首页 > 最新文献

Molecular Cancer最新文献

英文 中文
Tumor-associated macrophages promote chemoresistance to Paclitaxel via activating NOTCH2-JAG1 juxtacrine signaling. 肿瘤相关巨噬细胞通过激活NOTCH2-JAG1近胞嘧啶信号通路促进紫杉醇化疗耐药。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1186/s12943-025-02546-w
Fazhi Yu,Qin Zhou,Weiqiang Yu,Tong Zhou,Cheng Cao,Yijia Xie,Peng Zhang,Hanyuan Liu,Wei He,Aoxing Cheng,Xiaopeng Ma,Qingfa Wu,Qi Zhao,Jing Guo,Kaiguang Zhang,Ying Zhou,Jue Shi,Zhenye Yang
BACKGROUNDTaxane-based chemotherapy is a main treatment modality for ovarian cancer and other solid tumors, but chemoresistance limits the clinical efficacy. Studies have shown tumor interaction with macrophages in the tumor microenvironment (TME) plays a significant role in taxane resistance, yet the underlying molecular mechanisms are poorly understood.METHODSIn this study, we employed translatome profiling of paclitaxel-treated cancer cells, live-cell imaging analysis, gene knockdown/knockout, and in vitro cancer-macrophage coculture assays to unravel a novel chemoresistance mechanism mediated by tumor-macrophage interaction via the NOTCH2-JAG1 axis. The in vitro data were further validated by multiple xenograft, syngeneic and patient-derived xenograft mouse tumor models of ovarian cancer as well as ovarian cancer patient samples.RESULTSWe found paclitaxel selectively induced translational upregulation of NOTCH2 via cytoplasmic polyadenylation, and this NOTCH2 upregulation persisted after mitotic exit. Subsequent NOTCH2 activation by JAG1 expressed mainly on the neighboring macrophages promoted tumor cell survival and simulated cytokine release, such as CSF1 and IL-1β, that recruited JAG1-expressing macrophages, thus forming a positive feedback loop that further enhanced the pro-tumor NOTCH2 activity. Genetic depletion or pharmacological inhibition of NOTCH2 with the γ-secretase inhibitor attenuated macrophage infiltration and sensitized tumor response to paclitaxel in multiple preclinical models of ovarian cancer. Moreover, single-cell RNA sequencing analysis identified a JAG1-high macrophage subset that was enriched by paclitaxel treatment and attenuated by NOTCH inhibition. Clinically, high NOTCH2 expression in ovarian tumors was associated with recurrence and shorter progression-free survival of ovarian cancer patients.CONCLUSIONSPaclitaxel-induced translational upregulation of NOTCH2 enables immediate juxtacrine activation by JAG1-positive macrophages, coupling tumor cell survival with immune remodeling in the tumor microenvironment to drive chemoresistance. Our results suggest NOTCH2 is a viable biomarker for paclitaxel resistance and that combining NOTCH2 inhibitor with taxane is an effective therapeutic strategy to selectively disrupt tumor-macrophage interaction and overcome macrophage-mediated taxane resistance in NOTCH2-positive tumors.
