qianghua Zhou, Qiong Wang, Ruihui Xie, Yinjie Su, Meihua Yang, Tianxin Lin, Jian Huang","doi":"10.1186/s12943-025-02528-y","DOIUrl":"10.1186/s12943-025-02528-y","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"24 1","pages":"309"},"PeriodicalIF":33.9,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12720428/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145810654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sitravatinib plus tislelizumab in locally recurrent or metastatic triple-negative breast cancer: a multi-cohort, single-arm, phase II clinical trial (SPARK Trial). 西特拉替尼联合替利单抗治疗局部复发或转移性三阴性乳腺癌:一项多队列、单组、II期临床试验(SPARK试验)
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-19 DOI: 10.1186/s12943-025-02505-5
Xi-Yu Liu,Xin-Yi Sui,Ying Xu,Fan Yang,Song-Yang Wu,Xiu-Zhi Zhu,Ke Zuo,Shuo-Wen Cao,Xi Jin,Li Chen,Lin-Xiaoxi Ma,Wen-Juan Zhang,Fu-Gui Ye,Fei-Lin Qu,Ding Ma,Yi Xiao,Gen-Hong Di,Guang-Yu Liu,Ke-Da Yu,Jiong Wu,Xin Hu,Yi-Zhou Jiang,Zhong-Hua Wang,Zhi-Ming Shao,Lei Fan
{"title":"Sitravatinib plus tislelizumab in locally recurrent or metastatic triple-negative breast cancer: a multi-cohort, single-arm, phase II clinical trial (SPARK Trial).","authors":"Xi-Yu Liu,Xin-Yi Sui,Ying Xu,Fan Yang,Song-Yang Wu,Xiu-Zhi Zhu,Ke Zuo,Shuo-Wen Cao,Xi Jin,Li Chen,Lin-Xiaoxi Ma,Wen-Juan Zhang,Fu-Gui Ye,Fei-Lin Qu,Ding Ma,Yi Xiao,Gen-Hong Di,Guang-Yu Liu,Ke-Da Yu,Jiong Wu,Xin Hu,Yi-Zhou Jiang,Zhong-Hua Wang,Zhi-Ming Shao,Lei Fan","doi":"10.1186/s12943-025-02505-5","DOIUrl":"https://doi.org/10.1186/s12943-025-02505-5","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"4 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145786074","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Next-generation CAR-T cells design: leveraging tumor features for enhanced efficacy. 下一代CAR-T细胞设计:利用肿瘤特征增强疗效。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-19 DOI: 10.1186/s12943-025-02515-3
Yanna Lei,Ning Liu,Diyuan Qin,Ming Liu,Yongsheng Wang
{"title":"Next-generation CAR-T cells design: leveraging tumor features for enhanced efficacy.","authors":"Yanna Lei,Ning Liu,Diyuan Qin,Ming Liu,Yongsheng Wang","doi":"10.1186/s12943-025-02515-3","DOIUrl":"https://doi.org/10.1186/s12943-025-02515-3","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"46 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145786076","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cancer-associated fibroblasts in the tumor microenvironment: heterogeneity, crosstalk mechanisms, and therapeutic implications. 肿瘤微环境中的癌症相关成纤维细胞:异质性、串扰机制和治疗意义。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-18 DOI: 10.1186/s12943-025-02533-1
Ziyue Huang,Jie Chen,Tianyu Zhu,Jinxin Li,Ho Yin Ng,Yixiong Zhou,Xiang Gu,Shiqiong Xu,Renbing Jia
The tumor microenvironment (TME) is increasingly recognized as a critical modulator of the initiation, progression, metastasis, and therapeutic resistance of various cancers. Cancer-associated fibroblasts (CAFs), the predominant stromal cell population within the TME, play pivotal roles in these processes through their remarkable phenotypic and functional heterogeneity. Emerging evidence underscores the diversity in the origins, phenotypes, and functions of CAFs, highlighting their ability to adaptively influence tumor biology in a context-dependent manner. CAFs facilitate cancer malignancy via multiple interconnected mechanisms, including the secretion of soluble bioactive factors, the release of exosomes, the metabolic reprogramming of tumor cells, the remodeling of the extracellular matrix (ECM), and the modulation of the immune microenvironment. CAFs have emerged as attractive and viable therapeutic targets. Recent efforts have focused on developing therapies that disrupt the protumorigenic activities of CAFs or reprogram them toward tumor-suppressive phenotypes. Several of these strategies have shown promise and are advancing into clinical trials. In this review, we comprehensively discuss recent advancements in our understanding of the heterogeneity of CAFs, elucidate their multifaceted interactions within the TME, and explore novel therapeutic strategies targeting CAFs across various cancer types. Our review aims to foster the translation of preclinical insights into clinically effective interventions targeting CAFs.
