首页 > 最新文献

MedComm – Oncology最新文献

英文 中文
Decreased M6A Modification Promotes Progression of Gastrointestinal Stromal Tumor and KIT-Derived Imatinib Resistance Through FTO-Regulated Axis 减少M6A修饰通过fto调节轴促进胃肠道间质肿瘤进展和kit衍生的伊马替尼耐药
IF 2.2 Pub Date : 2025-12-04 DOI: 10.1002/mog2.70047
Jiehan Li, Ge Zhang, Yizheng Zhang, Zhao Sun, Wunan Mi, Meimei Jiang, Guiyun Jia, Nannan Liu, Lingling Zhang, Zhenqiang Sun, Jianwu Jiang, Yingjie Zhang, Yang Fu

Imatinib (IM) is the first-line therapy for high-risk gastrointestinal stromal tumor (GIST) patients; however, over 50% of those with advanced stage or metastasis develop IM resistance within 2 years, and effective strategies to overcome this resistance remain elusive. In this study, we identified that decreased N6-methyladenosine (m6A) modification by the demethylase FTO regulated GIST progression and IM resistance. Long noncoding RNA XIST (XIST) was identified as the main demethylated RNA by FTO in GIST. FTO leaded to a decrease in m6A modification at the 10517-10633 site of XIST, thereby protecting it from degradation mediated by YTHDF2's recognition and binding. Stabilized XIST enhanced IM resistance by acting as a posttranscriptional regulator of KIT, the primary oncogenic driver in GIST. In vitro and in vivo functional assays confirmed the roles of both FTO and XIST in promoting GIST progression and IM resistance. Importantly, pharmacological inhibition of FTO using FB23-2 effectively restored IM sensitivity in murine xenograft models of GIST. Together, our findings establish a mechanistic link among FTO-mediated m6A demethylation, XIST stabilization, and posttranscriptional regulation of KIT in GIST. These insights highlight the therapeutic potential of targeting m6A-FTO axis to overcome IM resistance in GIST treatment.

伊马替尼(IM)是高危胃肠道间质瘤(GIST)患者的一线治疗药物;然而,超过50%的晚期或转移患者在2年内出现IM耐药,而克服这种耐药的有效策略仍然难以捉摸。在这项研究中,我们发现去甲基化酶FTO对n6 -甲基腺苷(m6A)修饰的减少调节GIST的进展和IM耐药性。长链非编码RNA XIST (XIST)被FTO鉴定为GIST的主要去甲基化RNA。FTO导致XIST 10517-10633位点的m6A修饰减少,从而保护XIST免受YTHDF2识别和结合介导的降解。稳定的XIST通过作为KIT的转录后调节剂增强了IM耐药性,KIT是GIST的主要致癌驱动因子。体外和体内功能分析证实了FTO和XIST在促进GIST进展和IM耐药中的作用。重要的是,FB23-2对FTO的药理学抑制有效地恢复了小鼠GIST异种移植模型的IM敏感性。总之,我们的研究结果建立了fto介导的m6A去甲基化、XIST稳定和GIST中KIT转录后调控之间的机制联系。这些发现突出了靶向m6A-FTO轴在GIST治疗中克服IM耐药的治疗潜力。
{"title":"Decreased M6A Modification Promotes Progression of Gastrointestinal Stromal Tumor and KIT-Derived Imatinib Resistance Through FTO-Regulated Axis","authors":"Jiehan Li,&nbsp;Ge Zhang,&nbsp;Yizheng Zhang,&nbsp;Zhao Sun,&nbsp;Wunan Mi,&nbsp;Meimei Jiang,&nbsp;Guiyun Jia,&nbsp;Nannan Liu,&nbsp;Lingling Zhang,&nbsp;Zhenqiang Sun,&nbsp;Jianwu Jiang,&nbsp;Yingjie Zhang,&nbsp;Yang Fu","doi":"10.1002/mog2.70047","DOIUrl":"https://doi.org/10.1002/mog2.70047","url":null,"abstract":"<p>Imatinib (IM) is the first-line therapy for high-risk gastrointestinal stromal tumor (GIST) patients; however, over 50% of those with advanced stage or metastasis develop IM resistance within 2 years, and effective strategies to overcome this resistance remain elusive. In this study, we identified that decreased <i>N</i>6-methyladenosine (m6A) modification by the demethylase FTO regulated GIST progression and IM resistance. Long noncoding RNA XIST (XIST) was identified as the main demethylated RNA by FTO in GIST. FTO leaded to a decrease in m6A modification at the 10517-10633 site of XIST, thereby protecting it from degradation mediated by YTHDF2's recognition and binding. Stabilized XIST enhanced IM resistance by acting as a posttranscriptional regulator of KIT, the primary oncogenic driver in GIST. In vitro and in vivo functional assays confirmed the roles of both FTO and XIST in promoting GIST progression and IM resistance. Importantly, pharmacological inhibition of FTO using FB23-2 effectively restored IM sensitivity in murine xenograft models of GIST. Together, our findings establish a mechanistic link among FTO-mediated m6A demethylation, XIST stabilization, and posttranscriptional regulation of KIT in GIST. These insights highlight the therapeutic potential of targeting m6A-FTO axis to overcome IM resistance in GIST treatment.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 4","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70047","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145686174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LncRNA SNHG5/miR-363-3p/USP28 Axis Promotes Lung Adenocarcinoma Progression Through Enhancing β-Catenin Stabilization LncRNA SNHG5/miR-363-3p/USP28轴通过增强β-Catenin稳定性促进肺腺癌进展
IF 2.2 Pub Date : 2025-11-24 DOI: 10.1002/mog2.70046
Jia-yu Zou, Wen-min Zhou, Xue-bing He, Xing-tao Wu, Fang-de Li, Xiao-ci Luo, Xin Wang, Hong Bi, Li-ping Zhao, Xin-zhu Chen, Ming-na Sun, Yan-yan Yan, Jian-ye Zhang