紫杉烷为基础的化疗是卵巢癌和其他实体肿瘤的主要治疗方式,但化疗耐药限制了其临床疗效。研究表明肿瘤与肿瘤微环境中巨噬细胞的相互作用(TME)在紫杉烷耐药中起重要作用,但其潜在的分子机制尚不清楚。方法在本研究中,我们利用紫杉醇处理的癌细胞的翻译组分析、活细胞成像分析、基因敲除/敲除以及体外癌症-巨噬细胞共培养实验来揭示肿瘤-巨噬细胞通过NOTCH2-JAG1轴相互作用介导的一种新的化疗耐药机制。通过多种异种移植、同基因和患者来源的卵巢癌小鼠肿瘤模型以及卵巢癌患者样本进一步验证了体外数据。结果紫杉醇通过胞质多聚腺苷化选择性诱导NOTCH2的翻译上调,且NOTCH2的上调在有丝分裂结束后持续存在。随后,主要在邻近巨噬细胞上表达的JAG1激活NOTCH2,促进肿瘤细胞存活,模拟细胞因子如CSF1、IL-1β的释放,招募表达JAG1的巨噬细胞,形成正反馈循环,进一步增强NOTCH2的促瘤活性。在多种卵巢癌临床前模型中,基因缺失或γ-分泌酶抑制剂对NOTCH2的药理抑制可减轻巨噬细胞浸润,并使肿瘤对紫杉醇的反应增敏。此外,单细胞RNA测序分析发现了一个高jag1的巨噬细胞亚群,该亚群通过紫杉醇治疗而富集,并通过NOTCH抑制而减弱。临床上,NOTCH2在卵巢肿瘤中的高表达与卵巢癌患者的复发和较短的无进展生存期相关。结论紫杉醇诱导的NOTCH2翻译上调可使jag1阳性巨噬细胞立即激活近胞嘧啶,将肿瘤细胞存活与肿瘤微环境中的免疫重塑耦合在一起,从而驱动化疗耐药。我们的研究结果表明,NOTCH2是紫杉醇耐药的可行生物标志物,NOTCH2抑制剂与紫杉醇联合是一种有效的治疗策略,可以选择性地破坏肿瘤-巨噬细胞相互作用,克服巨噬细胞介导的紫杉醇耐药。
{"title":"Tumor-associated macrophages promote chemoresistance to Paclitaxel via activating NOTCH2-JAG1 juxtacrine signaling.","authors":"Fazhi Yu,Qin Zhou,Weiqiang Yu,Tong Zhou,Cheng Cao,Yijia Xie,Peng Zhang,Hanyuan Liu,Wei He,Aoxing Cheng,Xiaopeng Ma,Qingfa Wu,Qi Zhao,Jing Guo,Kaiguang Zhang,Ying Zhou,Jue Shi,Zhenye Yang","doi":"10.1186/s12943-025-02546-w","DOIUrl":"https://doi.org/10.1186/s12943-025-02546-w","url":null,"abstract":"BACKGROUNDTaxane-based chemotherapy is a main treatment modality for ovarian cancer and other solid tumors, but chemoresistance limits the clinical efficacy. Studies have shown tumor interaction with macrophages in the tumor microenvironment (TME) plays a significant role in taxane resistance, yet the underlying molecular mechanisms are poorly understood.METHODSIn this study, we employed translatome profiling of paclitaxel-treated cancer cells, live-cell imaging analysis, gene knockdown/knockout, and in vitro cancer-macrophage coculture assays to unravel a novel chemoresistance mechanism mediated by tumor-macrophage interaction via the NOTCH2-JAG1 axis. The in vitro data were further validated by multiple xenograft, syngeneic and patient-derived xenograft mouse tumor models of ovarian cancer as well as ovarian cancer patient samples.RESULTSWe found paclitaxel selectively induced translational upregulation of NOTCH2 via cytoplasmic polyadenylation, and this NOTCH2 upregulation persisted after mitotic exit. Subsequent NOTCH2 activation by JAG1 expressed mainly on the neighboring macrophages promoted tumor cell survival and simulated cytokine release, such as CSF1 and IL-1β, that recruited JAG1-expressing macrophages, thus forming a positive feedback loop that further enhanced the pro-tumor NOTCH2 activity. Genetic depletion or pharmacological inhibition of NOTCH2 with the γ-secretase inhibitor attenuated macrophage infiltration and sensitized tumor response to paclitaxel in multiple preclinical models of ovarian cancer. Moreover, single-cell RNA sequencing analysis identified a JAG1-high macrophage subset that was enriched by paclitaxel treatment and attenuated by NOTCH inhibition. Clinically, high NOTCH2 expression in ovarian tumors was associated with recurrence and shorter progression-free survival of ovarian cancer patients.CONCLUSIONSPaclitaxel-induced translational upregulation of NOTCH2 enables immediate juxtacrine activation by JAG1-positive macrophages, coupling tumor cell survival with immune remodeling in the tumor microenvironment to drive chemoresistance. Our results suggest NOTCH2 is a viable biomarker for paclitaxel resistance and that combining NOTCH2 inhibitor with taxane is an effective therapeutic strategy to selectively disrupt tumor-macrophage interaction and overcome macrophage-mediated taxane resistance in NOTCH2-positive tumors.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"46 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947418","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