肿瘤微环境(tumor microenvironment, TME)越来越被认为是各种癌症发生、进展、转移和治疗耐药的关键调节因子。癌症相关成纤维细胞(CAFs)是TME中主要的基质细胞群,通过其显著的表型和功能异质性在这些过程中发挥关键作用。新出现的证据强调了caf的起源、表型和功能的多样性,强调了它们以环境依赖的方式适应性地影响肿瘤生物学的能力。CAFs通过多种相互关联的机制促进肿瘤恶性发展,包括可溶性生物活性因子的分泌、外泌体的释放、肿瘤细胞的代谢重编程、细胞外基质(ECM)的重塑以及免疫微环境的调节。CAFs已成为有吸引力和可行的治疗靶点。最近的努力集中在开发破坏caf的致瘤活性或将其重新编程为肿瘤抑制表型的治疗方法。其中一些策略已经显示出前景,并正在进入临床试验阶段。在这篇综述中,我们全面讨论了我们对CAFs异质性的理解的最新进展,阐明了它们在TME中的多方面相互作用,并探索了针对不同癌症类型的CAFs的新治疗策略。我们的综述旨在促进临床前见解转化为针对CAFs的临床有效干预措施。
{"title":"Cancer-associated fibroblasts in the tumor microenvironment: heterogeneity, crosstalk mechanisms, and therapeutic implications.","authors":"Ziyue Huang,Jie Chen,Tianyu Zhu,Jinxin Li,Ho Yin Ng,Yixiong Zhou,Xiang Gu,Shiqiong Xu,Renbing Jia","doi":"10.1186/s12943-025-02533-1","DOIUrl":"https://doi.org/10.1186/s12943-025-02533-1","url":null,"abstract":"The tumor microenvironment (TME) is increasingly recognized as a critical modulator of the initiation, progression, metastasis, and therapeutic resistance of various cancers. Cancer-associated fibroblasts (CAFs), the predominant stromal cell population within the TME, play pivotal roles in these processes through their remarkable phenotypic and functional heterogeneity. Emerging evidence underscores the diversity in the origins, phenotypes, and functions of CAFs, highlighting their ability to adaptively influence tumor biology in a context-dependent manner. CAFs facilitate cancer malignancy via multiple interconnected mechanisms, including the secretion of soluble bioactive factors, the release of exosomes, the metabolic reprogramming of tumor cells, the remodeling of the extracellular matrix (ECM), and the modulation of the immune microenvironment. CAFs have emerged as attractive and viable therapeutic targets. Recent efforts have focused on developing therapies that disrupt the protumorigenic activities of CAFs or reprogram them toward tumor-suppressive phenotypes. Several of these strategies have shown promise and are advancing into clinical trials. In this review, we comprehensively discuss recent advancements in our understanding of the heterogeneity of CAFs, elucidate their multifaceted interactions within the TME, and explore novel therapeutic strategies targeting CAFs across various cancer types. Our review aims to foster the translation of preclinical insights into clinically effective interventions targeting CAFs.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"24 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145777317","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting the tumor cell-intrinsic ITGB2 axis inhibits melanoma progression. 靶向肿瘤细胞固有ITGB2轴抑制黑色素瘤进展。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-18 DOI: 10.1186/s12943-025-02527-z
Erik Rasbach,Laure Migayron,Anne Brandenburg,Praveen Singh,Christina Martins,Ali Kassem,Anastasia I Karkempetzaki,Nadine Suessner,Zsofi Kulcsar,Jessica Riopedre,Mariana Silva,Ethan Zhen,Shuyun Xu,Kyla Mucciarone,Julia Holzgruber,Jason B Williams,Emrullah Birgin,Mitchell P Levesque,Julia M Martínez-Gómez,Reinhard Dummer,Jennifer Landsberg,Christine G Lian,George F Murphy,Emma L Berdan,Shannan Ho Sui,Thomas S Kupper,Nuh N Rahbari,Nayoung Lee,Steven R Barthel,Tobias Schatton
BACKGROUNDITGB2 is a critical integrin mediator of immune cell activation and trafficking. Its expression has been claimed as exclusive to hematopoietic cells. Consequently, the significance of cancer cell-intrinsic ITGB2 in solid tumor progression and therapy has not been rigorously evaluated.METHODSWe leveraged single-cell and bulk RNA sequencing, real-time quantitative PCR, multiplex immunofluorescence, flow cytometry, immunoblotting, and intercellular adhesion molecule (ICAM)-1-dependent adhesion and proliferation assays to uncover melanoma cell-intrinsic ITGB2 functional expression, association with clinical tumor progression, activation, protumorigenic signaling, adhesive and proliferative functions utilizing patient melanoma biospecimens, established human and murine melanoma lines. In vivo tumorigenicity studies in immunocompromised NOD/SCID interleukin-2 receptor γ chain null (NSG), immunocompetent wildtype, and Icam1 knockout (KO) C57BL/6 mice were performed to dissect melanoma-ITGB2 downstream pathway activity and functions in tumor growth and metastasis. The cancer cell-intrinsic ITGB2 axis was targeted using CRISPR/Cas9-based Itgb2 KO, blocking ITGB2 antibodies, ITGB2-activating CD44 crosslinking, and pharmacologic inhibition of ITGB2-dependent Wnt signaling using LGK974, zamaporvint, and FDA-approved pyrvinium pamoate repurposed for cancer therapy.RESULTSThis work demonstrates nonhematopoietic expression and protumorigenic functions of ITGB2 intrinsic to melanoma cells. Tumor cell-ITGB2 mediated adhesion to ICAM-1, promoted cancer progression in preclinical melanoma models, was enriched in clinical metastatic versus primary melanomas or benign nevi, and predicted sentinel lymph node metastasis in patients with primary disease. Consistently, inhibition of melanoma cell-intrinsic ITGB2 using blocking antibodies or Itgb2 gene KO potently suppressed ICAM-1-mediated melanoma cell adhesion, tumor growth, and metastatic dissemination. Melanoma cell-ITGB2:ICAM-1 interaction activated downstream Wnt signaling, the pharmacologic inhibition of which suppressed melanoma-ITGB2-mediated tumorigenesis.CONCLUSIONSThis work overturns the longstanding paradigm that ITGB2 is restricted to leukocytes by discovering a tumor cell-intrinsic ITGB2:ICAM-1:Wnt protumorigenic axis as a bona fide cancer therapeutic target in melanoma.