Lung adenocarcinoma (LUAD) is the most common histological subtype of lung cancer, accounting for approximately 50% of global lung cancer-related mortality, which underscores the urgent need for identifying novel biomarkers and therapeutic targets. Long noncoding RNAs (lncRNAs) have been increasingly recognized as pivotal regulators in cancer development; however, the specific function of lncRNA small nucleolar RNA host gene 5 (SNHG5) in LUAD remained unclear. This study aimed to investigate the clinical significance, biological roles, and molecular mechanisms of SNHG5 in LUAD pathogenesis. Through integrated bioinformatics analysis and experimental validation, we found that SNHG5 was significantly upregulated in LUAD tissues and cell lines. Functional in vitro and in vivo assays—including gain-/loss-of-function studies, luciferase reporter assays, RNA immunoprecipitation, co-immunoprecipitation, and tumor xenograft models—demonstrated that SNHG5 promoted malignant phenotypes by acting as a competing endogenous RNA (ceRNA) for miR-363-3p. This sponge activity elevated the expression of ubiquitin-specific peptidase 28 (USP28), which in turn stabilized β-catenin and activated oncogenic signaling. Rescue experiments confirmed the functional importance of the SNHG5/miR-363-3p/USP28/β-catenin axis. In conclusion, these results indicate that SNHG5 drives LUAD progression through a novel ceRNA mechanism, highlighting its potential as both a prognostic biomarker and a therapeutic target.