Multi-omics profiling uncovers immune-molecular clusters with distinct chemo-immunotherapeutic vulnerabilities in a mouse model of triple-negative breast cancer. 多组学分析揭示了三阴性乳腺癌小鼠模型中具有不同化学免疫治疗脆弱性的免疫分子簇。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-10 DOI: 10.1186/s12943-025-02547-9
Olivier Castellanet,Jean Monatte,Nathan Corvaisier,Abdessamad El Kaoutari,Muge Kaya,Lorène Ferreira,Stephane Audebert,Luc Camoin,Alexandre de Nonneville,Anthony Gonçalves,Jean-Paul Borg,Paula Michea,Flavio Maina,Fabienne Lamballe
BACKGROUNDTriple-negative breast cancer (TNBC) is a highly aggressive and heterogeneous breast cancer subtype with limited treatment options. Predicting patient response to chemo-immunotherapy remains challenging, highlighting the need for robust stratification strategies.METHODSWe performed a multi-parametric analysis combining histological, genomic, transcriptomic, proteomic, and immune profiling in the immunocompetent MMTV-R26Met TNBC mouse model and compared outcomes with patient data from human TNBC cohorts and TNBC tumor microarray. To enable therapeutic testing and functional validation, we established syngeneic grafts from primary tumors and used them to evaluate combined chemotherapy (epirubicin) and anti-PD-1 immunotherapy.RESULTSMulti-parametric analysis of TNBC heterogeneity modeled by the MMTV-R26Met mice identified four distinct TNBC clusters, defined by unique intrinsic (molecular/genomic) and extrinsic (immune) features, which closely parallel patient subtypes, including rare metaplastic forms, and correlate with clinical outcomes. Both intrinsic and immune hallmarks of primary tumors were conserved across serial syngeneic transplantations, confirming the translational value of this preclinical platform. Treatment assessments indicated cluster-specific therapeutic vulnerabilities associated with molecular and immune traits. Specifically, whereas chemo-immunotherapy is beneficial to neutrophil-enriched tumors, immunotherapy alone appears to be more effective in macrophage-enriched tumors. Our findings indicate that TNBC treatment response is shaped by the interplay between tumor-intrinsic and immune features.CONCLUSIONOur study provides a robust preclinical platform for precision immuno-oncology, enabling stratification of TNBC patients for tailored onco-immunotherapies.
背景:三阴性乳腺癌(TNBC)是一种高度侵袭性和异质性的乳腺癌亚型,治疗选择有限。预测患者对化学免疫治疗的反应仍然具有挑战性,强调需要强有力的分层策略。方法我们对免疫功能良好的MMTV-R26Met TNBC小鼠模型进行了多参数分析,包括组织学、基因组学、转录组学、蛋白质组学和免疫谱分析,并将结果与人类TNBC队列和TNBC肿瘤微阵列的患者数据进行了比较。为了进行治疗测试和功能验证,我们建立了来自原发肿瘤的同基因移植物,并用它们来评估联合化疗(表柔比星)和抗pd -1免疫治疗。结果MMTV-R26Met小鼠模型对TNBC异质性的多参数分析确定了四种不同的TNBC集群,这些集群由独特的内在(分子/基因组)和外在(免疫)特征定义,它们与患者亚型密切相似,包括罕见的化生形式,并与临床结果相关。原发肿瘤的固有特征和免疫特征在连续的同基因移植中都是保守的,这证实了该临床前平台的转化价值。治疗评估显示与分子和免疫特性相关的簇特异性治疗脆弱性。具体来说,虽然化学免疫治疗对嗜中性粒细胞富集的肿瘤有益,但单独免疫治疗似乎对巨噬细胞富集的肿瘤更有效。我们的研究结果表明,TNBC治疗反应是由肿瘤固有特征和免疫特征之间的相互作用形成的。结论:我们的研究为精确的免疫肿瘤学提供了一个强大的临床前平台,使TNBC患者能够分层以进行量身定制的肿瘤免疫治疗。
{"title":"Multi-omics profiling uncovers immune-molecular clusters with distinct chemo-immunotherapeutic vulnerabilities in a mouse model of triple-negative breast cancer.","authors":"Olivier Castellanet,Jean Monatte,Nathan Corvaisier,Abdessamad El Kaoutari,Muge Kaya,Lorène Ferreira,Stephane Audebert,Luc Camoin,Alexandre de Nonneville,Anthony Gonçalves,Jean-Paul Borg,Paula Michea,Flavio Maina,Fabienne Lamballe","doi":"10.1186/s12943-025-02547-9","DOIUrl":"https://doi.org/10.1186/s12943-025-02547-9","url":null,"abstract":"BACKGROUNDTriple-negative breast cancer (TNBC) is a highly aggressive and heterogeneous breast cancer subtype with limited treatment options. Predicting patient response to chemo-immunotherapy remains challenging, highlighting the need for robust stratification strategies.METHODSWe performed a multi-parametric analysis combining histological, genomic, transcriptomic, proteomic, and immune profiling