背景gb2是免疫细胞激活和运输的重要整合素介质。它的表达被认为是造血细胞所独有的。因此,癌细胞固有的ITGB2在实体瘤进展和治疗中的意义尚未得到严格的评估。方法利用黑色素瘤患者生物标本,利用单细胞和大量RNA测序、实时定量PCR、多重免疫荧光、流式细胞术、免疫印迹和细胞间粘附分子(ICAM)-1依赖性粘附和增殖检测,揭示黑色素瘤细胞内在ITGB2功能表达、与临床肿瘤进展、激活、致瘤信号、粘附和增殖功能的关联。建立了人类和小鼠黑色素瘤细胞系。通过对NOD/SCID白介素-2受体γ链缺失(NSG)、免疫正常野生型和Icam1敲除(KO) C57BL/6小鼠的体内致瘤性研究,探讨了黑色素瘤- itgb2下游通路在肿瘤生长和转移中的活性和功能。使用基于CRISPR/ cas9的ITGB2 KO靶向癌细胞固有的ITGB2轴,阻断ITGB2抗体,激活ITGB2的CD44交联,并使用LGK974, zamaporvint和fda批准的用于癌症治疗的pyrvinium pamoate对ITGB2依赖性Wnt信号进行药理学抑制。结果本研究证实了ITGB2在黑色素瘤细胞中的非造血表达和致瘤功能。肿瘤细胞itgb2介导对ICAM-1的粘附,在临床前黑色素瘤模型中促进肿瘤进展,在临床转移性黑色素瘤与原发性黑色素瘤或良性痣中富集,并预测原发性疾病患者的前哨淋巴结转移。一致地,使用阻断抗体或ITGB2基因KO抑制黑色素瘤细胞固有的ITGB2可以有效地抑制icam -1介导的黑色素瘤细胞粘附、肿瘤生长和转移传播。黑色素瘤细胞- itgb2:ICAM-1相互作用激活下游Wnt信号,其药理抑制抑制黑色素瘤- itgb2介导的肿瘤发生。本研究通过发现肿瘤细胞固有的ITGB2:ICAM-1:Wnt蛋白原轴作为黑色素瘤的真正癌症治疗靶点,推翻了ITGB2局限于白细胞的长期范式。
{"title":"Targeting the tumor cell-intrinsic ITGB2 axis inhibits melanoma progression.","authors":"Erik Rasbach,Laure Migayron,Anne Brandenburg,Praveen Singh,Christina Martins,Ali Kassem,Anastasia I Karkempetzaki,Nadine Suessner,Zsofi Kulcsar,Jessica Riopedre,Mariana Silva,Ethan Zhen,Shuyun Xu,Kyla Mucciarone,Julia Holzgruber,Jason B Williams,Emrullah Birgin,Mitchell P Levesque,Julia M Martínez-Gómez,Reinhard Dummer,Jennifer Landsberg,Christine G Lian,George F Murphy,Emma L Berdan,Shannan Ho Sui,Thomas S Kupper,Nuh N Rahbari,Nayoung Lee,Steven R Barthel,Tobias Schatton","doi":"10.1186/s12943-025-02527-z","DOIUrl":"https://doi.org/10.1186/s12943-025-02527-z","url":null,"abstract":"BACKGROUNDITGB2 is a critical integrin mediator of immune cell activation and trafficking. Its expression has been claimed as exclusive to hematopoietic cells. Consequently, the significance of cancer cell-intrinsic ITGB2 in solid tumor progression and therapy has not been rigorously evaluated.METHODSWe leveraged single-cell and bulk RNA sequencing, real-time quantitative PCR, multiplex immunofluorescence, flow cytometry, immunoblotting, and intercellular adhesion molecule (ICAM)-1-dependent adhesion and proliferation assays to uncover melanoma cell-intrinsic ITGB2 functional expression, association with clinical tumor progression, activation, protumorigenic signaling, adhesive and proliferative functions utilizing patient melanoma biospecimens, established human and murine melanoma lines. In vivo tumorigenicity studies in immunocompromised NOD/SCID interleukin-2 receptor γ chain null (NSG), immunocompetent wildtype, and Icam1 knockout (KO) C57BL/6 mice were performed to dissect melanoma-ITGB2 downstream pathway activity and functions in tumor growth and metastasis. The cancer cell-intrinsic ITGB2 axis was targeted using CRISPR/Cas9-based Itgb2 KO, blocking ITGB2 antibodies, ITGB2-activating CD44 crosslinking, and pharmacologic inhibition of ITGB2-dependent Wnt signaling using LGK974, zamaporvint, and FDA-approved pyrvinium pamoate repurposed for cancer therapy.RESULTSThis work demonstrates nonhematopoietic expression and protumorigenic functions of ITGB2 intrinsic to melanoma cells. Tumor cell-ITGB2 mediated adhesion to ICAM-1, promoted cancer progression in preclinical melanoma models, was enriched in clinical metastatic versus primary melanomas or benign nevi, and predicted sentinel lymph node metastasis in patients with primary disease. Consistently, inhibition of melanoma cell-intrinsic ITGB2 using blocking antibodies or Itgb2 gene KO potently suppressed ICAM-1-mediated melanoma cell adhesion, tumor growth, and metastatic dissemination. Melanoma cell-ITGB2:ICAM-1 interaction activated downstream Wnt signaling, the pharmacologic inhibition of which suppressed melanoma-ITGB2-mediated tumorigenesis.CONCLUSIONSThis work overturns the longstanding paradigm that ITGB2 is restricted to leukocytes by discovering a tumor cell-intrinsic ITGB2:ICAM-1:Wnt protumorigenic axis as a bona fide cancer therapeutic target in melanoma.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"31 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145777319","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting OxLDL-mediated CD36 + CAF reprogramming potentiates PD-1 immunotherapy in osteosarcoma. 靶向oxldl介导的CD36 + CAF重编程增强骨肉瘤的PD-1免疫治疗。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-18 DOI: 10.1186/s12943-025-02516-2
Anyu Zeng,Hongmin Chen,Tianqi Luo,Weiqing Chen,Yihui Song,Yanyang Xu,Zhihao Chen,Qinglian Tang,Xiaojun Zhu,Chuangzhong Deng,Huaiyuan Xu,Anqi Wang,Hao Wu,Guohui Song,Jinchang Lu,Jin Wang