肺腺癌(LUAD)是肺癌最常见的组织学亚型,约占全球肺癌相关死亡率的50%,这凸显了鉴定新的生物标志物和治疗靶点的迫切需要。长链非编码rna (lncRNAs)越来越被认为是癌症发展的关键调控因子;然而,lncRNA小核果RNA宿主基因5 (SNHG5)在LUAD中的具体功能尚不清楚。本研究旨在探讨SNHG5在LUAD发病中的临床意义、生物学作用及分子机制。通过综合生物信息学分析和实验验证,我们发现SNHG5在LUAD组织和细胞系中显著上调。体外和体内功能分析——包括功能获得/丧失研究、荧光素酶报告基因测定、RNA免疫沉淀、共免疫沉淀和肿瘤异种移植模型——表明,SNHG5通过作为miR-363-3p的竞争内源性RNA (ceRNA)促进恶性表型。这种海绵活性提高了泛素特异性肽酶28 (USP28)的表达,从而稳定了β-连环蛋白并激活了致癌信号传导。救援实验证实了SNHG5/miR-363-3p/USP28/β-catenin轴的功能重要性。总之,这些结果表明SNHG5通过一种新的ceRNA机制驱动LUAD的进展,突出了其作为预后生物标志物和治疗靶点的潜力。
{"title":"LncRNA SNHG5/miR-363-3p/USP28 Axis Promotes Lung Adenocarcinoma Progression Through Enhancing β-Catenin Stabilization","authors":"Jia-yu Zou,&nbsp;Wen-min Zhou,&nbsp;Xue-bing He,&nbsp;Xing-tao Wu,&nbsp;Fang-de Li,&nbsp;Xiao-ci Luo,&nbsp;Xin Wang,&nbsp;Hong Bi,&nbsp;Li-ping Zhao,&nbsp;Xin-zhu Chen,&nbsp;Ming-na Sun,&nbsp;Yan-yan Yan,&nbsp;Jian-ye Zhang","doi":"10.1002/mog2.70046","DOIUrl":"https://doi.org/10.1002/mog2.70046","url":null,"abstract":"<p>Lung adenocarcinoma (LUAD) is the most common histological subtype of lung cancer, accounting for approximately 50% of global lung cancer-related mortality, which underscores the urgent need for identifying novel biomarkers and therapeutic targets. Long noncoding RNAs (lncRNAs) have been increasingly recognized as pivotal regulators in cancer development; however, the specific function of lncRNA small nucleolar RNA host gene 5 (SNHG5) in LUAD remained unclear. This study aimed to investigate the clinical significance, biological roles, and molecular mechanisms of SNHG5 in LUAD pathogenesis. Through integrated bioinformatics analysis and experimental validation, we found that SNHG5 was significantly upregulated in LUAD tissues and cell lines. Functional in vitro and in vivo assays—including gain-/loss-of-function studies, luciferase reporter assays, RNA immunoprecipitation, co-immunoprecipitation, and tumor xenograft models—demonstrated that SNHG5 promoted malignant phenotypes by acting as a competing endogenous RNA (ceRNA) for miR-363-3p. This sponge activity elevated the expression of ubiquitin-specific peptidase 28 (USP28), which in turn stabilized β-catenin and activated oncogenic signaling. Rescue experiments confirmed the functional importance of the SNHG5/miR-363-3p/USP28/β-catenin axis. In conclusion, these results indicate that SNHG5 drives LUAD progression through a novel ceRNA mechanism, highlighting its potential as both a prognostic biomarker and a therapeutic target.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 4","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70046","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145618846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting Metabolic Reprogramming in Tumor: From Mechanisms to Precision Immunotherapies 肿瘤中靶向代谢重编程:从机制到精确免疫治疗
IF 2.2 Pub Date : 2025-11-14 DOI: 10.1002/mog2.70045
Tong Zhang, Ping Gao, Linchong Sun

Metabolic reprogramming is a core hallmark of malignant tumors. It facilitates the rapid growth of tumor cells and significantly modulates antitumour immune responses through metabolic interactions, affecting the success of immunotherapy. Despite recent breakthroughs in immunotherapy, most patients exhibit limited responses, and the underlying mechanisms are closely related to metabolic dysregulation within the tumor immune microenvironment. However, a comprehensive review of how to systematically leverage metabolic interventions to enhance immunotherapy efficacy is lacking. This review examines the competitive interactions between tumor and immune cells within essential metabolic pathways, including those involving glucose, amino acids, lipids, and nucleotides. This metabolic stress leads to the functional exhaustion of effector immune cells and activation of immunosuppressive cells, thereby promoting immune escape. Based on these mechanisms, we further summarize therapeutic strategies that target key metabolic enzymes to reshape the immune microenvironment and discuss their integration with strategies and clinical advances such as immune checkpoint blockade or CAR-T cell therapy. This review systematically integrates the core mechanisms and cutting-edge strategies at the intersection of metabolism and immunity, provides a theoretical framework and methodological reference for basic research and clinical translation in this area, and offers a theoretical basis and translational perspective for the development of synergistic metabolic–immunological therapeutic strategies.