in the immunocompetent MMTV-R26Met TNBC mouse model and compared outcomes with patient data from human TNBC cohorts and TNBC tumor microarray. To enable therapeutic testing and functional validation, we established syngeneic grafts from primary tumors and used them to evaluate combined chemotherapy (epirubicin) and anti-PD-1 immunotherapy.RESULTSMulti-parametric analysis of TNBC heterogeneity modeled by the MMTV-R26Met mice identified four distinct TNBC clusters, defined by unique intrinsic (molecular/genomic) and extrinsic (immune) features, which closely parallel patient subtypes, including rare metaplastic forms, and correlate with clinical outcomes. Both intrinsic and immune hallmarks of primary tumors were conserved across serial syngeneic transplantations, confirming the translational value of this preclinical platform. Treatment assessments indicated cluster-specific therapeutic vulnerabilities associated with molecular and immune traits. Specifically, whereas chemo-immunotherapy is beneficial to neutrophil-enriched tumors, immunotherapy alone appears to be more effective in macrophage-enriched tumors. Our findings indicate that TNBC treatment response is shaped by the interplay between tumor-intrinsic and immune features.CONCLUSIONOur study provides a robust preclinical platform for precision immuno-oncology, enabling stratification of TNBC patients for tailored onco-immunotherapies.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"81 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947423","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
Unlocking the power of non-coding RNAs: toward real-time cancer monitoring in precision oncology. 解锁非编码rna的力量:在精确肿瘤学中实现实时癌症监测。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-09 DOI: 10.1186/s12943-025-02536-y
Manon Chang,Thomas Papazyan,Elvire Pons-Tostivint,Delphine Fradin
{"title":"Unlocking the power of non-coding RNAs: toward real-time cancer monitoring in precision oncology.","authors":"Manon Chang,Thomas Papazyan,Elvire Pons-Tostivint,Delphine Fradin","doi":"10.1186/s12943-025-02536-y","DOIUrl":"https://doi.org/10.1186/s12943-025-02536-y","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"19 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145937771","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
Metabolic characteristics in hepatocellular carcinoma: amino acid metabolic reprogramming. 肝细胞癌的代谢特征:氨基酸代谢重编程。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-08 DOI: 10.1186/s12943-025-02492-7
Ran Zhou, Yuejun Li, Guanghui Li, Yan Li, Lie Luo, Bin Wang, Liping Wang

Hepatocellular carcinoma (HCC) is a common type of primary liver cancer and is considered the third leading cause of cancer-related deaths worldwide. The high aggressiveness and resistance to therapies exhibited by HCC present significant challenges to global public health. As the primary metabolic organ in the human body, the liver undergoes substantial metabolic reprogramming during carcinogenesis, affecting various metabolic pathways including those involved in carbohydrates, lipids, and amino acids. Notably, disruptions in amino acid metabolism play a critical role in the initiation and progression of HCC, helping to sustain its malignant characteristics. This review aims to provide an in-depth analysis of the alterations observed in aromatic amino acids metabolism, branched chain amino acids (BCAAs) metabolism, glutamine metabolism, and other amino acid metabolism processes, including serine, arginine, and methionine, along with the expression patterns of associated metabolic enzymes. Furthermore, it discusses potential therapeutic approaches and their clinical relevance, offering insights and strategies for improving HCC diagnosis and treatment in the future.