BACKGROUNDOsteosarcoma demonstrates limited responsiveness to PD-1 blockade, largely due to its immunosuppressive tumor microenvironment (TME). The specific mechanisms by which cancer-associated fibroblasts (CAFs) contribute to immunosuppression in osteosarcoma are not fully understood.METHODSWe performed single-cell RNA sequencing (scRNA-seq) on osteosarcoma tissues from patients treated with neoadjuvant chemotherapy and anti-PD-1 therapy to investigate the tumor microenvironment. Cellular composition, gene expression programs, and signaling pathways were analyzed. Functional assays, pull-down and PLA-flow binding validation, and in vivo mouse models were used to dissect the mechanisms by which CAF-derived factors influence CD8⁺ T cell function and contribute to immunotherapy response.RESULTSWe identified a subpopulation of CD36⁺ CAFs, characterized by adaptive uptake of oxidized low-density lipoprotein (OxLDL) and activation of the PPARG-FABP4 axis. This metabolic program promoted ANGPTL4 secretion, which bound integrin on CD8⁺ T cells and activated the JAK2-STAT3 pathway, leading to T cell exhaustion and impaired effector function. In vivo, administration of VitE effectively scavenged OxLDL, reprogrammed the TME, enhanced CD8⁺ T cell infiltration, and synergized with PD-1 blockade to improve tumor control.CONCLUSIONSCD36⁺ CAFs drive immunosuppressive metabolic reprogramming via the OxLDL-PPARG-ANGPTL4 axis, promoting CD8⁺ T cell exhaustion and resistance to immunotherapy in osteosarcoma. Targeting this pathway with VitE alleviated CAF-mediated immune suppression and enhanced PD-1 blockade responses in preclinical models, providing a rationale for metabolism-based combinatorial strategies in osteosarcoma.
背景:骨肉瘤对PD-1阻断的反应有限,主要是由于其免疫抑制肿瘤微环境(TME)。癌症相关成纤维细胞(CAFs)参与骨肉瘤免疫抑制的具体机制尚不完全清楚。方法对接受新辅助化疗和抗pd -1治疗的骨肉瘤患者的组织进行单细胞RNA测序(scRNA-seq),研究肿瘤微环境。分析了细胞组成、基因表达程序和信号通路。通过功能分析、pull-down和PLA-flow结合验证以及体内小鼠模型,研究了cafd衍生因子影响CD8 + T细胞功能并促进免疫治疗反应的机制。我们鉴定了一个CD36 + CAFs亚群,其特征是适应性地摄取氧化低密度脂蛋白(OxLDL)和激活PPARG-FABP4轴。这种代谢程序促进ANGPTL4的分泌,将整合素结合在CD8 + T细胞上,激活JAK2-STAT3通路,导致T细胞衰竭和效应功能受损。在体内,给药VitE可有效清除OxLDL,重编程TME,增强CD8 + T细胞浸润,并与PD-1阻断协同改善肿瘤控制。结论scd36 + CAFs通过oxldl - ppar - angptl4轴驱动免疫抑制性代谢重编程,促进CD8 + T细胞衰竭和对骨肉瘤免疫治疗的抵抗。在临床前模型中,用VitE靶向这一途径减轻了cafc介导的免疫抑制和增强的PD-1阻断反应,为骨肉瘤基于代谢的组合策略提供了理论依据。
{"title":"Targeting OxLDL-mediated CD36 + CAF reprogramming potentiates PD-1 immunotherapy in osteosarcoma.","authors":"Anyu Zeng,Hongmin Chen,Tianqi Luo,Weiqing Chen,Yihui Song,Yanyang Xu,Zhihao Chen,Qinglian Tang,Xiaojun Zhu,Chuangzhong Deng,Huaiyuan Xu,Anqi Wang,Hao Wu,Guohui Song,Jinchang Lu,Jin Wang","doi":"10.1186/s12943-025-02516-2","DOIUrl":"https://doi.org/10.1186/s12943-025-02516-2","url":null,"abstract":"BACKGROUNDOsteosarcoma demonstrates limited responsiveness to PD-1 blockade, largely due to its immunosuppressive tumor microenvironment (TME). The specific mechanisms by which cancer-associated fibroblasts (CAFs) contribute to immunosuppression in osteosarcoma are not fully understood.METHODSWe performed single-cell RNA sequencing (scRNA-seq) on osteosarcoma tissues from patients treated with neoadjuvant chemotherapy and anti-PD-1 therapy to investigate the tumor microenvironment. Cellular composition, gene expression programs, and signaling pathways were analyzed. Functional assays, pull-down and PLA-flow binding validation, and in vivo mouse models were used to dissect the mechanisms by which CAF-derived factors influence CD8⁺ T cell function and contribute to immunotherapy response.RESULTSWe identified a subpopulation of CD36⁺ CAFs, characterized by adaptive uptake of oxidized low-density lipoprotein (OxLDL) and activation of the PPARG-FABP4 axis. This metabolic program promoted ANGPTL4 secretion, which bound integrin on CD8⁺ T cells and activated the JAK2-STAT3 pathway, leading to T cell exhaustion and impaired effector function. In vivo, administration of VitE effectively scavenged OxLDL, reprogrammed the TME, enhanced CD8⁺ T cell infiltration, and synergized with PD-1 blockade to improve tumor control.CONCLUSIONSCD36⁺ CAFs drive immunosuppressive metabolic reprogramming via the OxLDL-PPARG-ANGPTL4 axis, promoting CD8⁺ T cell exhaustion and resistance to immunotherapy in osteosarcoma. Targeting this pathway with VitE alleviated CAF-mediated immune suppression and enhanced PD-1 blockade responses in preclinical models, providing a rationale for metabolism-based combinatorial strategies in osteosarcoma.