代谢重编程是恶性肿瘤的核心特征。它促进肿瘤细胞的快速生长,并通过代谢相互作用显著调节抗肿瘤免疫反应,影响免疫治疗的成功。尽管最近在免疫治疗方面取得了突破,但大多数患者表现出有限的反应,其潜在机制与肿瘤免疫微环境中的代谢失调密切相关。然而,关于如何系统地利用代谢干预来增强免疫治疗效果的全面综述是缺乏的。本文综述了肿瘤和免疫细胞在基本代谢途径中的竞争性相互作用,包括涉及葡萄糖、氨基酸、脂质和核苷酸的代谢途径。这种代谢应激导致效应免疫细胞的功能衰竭和免疫抑制细胞的激活,从而促进免疫逃逸。基于这些机制,我们进一步总结了针对关键代谢酶重塑免疫微环境的治疗策略,并讨论了它们与免疫检查点阻断或CAR-T细胞治疗等策略和临床进展的整合。本综述系统整合了代谢与免疫交叉领域的核心机制和前沿策略,为该领域的基础研究和临床转化提供理论框架和方法参考,为代谢-免疫协同治疗策略的发展提供理论基础和转化视角。
{"title":"Targeting Metabolic Reprogramming in Tumor: From Mechanisms to Precision Immunotherapies","authors":"Tong Zhang,&nbsp;Ping Gao,&nbsp;Linchong Sun","doi":"10.1002/mog2.70045","DOIUrl":"https://doi.org/10.1002/mog2.70045","url":null,"abstract":"<p>Metabolic reprogramming is a core hallmark of malignant tumors. It facilitates the rapid growth of tumor cells and significantly modulates antitumour immune responses through metabolic interactions, affecting the success of immunotherapy. Despite recent breakthroughs in immunotherapy, most patients exhibit limited responses, and the underlying mechanisms are closely related to metabolic dysregulation within the tumor immune microenvironment. However, a comprehensive review of how to systematically leverage metabolic interventions to enhance immunotherapy efficacy is lacking. This review examines the competitive interactions between tumor and immune cells within essential metabolic pathways, including those involving glucose, amino acids, lipids, and nucleotides. This metabolic stress leads to the functional exhaustion of effector immune cells and activation of immunosuppressive cells, thereby promoting immune escape. Based on these mechanisms, we further summarize therapeutic strategies that target key metabolic enzymes to reshape the immune microenvironment and discuss their integration with strategies and clinical advances such as immune checkpoint blockade or CAR-T cell therapy. This review systematically integrates the core mechanisms and cutting-edge strategies at the intersection of metabolism and immunity, provides a theoretical framework and methodological reference for basic research and clinical translation in this area, and offers a theoretical basis and translational perspective for the development of synergistic metabolic–immunological therapeutic strategies.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 4","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70045","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145522101","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Parkinson's Disease and Cancer: Mechanistic Insights and Therapeutic Opportunities From Cancer Neuroscience 帕金森氏病和癌症:来自癌症神经科学的机制见解和治疗机会
IF 2.2 Pub Date : 2025-11-04 DOI: 10.1002/mog2.70044
Tianli Pan, Lin Yuan, Ruoyang Zhao, Liting Lu, Yinghao Zhi, Yuying Li, Min Wu, Yongye Huang

There is a complex pathological association between neurodegenerative diseases and cancer. The epidemiological negative correlation between Parkinson's disease (PD) and brain tumor is particularly noteworthy. PD is characterized by the loss of dopaminergic neurons and the formation of Lewy bodies, while glioma, the representative of brain tumors, originates from the malignant transformation of glial cells. The molecular interaction network between these two diseases is elusive, limiting the development of cross-disease treatment strategies. This review systematically summarizes the associations between PD and glioma in genetic predispositions, epigenetic modifications, alterations in subcellular compartments, and cellular mechanisms concerning neurons, glial cells, and stem cells. Additional links arise from circadian rhythm regulation, oxidative stress, and gut microbiota, underscoring the importance of systemic pathways that connect neurodegeneration and tumorigenesis. Within this context, cancer neuroscience emerges as a critical framework, demonstrating how neuronal activity drives cancer progression by shaping the tumor microenvironment. Therapeutic opportunities build upon these mechanistic insights, including engineering neuron types to suppress cancer growth, modulating synaptic genes, inducing neuronal cell death cascades, and controlling inflammation to disrupt tumor-nerve crosstalk. Emerging neuroscience-inspired technologies may drastically expand the treatment landscape. This review tries to unveil a potential theoretical paradigm for developing precise therapies with both neuroprotection and antitumor effects.