肝细胞癌(HCC)是一种常见的原发性肝癌,被认为是全球癌症相关死亡的第三大原因。HCC表现出的高侵袭性和耐药性对全球公共卫生构成了重大挑战。肝脏作为人体的主要代谢器官,在癌变过程中经历了大量的代谢重编程,影响了碳水化合物、脂质和氨基酸等多种代谢途径。值得注意的是,氨基酸代谢的中断在HCC的发生和发展中起着关键作用,有助于维持其恶性特征。本文旨在深入分析芳香氨基酸代谢、支链氨基酸(BCAAs)代谢、谷氨酰胺代谢以及其他氨基酸代谢过程(包括丝氨酸、精氨酸和蛋氨酸)的变化以及相关代谢酶的表达模式。此外,它还讨论了潜在的治疗方法及其临床相关性,为未来改善HCC的诊断和治疗提供了见解和策略。
{"title":"Metabolic characteristics in hepatocellular carcinoma: amino acid metabolic reprogramming.","authors":"Ran Zhou, Yuejun Li, Guanghui Li, Yan Li, Lie Luo, Bin Wang, Liping Wang","doi":"10.1186/s12943-025-02492-7","DOIUrl":"https://doi.org/10.1186/s12943-025-02492-7","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) is a common type of primary liver cancer and is considered the third leading cause of cancer-related deaths worldwide. The high aggressiveness and resistance to therapies exhibited by HCC present significant challenges to global public health. As the primary metabolic organ in the human body, the liver undergoes substantial metabolic reprogramming during carcinogenesis, affecting various metabolic pathways including those involved in carbohydrates, lipids, and amino acids. Notably, disruptions in amino acid metabolism play a critical role in the initiation and progression of HCC, helping to sustain its malignant characteristics. This review aims to provide an in-depth analysis of the alterations observed in aromatic amino acids metabolism, branched chain amino acids (BCAAs) metabolism, glutamine metabolism, and other amino acid metabolism processes, including serine, arginine, and methionine, along with the expression patterns of associated metabolic enzymes. Furthermore, it discusses potential therapeutic approaches and their clinical relevance, offering insights and strategies for improving HCC diagnosis and treatment in the future.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":" ","pages":""},"PeriodicalIF":33.9,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918085","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
BRRIAR lncRNA alters breast cancer risk by modulating interferon signaling in cis and in trans. BRRIAR lncRNA通过调节顺式和反式干扰素信号改变乳腺癌风险。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1186/s12943-025-02510-8
Haran Sivakumaran, Sneha Nair, Mainá Bitar, Xue Lu, Lu Wang, Ji Liu, Deshapriya S Karunarathne, P Prakrithi, Sebastien Jacquelin, Isela Sarahi Rivera, Kristine M Hillman, Susanne Kaufmann, Rebekah Ziegman, Wei Shi, Sarah Alexandrou, C Elizabeth Caldon, Rakesh N Veedu, Quan H Nguyen, Jonathan Beesley, Michelle N Wykes, Juliet D French, Stacey L Edwards

Background: Interferons (IFNs) are key cytokines that drive immune responses against infections and cancer, yet few therapies have successfully leveraged IFN signaling for cancer treatment. Long noncoding RNAs (lncRNAs) are emerging as promising therapeutic candidates, but their roles in immune modulation remain largely unexplored. Here, we functionally characterize a breast cancer-associated lncRNA, BRRIAR, which primes the IFN signaling pathway in specific cancer contexts and represents a potential therapeutic strategy for estrogen receptor-positive (ER+) breast cancer.

Methods: BRRIAR expression and subcellular localization were examined using qPCR, in situ hybridization, single-cell RNA sequencing and spatial transcriptomics. BRRIAR target genes were identified through CRISPR interference, chromatin interaction assays and ChIP sequencing. Mechanistic studies in ER + breast cancer cells included CRISPR-Cas9 genome-wide screens, RNA sequencing, RNA pull-down followed by mass spectrometry, proliferation assays and Western blotting. The therapeutic potential of BRRIAR was evaluated via intratumoral delivery of lipid nanoparticle-encapsulated BRRIAR in ER + breast cancer xenograft models. Immune activation was assessed using flow cytometry and cytokine profiling of human peripheral blood mononuclear cells (PBMCs).

Results: We demonstrate that BRRIAR is a key target gene at the 3p26 breast cancer risk region. Primarily expressed in ER + breast tumors, BRRIAR acts both in cis and in trans. Nuclear BRRIAR regulates BHLHE40 expression in cis through chromatin interactions, while cytoplasmic BRRIAR binds in trans to the pattern recognition receptor RIG-I, priming IFN signaling. Overexpression of BRRIAR RNA triggers RIG-I signaling, inducing IFN responses, drives rapid, dose-dependent apoptosis of ER + breast cancer cells in vitro and in vivo, and promotes immune activation in human PBMCs.

Conclusions: These findings establish lncRNAs as key regulators of tumor immunity and uncover a critical link between genetic risk, lncRNAs, cancer immunosurveillance and breast cancer development, positioning BRRIAR as a promising lncRNA-based RIG-I activator for ER + breast cancer therapy.