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"40 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145777322","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immune-related deubiquitylation spectrum of microsatellite stability colorectal cancer reveals USP7 as a potential immunotherapeutic target. 微卫星稳定性结直肠癌的免疫相关去泛素化谱显示USP7是一个潜在的免疫治疗靶点。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-18 DOI: 10.1186/s12943-025-02502-8
Xiaomao Yin,Jinran Wu,Mi-Die Xu,Tongguan Tian,Lin Zhu,Jiexuan Wang,Xuan Dai,Xin Yang,Jingjing Qian,Wenqiang Wang,Liangchen Zhu,Zekun Zhao,Kai Xu,Yanping Xu,Xinxing Li,Zhiqian Hu
The efficacy of immune checkpoint inhibitors (ICIs) in microsatellite stable colorectal cancer (MSS CRC) remains limited, highlighting an urgent need for predictive biomarkers. Through multi-omics analysis, we identified two novel MSS CRC subtypes, termed DUB-H and DUB-L. The DUB-L subtype exhibited an inflamed tumor immune microenvironment, a superior response to immune therapy, and better recurrence-free survival (RFS) compared to DUB-H. The classifier gene USP7 was selected as a gene of interest due to its specific expression profile, which is highly expressed in MSS CRC but not in microsatellite instability-high (MSI-H) tumors, and strongly correlated with suppressed immune infiltration. Large-scale clinical analyses confirmed associations between high USP7 expression, microsatellite stability, specific consensus molecular subtypes (CMS), and unfavorable prognosis. Single-cell analysis and multiplex immunofluorescence validated an immune-desert phenotype in USP7-high MSS tumors. Mechanistically, USP7 knockdown in MSS CRC cells enhances the secretion of T-cell-recruiting chemokines (CXCL9/10/11), promoting CD8⁺ T cell recruitment and cytotoxicity in vitro. In vivo experiments demonstrated that USP7 blockade enhanced the efficacy of anti-PD-1 treatment in MSS CRC models by remodeling the tumor immune microenvironment, increasing infiltration and function of CD8⁺ T and NK cells. Consistently, low USP7 expression is associated with a better response to anti-PD-1 therapy. Overall, we propose a novel DUB-based classification system for MSS CRC and demonstrate that targeting USP7 may overcome immunotherapy resistance by converting immunologically "cold" tumors into "hot" ones.
免疫检查点抑制剂(ICIs)在微卫星稳定型结直肠癌(MSS CRC)中的疗效仍然有限,这凸显了对预测性生物标志物的迫切需求。通过多组学分析,我们确定了两种新的MSS CRC亚型,称为DUB-H和DUB-L。与DUB-H相比,DUB-L亚型表现出炎症性肿瘤免疫微环境,对免疫治疗的反应更好,无复发生存期(RFS)更好。选择分类基因USP7作为感兴趣的基因是因为其特异性表达谱,该基因在MSS CRC中高表达,而在微卫星不稳定性高(MSI-H)肿瘤中不表达,并且与抑制免疫浸润密切相关。大规模临床分析证实了USP7高表达、微卫星稳定性、特异性共识分子亚型(CMS)和不良预后之间的关联。单细胞分析和多重免疫荧光证实了usp7 -高MSS肿瘤的免疫荒漠表型。在机制上,MSS CRC细胞中USP7敲低可增强T细胞募集趋化因子(CXCL9/10/11)的分泌,促进CD8 +在体外募集T细胞和细胞毒性。体内实验表明,USP7阻断剂通过重塑肿瘤免疫微环境,增加CD8 + T和NK细胞的浸润和功能,增强了MSS CRC模型抗pd -1治疗的效果。一致地,低USP7表达与抗pd -1治疗的更好反应相关。总之,我们提出了一种新的基于dub的MSS CRC分类系统,并证明靶向USP7可以通过将免疫“冷”肿瘤转化为“热”肿瘤来克服免疫治疗耐药性。
{"title":"Immune-related deubiquitylation spectrum of microsatellite stability colorectal cancer reveals USP7 as a potential immunotherapeutic target.","authors":"Xiaomao Yin,Jinran Wu,Mi-Die Xu,Tongguan Tian,Lin Zhu,Jiexuan Wang,Xuan Dai,Xin Yang,Jingjing Qian,Wenqiang Wang,Liangchen Zhu,Zekun Zhao,Kai Xu,Yanping Xu,Xinxing Li,Zhiqian Hu","doi":"10.1186/s12943-025-02502-8","DOIUrl":"https://doi.org/10.1186/s12943-025-02502-8","url":null,"abstract":"The efficacy of immune checkpoint inhibitors (ICIs) in microsatellite stable colorectal cancer (MSS CRC) remains limited, highlighting an urgent need for predictive biomarkers. Through multi-omics analysis, we identified two novel MSS CRC subtypes, termed DUB-H and DUB-L. The DUB-L subtype exhibited an inflamed tumor immune microenvironment, a superior response to immune therapy, and better recurrence-free survival (RFS) compared to DUB-H. The classifier gene USP7 was selected as a gene of interest due to its specific expression profile, which is highly expressed in MSS CRC but not in microsatellite instability-high (MSI-H) tumors, and strongly correlated with suppressed immune infiltration. Large-scale clinical analyses confirmed associations between high USP7 expression, microsatellite stability, specific consensus molecular subtypes (CMS), and unfavorable prognosis. Single-cell analysis and multiplex immunofluorescence validated an immune-desert phenotype in USP7-high MSS tumors. Mechanistically, USP7 knockdown in MSS CRC cells enhances the secretion of T-cell-recruiting chemokines (CXCL9/10/11), promoting CD8⁺ T cell recruitment and cytotoxicity in vitro. In vivo experiments demonstrated that USP7 blockade enhanced the efficacy of anti-PD-1 treatment in MSS CRC models by remodeling the tumor immune microenvironment, increasing infiltration and function of CD8⁺ T and NK cells. Consistently, low USP7 expression is associated with a better response to anti-PD-1 therapy. Overall, we propose a novel DUB-based classification system for MSS CRC and demonstrate that targeting USP7 may overcome immunotherapy resistance by converting immunologically \"cold\" tumors into \"hot\" ones.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"89 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145777320","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Amino acids metabolism: a potential target for cancer treatment. 氨基酸代谢:癌症治疗的潜在靶点。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-16 DOI: 10.1186/s12943-025-02523-3
Shiqi Ren,Xinyi Zhou,Zhen Wang,Kefei Yuan
Metabolic reprogramming of amino acids has been recognized as a significant characteristic in various types of cancers. Numerous studies have indicated that the metabolic reprogramming of amino acids in tumors significantly supports certain malignant behaviors, including tumor proliferation, survival, invasion, and even immune escape. Amino acids can provide biomolecules such as nucleotides and glutathione (GSH) for tumors, and the bioavailability of amino acids influences tumor progression. Meanwhile, as essential metabolites, amino acids are closely associated with immune cell activation and can contribute to tumor immune processes by modulating the function of immune cells. Thus, targeting amino acids metabolism has emerged as a promising therapeutic strategy. Herein, we provide an overview of the effects of amino acids on the central carbon cycle and autophagy. We also provide an in-depth review of potential therapies for cancer treatment associated with amino acids, including metabolic enzymes of amino acids, dietary therapy of amino acids, and so on. Furthermore, we summarize some current nano-systems relevant to amino acids. This review aims to offer a theoretical foundation for understanding amino acids metabolism in cancer and identifying potential therapeutic strategies.
氨基酸的代谢重编程已被认为是各种类型癌症的一个重要特征。大量研究表明,肿瘤中氨基酸的代谢重编程显著支持某些恶性行为,包括肿瘤的增殖、存活、侵袭甚至免疫逃逸。氨基酸可以为肿瘤提供生物分子,如核苷酸和谷胱甘肽(GSH),氨基酸的生物利用度影响肿瘤的进展。同时,氨基酸作为必需代谢物,与免疫细胞活化密切相关,可通过调节免疫细胞功能参与肿瘤免疫过程。因此,靶向氨基酸代谢已成为一种有前景的治疗策略。在这里,我们提供了氨基酸对中心碳循环和自噬的影响的概述。我们还对与氨基酸相关的潜在癌症治疗方法进行了深入的综述,包括氨基酸代谢酶、氨基酸饮食疗法等。此外,我们还总结了目前一些与氨基酸相关的纳米系统。本文旨在为了解氨基酸在癌症中的代谢和确定潜在的治疗策略提供理论基础。
{"title":"Amino acids metabolism: a potential target for cancer treatment.","authors":"Shiqi Ren,Xinyi Zhou,Zhen Wang,Kefei Yuan","doi":"10.1186/s12943-025-02523-3","DOIUrl":"https://doi.org/10.1186/s12943-025-02523-3","url":null,"abstract":"Metabolic reprogramming of amino acids has been recognized as a significant characteristic in various types of cancers. Numerous studies have indicated that the metabolic reprogramming of amino acids in tumors significantly supports certain malignant behaviors, including tumor proliferation, survival, invasion, and even immune escape. Amino acids can provide biomolecules such as nucleotides and glutathione (GSH) for tumors, and the bioavailability of amino acids influences tumor progression. Meanwhile, as essential metabolites, amino acids are closely associated with immune cell activation and can contribute to tumor immune processes by modulating the function of immune cells. Thus, targeting amino acids metabolism has emerged as a promising therapeutic strategy. Herein, we provide an overview of the effects of amino acids on the central carbon cycle and autophagy. We also provide an in-depth review of potential therapies for cancer treatment associated with amino acids, including metabolic enzymes of amino acids, dietary therapy of amino acids, and so on. Furthermore, we summarize some current nano-systems relevant to amino acids. This review aims to offer a theoretical foundation for understanding amino acids metabolism in cancer and identifying potential therapeutic strategies.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"56 1","pages":"307"},"PeriodicalIF":37.3,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145765479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epithelial WNT secretion drives niche escape of developing gastric cancer. 上皮细胞WNT分泌驱动发生胃癌的生态位逃逸。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-16 DOI: 10.1186/s12943-025-02543-z