背景:干扰素(IFN)是驱动免疫反应对抗感染和癌症的关键细胞因子,然而很少有治疗方法成功地利用IFN信号来治疗癌症。长链非编码rna (lncRNAs)作为一种有前景的治疗候选者正在出现,但它们在免疫调节中的作用在很大程度上仍未被探索。在这里,我们对乳腺癌相关的lncRNA BRRIAR进行了功能表征,BRRIAR在特定的癌症背景下启动IFN信号通路,并代表了雌激素受体阳性(ER+)乳腺癌的潜在治疗策略。方法:采用qPCR、原位杂交、单细胞RNA测序和空间转录组学检测BRRIAR的表达和亚细胞定位。通过CRISPR干扰、染色质相互作用和ChIP测序鉴定BRRIAR靶基因。ER +乳腺癌细胞的机制研究包括CRISPR-Cas9全基因组筛选、RNA测序、RNA下拉、质谱分析、增殖试验和Western blotting。BRRIAR的治疗潜力是通过在ER +乳腺癌异种移植模型中给药脂质纳米颗粒包裹的BRRIAR来评估的。利用流式细胞术和人外周血单个核细胞(PBMCs)的细胞因子谱来评估免疫激活。结果:我们证明BRRIAR是3p26乳腺癌危险区域的关键靶基因。BRRIAR主要在ER +乳腺肿瘤中表达,并以顺式和反式两种方式起作用。核BRRIAR通过染色质相互作用顺式调节BHLHE40的表达,而细胞质BRRIAR通过反式结合模式识别受体RIG-I,启动IFN信号传导。BRRIAR RNA的过表达触发RIG-I信号,诱导IFN反应,在体外和体内驱动ER +乳腺癌细胞的快速、剂量依赖性凋亡,并促进人PBMCs的免疫激活。结论:这些发现确立了lncrna是肿瘤免疫的关键调控因子,揭示了遗传风险、lncrna、癌症免疫监测和乳腺癌发展之间的关键联系,将BRRIAR定位为一种有前途的基于lncrna的RIG-I激活剂,用于ER +乳腺癌治疗。
{"title":"BRRIAR lncRNA alters breast cancer risk by modulating interferon signaling in cis and in trans.","authors":"Haran Sivakumaran, Sneha Nair, Mainá Bitar, Xue Lu, Lu Wang, Ji Liu, Deshapriya S Karunarathne, P Prakrithi, Sebastien Jacquelin, Isela Sarahi Rivera, Kristine M Hillman, Susanne Kaufmann, Rebekah Ziegman, Wei Shi, Sarah Alexandrou, C Elizabeth Caldon, Rakesh N Veedu, Quan H Nguyen, Jonathan Beesley, Michelle N Wykes, Juliet D French, Stacey L Edwards","doi":"10.1186/s12943-025-02510-8","DOIUrl":"10.1186/s12943-025-02510-8","url":null,"abstract":"<p><strong>Background: </strong>Interferons (IFNs) are key cytokines that drive immune responses against infections and cancer, yet few therapies have successfully leveraged IFN signaling for cancer treatment. Long noncoding RNAs (lncRNAs) are emerging as promising therapeutic candidates, but their roles in immune modulation remain largely unexplored. Here, we functionally characterize a breast cancer-associated lncRNA, BRRIAR, which primes the IFN signaling pathway in specific cancer contexts and represents a potential therapeutic strategy for estrogen receptor-positive (ER+) breast cancer.</p><p><strong>Methods: </strong>BRRIAR expression and subcellular localization were examined using qPCR, in situ hybridization, single-cell RNA sequencing and spatial transcriptomics. BRRIAR target genes were identified through CRISPR interference, chromatin interaction assays and ChIP sequencing. Mechanistic studies in ER + breast cancer cells included CRISPR-Cas9 genome-wide screens, RNA sequencing, RNA pull-down followed by mass spectrometry, proliferation assays and Western blotting. The therapeutic potential of BRRIAR was evaluated via intratumoral delivery of lipid nanoparticle-encapsulated BRRIAR in ER + breast cancer xenograft models. Immune activation was assessed using flow cytometry and cytokine profiling of human peripheral blood mononuclear cells (PBMCs).</p><p><strong>Results: </strong>We demonstrate that BRRIAR is a key target gene at the 3p26 breast cancer risk region. Primarily expressed in ER + breast tumors, BRRIAR acts both in cis and in trans. Nuclear BRRIAR regulates BHLHE40 expression in cis through chromatin interactions, while cytoplasmic BRRIAR binds in trans to the pattern recognition receptor RIG-I, priming IFN signaling. Overexpression of BRRIAR RNA triggers RIG-I signaling, inducing IFN responses, drives rapid, dose-dependent apoptosis of ER + breast cancer cells in vitro and in vivo, and promotes immune activation in human PBMCs.</p><p><strong>Conclusions: </strong>These findings establish lncRNAs as key regulators of tumor immunity and uncover a critical link between genetic risk, lncRNAs, cancer immunosurveillance and breast cancer development, positioning BRRIAR as a promising lncRNA-based RIG-I activator for ER + breast cancer therapy.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"25 1","pages":"5"},"PeriodicalIF":33.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12777463/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918137","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
HDAC6 orchestrates metastatic and immunosuppressive programs in small cell lung cancer through S100A2-TGF-β/SMAD and CSF1R signaling. HDAC6通过S100A2-TGF-β/SMAD和CSF1R信号调控小细胞肺癌的转移和免疫抑制程序。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1186/s12943-025-02552-y