Jaehun Lee,Soomin Kim,Youngchul Oh,Stephan R Jahn,Jihoon Kim,Yeongjun Kim,Tim Schmäche,Sang-Min Kim,Isaree Teriyapirom,Thomas Groß,Ohbin Kwon,Jungmin Kim,Somi Kim,Anne-Marlen Ada,Andrea Català-Bordes,Youngwon Cho,Jinho Kim,Amanda Andersson-Rolf,Sebastian R Merker,Joo Yeon Lim,Ji-Yeon Park,Thomas M Klompstra,Ki-Jun Yoon,Dae-Sik Lim,Ho-Seok Lee,Jong Kyoung Kim,Eunyoung Choi,James R Goldenring,Jae-Ho Cheong,Hyunki Kim,Daniel E Stange,Heetak Lee,Bon-Kyoung Koo,Ji-Hyun Lee
BACKGROUNDWNT signaling plays a key role in maintaining the gastric epithelium and promoting tumorigenesis. However, how gastric tumors achieve WNT niche independence remains unclear, as mutations on APC or CTNNB1-common mechanisms of ligand-independent WNT activation in colorectal cancer-are infrequent in gastric cancer. Understanding how WNT self-sufficiency is acquired in the stomach is therefore critical.METHODSWe analyzed mouse gastric organoids harboring oncogenic KRASG12D with or without RNF43/ZNRF3 (RZ) or CDH1/TP53 (CP) mutations, along with corresponding in vivo mouse models. Niche independence was assessed through growth factor withdrawal, Porcupine and pathway-specific inhibitor treatments, and WNT rescue assays. We performed single-nucleus multiome sequencing (RNA + ATAC) to investigate transcriptional and chromatin dynamics. Findings from mouse models were validated using patient-derived gastric cancer organoids, and pan-cancer cell line datasets were analyzed to evaluate clinical and cross-tissue relevance.RESULTSGastric fibroblasts secreted canonical WNT2B to maintain the homeostatic gastric epithelium. Upon KRAS activation, epithelial cells were reprogrammed to secrete WNT ligands independently of additional mutations. Single-nucleus multiome analysis revealed that KRAS-driven MAPK signaling opened SMAD2/3-bound enhancers at the WNT7B locus, leading to the emergence of WNT7B-expressing subpopulations. Inhibition of SMAD2/3 phosphorylation suppressed both organoid growth and WNT7B transcription, whereas exogenous WNT restored organoid proliferation. Patient-derived organoids with HER2 amplification, KRAS amplification, or WNT2 copy-number gain exhibited Porcupine inhibitor-sensitive growth, indicating dependence on WNT secretion from the organoids. Analysis of public transcriptomic datasets further demonstrated that the KRAS-MAPK-WNT7B axis is conserved across other cancer types, including lung cancer.CONCLUSIONSGastric tumors can bypass niche dependence by acquiring KRAS-MAPK-SMAD2/3-driven epithelial WNT secretion. Targeting this axis-through MAPK inhibition, SMAD2/3 blockade, or suppression of WNT secretion-may represent a therapeutic vulnerability in gastric cancer and other KRAS-high malignancies.
背景wnt信号在维持胃上皮和促进肿瘤发生中起关键作用。然而,胃肿瘤如何实现WNT生态位独立性仍不清楚,因为APC或ctnnb1(结直肠癌中常见的不依赖配体的WNT激活机制)的突变在胃癌中并不常见。因此,了解WNT如何在胃中获得自给自足是至关重要的。方法我们分析了携带或不携带RNF43/ZNRF3 (RZ)或CDH1/TP53 (CP)突变的胃癌类器官KRASG12D,以及相应的小鼠体内模型。通过生长因子停药、豪猪和通路特异性抑制剂治疗以及WNT拯救试验来评估生态位独立性。我们进行了单核多组测序(RNA + ATAC)来研究转录和染色质动力学。使用患者来源的胃癌类器官验证小鼠模型的发现,并分析泛癌细胞系数据集以评估临床和跨组织相关性。结果胃成纤维细胞分泌典型的WNT2B维持胃上皮的稳态。KRAS激活后,上皮细胞被重编程为独立于其他突变分泌WNT配体。单核多组分析显示,kras驱动的MAPK信号在WNT7B位点打开smad2 /3结合的增强子,导致表达WNT7B的亚群出现。抑制SMAD2/3磷酸化抑制类器官生长和WNT7B转录,而外源WNT恢复类器官增殖。HER2扩增、KRAS扩增或WNT2拷贝数增加的患者源性类器官表现出豪猪抑制剂敏感生长,表明依赖于类器官的WNT分泌。对公共转录组数据集的分析进一步表明,KRAS-MAPK-WNT7B轴在包括肺癌在内的其他癌症类型中是保守的。结论胃肿瘤可通过获得kras - mapk - smad2 /3驱动的上皮细胞WNT分泌而绕过生态位依赖。通过抑制MAPK、阻断SMAD2/3或抑制WNT分泌来靶向这条轴,可能代表了胃癌和其他kras高恶性肿瘤的治疗脆弱性。
{"title":"Epithelial WNT secretion drives niche escape of developing gastric cancer.","authors":"Jaehun Lee,Soomin Kim,Youngchul Oh,Stephan R Jahn,Jihoon Kim,Yeongjun Kim,Tim Schmäche,Sang-Min Kim,Isaree Teriyapirom,Thomas Groß,Ohbin Kwon,Jungmin Kim,Somi Kim,Anne-Marlen Ada,Andrea Català-Bordes,Youngwon Cho,Jinho Kim,Amanda Andersson-Rolf,Sebastian R Merker,Joo Yeon Lim,Ji-Yeon Park,Thomas M Klompstra,Ki-Jun Yoon,Dae-Sik Lim,Ho-Seok Lee,Jong Kyoung Kim,Eunyoung Choi,James R Goldenring,Jae-Ho Cheong,Hyunki Kim,Daniel E Stange,Heetak Lee,Bon-Kyoung Koo,Ji-Hyun Lee","doi":"10.1186/s12943-025-02543-z","DOIUrl":"https://doi.org/10.1186/s12943-025-02543-z","url":null,"abstract":"BACKGROUNDWNT signaling plays a key role in maintaining the gastric epithelium and promoting tumorigenesis. However, how gastric tumors achieve WNT niche independence remains unclear, as mutations on APC or CTNNB1-common mechanisms of ligand-independent WNT activation in colorectal cancer-are infrequent in gastric cancer. Understanding how WNT self-sufficiency is acquired in the stomach is therefore critical.METHODSWe analyzed mouse gastric organoids harboring oncogenic KRASG12D with or without RNF43/ZNRF3 (RZ) or CDH1/TP53 (CP) mutations, along with corresponding in vivo mouse models. Niche independence was assessed through growth factor withdrawal, Porcupine and pathway-specific inhibitor treatments, and WNT rescue assays. We performed single-nucleus multiome sequencing (RNA + ATAC) to investigate transcriptional and chromatin dynamics. Findings from mouse models were validated using patient-derived gastric cancer organoids, and pan-cancer cell line datasets were analyzed to evaluate clinical and cross-tissue relevance.RESULTSGastric fibroblasts secreted canonical WNT2B to maintain the homeostatic gastric epithelium. Upon KRAS activation, epithelial cells were reprogrammed to secrete WNT ligands independently of additional mutations. Single-nucleus multiome analysis revealed that KRAS-driven MAPK signaling opened SMAD2/3-bound enhancers at the WNT7B locus, leading to the emergence of WNT7B-expressing subpopulations. Inhibition of SMAD2/3 phosphorylation suppressed both organoid growth and WNT7B transcription, whereas exogenous WNT restored organoid proliferation. Patient-derived organoids with HER2 amplification, KRAS amplification, or WNT2 copy-number gain exhibited Porcupine inhibitor-sensitive growth, indicating dependence on WNT secretion from the organoids. Analysis of public transcriptomic datasets further demonstrated that the KRAS-MAPK-WNT7B axis is conserved across other cancer types, including lung cancer.CONCLUSIONSGastric tumors can bypass niche dependence by acquiring KRAS-MAPK-SMAD2/3-driven epithelial WNT secretion. Targeting this axis-through MAPK inhibition, SMAD2/3 blockade, or suppression of WNT secretion-may represent a therapeutic vulnerability in gastric cancer and other KRAS-high malignancies.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"150 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145759964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
c-Rel drives pancreatic cancer metastasis through fibronectin-integrin signaling-induced isolation stress resistance and EMT. c-Rel通过纤维连接蛋白-整合素信号诱导的分离、应激抵抗和EMT驱动胰腺癌转移。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-15 DOI: 10.1186/s12943-025-02486-5
D Bakırdöğen, K Görgülü, J Xin, L Richter, S Alcalá, L Ruiz-Cañas, C Dai, K J Frank, N Wu, K N Diakopoulos, H Ozturk, D Demircioğlu, K Peschke, R Ranjan, F Fusco, J Martinez-Useros, M J Fernandez-Aceñero, N F Chhabra, J C López-Gil, J Ai, D A Ruess, E Kaya-Aksoy, F Schmidt, L Kohlmann, A Berninger, H Yangin, F Schicktanz, K Steiger, I E Demir, R M Schmid, M Reichert, M Adli, M Lesina, B Sainz, H Algül
{"title":"c-Rel drives pancreatic cancer metastasis through fibronectin-integrin signaling-induced isolation stress resistance and EMT.","authors":"D Bakırdöğen, K Görgülü, J Xin, L Richter, S Alcalá, L Ruiz-Cañas, C Dai, K J Frank, N Wu, K N Diakopoulos, H Ozturk, D Demircioğlu, K Peschke, R Ranjan, F Fusco, J Martinez-Useros, M J Fernandez-Aceñero, N F Chhabra, J C López-Gil, J Ai, D A Ruess, E Kaya-Aksoy, F Schmidt, L Kohlmann, A Berninger, H Yangin, F Schicktanz, K Steiger, I E Demir, R M Schmid, M Reichert, M Adli, M Lesina, B Sainz, H Algül","doi":"10.1186/s12943-025-02486-5","DOIUrl":"10.1186/s12943-025-02486-5","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":" ","pages":""},"PeriodicalIF":33.9,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145763358","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
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