Yantao Jiang, Junjie Yu, Ting Wang, Qingwu Du, Jingya Wang, Yi Lu, Qi Xu, Huiyan Liu, Xueyang Li, Luyao Tong, Tingting Qin, Dingzhi Huang
{"title":"HDAC6 orchestrates metastatic and immunosuppressive programs in small cell lung cancer through S100A2-TGF-β/SMAD and CSF1R signaling.","authors":"Yantao Jiang, Junjie Yu, Ting Wang, Qingwu Du, Jingya Wang, Yi Lu, Qi Xu, Huiyan Liu, Xueyang Li, Luyao Tong, Tingting Qin, Dingzhi Huang","doi":"10.1186/s12943-025-02552-y","DOIUrl":"https://doi.org/10.1186/s12943-025-02552-y","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":" ","pages":""},"PeriodicalIF":33.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918070","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
Retraction Note: Circular RNA circDCUN1D4 suppresses hepatocellular carcinoma development via targeting the miR-590-5p/ TIMP3 axis. 注:环状RNA circDCUN1D4通过靶向miR-590-5p/ TIMP3轴抑制肝细胞癌的发展。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-06 DOI: 10.1186/s12943-025-02550-0
Hongyu Li, Bing Su, Yan Jiang, Boyang Zhang, Rulong Du, Can Song, Bin Hou, Kun Xu, Lida Wu, Yuchun Gu
{"title":"Retraction Note: Circular RNA circDCUN1D4 suppresses hepatocellular carcinoma development via targeting the miR-590-5p/ TIMP3 axis.","authors":"Hongyu Li, Bing Su, Yan Jiang, Boyang Zhang, Rulong Du, Can Song, Bin Hou, Kun Xu, Lida Wu, Yuchun Gu","doi":"10.1186/s12943-025-02550-0","DOIUrl":"10.1186/s12943-025-02550-0","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"25 1","pages":"4"},"PeriodicalIF":33.9,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12771894/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145912459","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
The puppet master in the breast cancer "microecological community": spatial and metabolic regulation of macrophage heterogeneity. 乳腺癌“微生态群落”中的木偶大师:巨噬细胞异质性的空间与代谢调控。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-06 DOI: 10.1186/s12943-025-02551-z
Hao Wu,Hong-Da Tian,Liuying Zhao,Dandan Liu,Baohang Lin,Xiaohong Wu
{"title":"The puppet master in the breast cancer \"microecological community\": spatial and metabolic regulation of macrophage heterogeneity.","authors":"Hao Wu,Hong-Da Tian,Liuying Zhao,Dandan Liu,Baohang Lin,Xiaohong Wu","doi":"10.1186/s12943-025-02551-z","DOIUrl":"https://doi.org/10.1186/s12943-025-02551-z","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"30 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903669","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
Lactate: elucidating its indispensable role in human health. 乳酸:阐明其在人体健康中不可或缺的作用。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-05 DOI: 10.1186/s12943-025-02519-z
Shengxin Zhang,Jie Wang,Ziyu Xu,Ziyang Cheng,Bin Shao,Jiayun Yu
Lactate, traditionally considered just a byproduct of metabolism, is now understood to be a vital regulator in energy metabolism, immune function, and epigenetic changes. Besides serving as an alternative energy source through the "lactate shuttle," it acts as a signaling molecule influencing both normal and abnormal processes in various organs. New research has emphasized its role in lactylation of histones and non-histones, a novel post-translational modification linking metabolic activity with gene expression and immune response. Lactate contributes to immunosuppression, angiogenesis, and the spread of tumors within the tumor microenvironment. Its accumulation is also linked to cardiovascular, metabolic, and neurodegenerative conditions. This shift in metabolism underscores lactate's growing importance in both health and disease, presenting novel therapeutic opportunities, especially in the treatment of cancer and metabolic disorders. This review synthesizes emerging insights into lactate's multifaceted roles and discusses promising therapeutic strategies targeting lactate metabolism, transport, and downstream signaling pathways, with an emphasis on candidates advancing toward clinical translation.
乳酸,传统上被认为只是代谢的副产物,现在被认为是能量代谢、免疫功能和表观遗传变化的重要调节因子。除了通过“乳酸穿梭”作为替代能源外,它还作为影响各种器官正常和异常过程的信号分子。新的研究强调了它在组蛋白和非组蛋白的乳酸化中的作用,这是一种新的翻译后修饰,将代谢活性与基因表达和免疫反应联系起来。乳酸有助于免疫抑制、血管生成和肿瘤微环境内肿瘤的扩散。它的积累也与心血管、代谢和神经退行性疾病有关。这种代谢的转变强调了乳酸在健康和疾病中日益重要的作用,提供了新的治疗机会,特别是在治疗癌症和代谢紊乱方面。这篇综述综合了对乳酸的多方面作用的新见解,并讨论了针对乳酸代谢、转运和下游信号通路的有前途的治疗策略,重点是向临床转化的候选药物。
{"title":"Lactate: elucidating its indispensable role in human health.","authors":"Shengxin Zhang,Jie Wang,Ziyu Xu,Ziyang Cheng,Bin Shao,Jiayun Yu","doi":"10.1186/s12943-025-02519-z","DOIUrl":"https://doi.org/10.1186/s12943-025-02519-z","url":null,"abstract":"Lactate, traditionally considered just a byproduct of metabolism, is now understood to be a vital regulator in energy metabolism, immune function, and epigenetic changes. Besides serving as an alternative energy source through the \"lactate shuttle,\" it acts as a signaling molecule influencing both normal and abnormal processes in various organs. New research has emphasized its role in lactylation of histones and non-histones, a novel post-translational modification linking metabolic activity with gene expression and immune response. Lactate contributes to immunosuppression, angiogenesis, and the spread of tumors within the tumor microenvironment. Its accumulation is also linked to cardiovascular, metabolic, and neurodegenerative conditions. This shift in metabolism underscores lactate's growing importance in both health and disease, presenting novel therapeutic opportunities, especially in the treatment of cancer and metabolic disorders. This review synthesizes emerging insights into lactate's multifaceted roles and discusses promising therapeutic strategies targeting lactate metabolism, transport, and downstream signaling pathways, with an emphasis on candidates advancing toward clinical translation.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"22 1","pages":"2"},"PeriodicalIF":37.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903672","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
EIF4A3/circPTGR1/miR-4725-5p positive- feedback loop promotes colorectal cancer progression via FAK/AKT signaling pathway. EIF4A3/circPTGR1/miR-4725-5p正反馈环通过FAK/AKT信号通路促进结直肠癌进展。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-05 DOI: 10.1186/s12943-025-02537-x
Yi Dong,Yi-Han Ding,Xiao Yang,Xiao-Hang Song,Xue-Tao Lei,Ya-Nan Tang,Xin-Er Zhang,Wei-Cai Huang,Kai Zhang,Jing-Song Chen
{"title":"EIF4A3/circPTGR1/miR-4725-5p positive- feedback loop promotes colorectal cancer progression via FAK/AKT signaling pathway.","authors":"Yi Dong,Yi-Han Ding,Xiao Yang,Xiao-Hang Song,Xue-Tao Lei,Ya-Nan Tang,Xin-Er Zhang,Wei-Cai Huang,Kai Zhang,Jing-Song Chen","doi":"10.1186/s12943-025-02537-x","DOIUrl":"https://doi.org/10.1186/s12943-025-02537-x","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"22 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903673","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