神经退行性疾病与癌症之间存在复杂的病理联系。帕金森病(PD)与脑肿瘤之间的流行病学负相关尤其值得注意。PD以多巴胺能神经元的丧失和路易小体的形成为特征,而脑肿瘤的代表胶质瘤则起源于胶质细胞的恶性转化。这两种疾病之间的分子相互作用网络是难以捉摸的,限制了跨疾病治疗策略的发展。本文系统总结了PD和胶质瘤在遗传易感性、表观遗传修饰、亚细胞区室改变以及神经元、胶质细胞和干细胞的细胞机制方面的联系。其他联系来自昼夜节律调节、氧化应激和肠道微生物群,强调了连接神经变性和肿瘤发生的系统性途径的重要性。在此背景下,癌症神经科学作为一个关键框架出现,展示了神经元活动如何通过塑造肿瘤微环境来驱动癌症进展。治疗机会建立在这些机制的基础上,包括设计神经元类型以抑制癌症生长,调节突触基因,诱导神经元细胞死亡级联,控制炎症以破坏肿瘤-神经串扰。受神经科学启发的新兴技术可能会极大地扩展治疗领域。这篇综述试图揭示一种潜在的理论范式,用于开发具有神经保护和抗肿瘤作用的精确疗法。
{"title":"Parkinson's Disease and Cancer: Mechanistic Insights and Therapeutic Opportunities From Cancer Neuroscience","authors":"Tianli Pan,&nbsp;Lin Yuan,&nbsp;Ruoyang Zhao,&nbsp;Liting Lu,&nbsp;Yinghao Zhi,&nbsp;Yuying Li,&nbsp;Min Wu,&nbsp;Yongye Huang","doi":"10.1002/mog2.70044","DOIUrl":"https://doi.org/10.1002/mog2.70044","url":null,"abstract":"<p>There is a complex pathological association between neurodegenerative diseases and cancer. The epidemiological negative correlation between Parkinson's disease (PD) and brain tumor is particularly noteworthy. PD is characterized by the loss of dopaminergic neurons and the formation of Lewy bodies, while glioma, the representative of brain tumors, originates from the malignant transformation of glial cells. The molecular interaction network between these two diseases is elusive, limiting the development of cross-disease treatment strategies. This review systematically summarizes the associations between PD and glioma in genetic predispositions, epigenetic modifications, alterations in subcellular compartments, and cellular mechanisms concerning neurons, glial cells, and stem cells. Additional links arise from circadian rhythm regulation, oxidative stress, and gut microbiota, underscoring the importance of systemic pathways that connect neurodegeneration and tumorigenesis. Within this context, cancer neuroscience emerges as a critical framework, demonstrating how neuronal activity drives cancer progression by shaping the tumor microenvironment. Therapeutic opportunities build upon these mechanistic insights, including engineering neuron types to suppress cancer growth, modulating synaptic genes, inducing neuronal cell death cascades, and controlling inflammation to disrupt tumor-nerve crosstalk. Emerging neuroscience-inspired technologies may drastically expand the treatment landscape. This review tries to unveil a potential theoretical paradigm for developing precise therapies with both neuroprotection and antitumor effects.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 4","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70044","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145469448","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Breast Cancer Brain Metastasis: Bridging Biological Mechanisms to Therapeutic Innovations 乳腺癌脑转移:连接生物学机制与治疗创新
IF 2.2 Pub Date : 2025-10-24 DOI: 10.1002/mog2.70043
Jing Feng, Yuwei Tao, Fengkai Li, Suling Liu

Breast cancer brain metastasis (BCBrM) remains a major clinical challenge with limited therapeutic options and poor prognosis. Despite advances in systemic therapy, the incidence of BCBrM is rising due to prolonged survival of patients with advanced breast cancer, yet effective brain-targeted strategies remain scarce, underscoring a critical research gap. This review integrates recent mechanistic insights that illuminate the complex biology underpinning BCBrM and explores how these discoveries are driving therapeutic innovation. We detail the metastatic cascade from local invasion to brain colonization, and examine key signaling pathways orchestrating brain-specific metastasis. Emphasis is placed on the dynamic crosstalk between tumor cells and the brain microenvironment, including astrocytes, microglia, and neurons, as well as metabolic reprogramming and immune evasion. We critically evaluate current preclinical models and their translational relevance, highlighting recent advances in humanized and imaging-based systems. Emerging therapies, such as central nervous system-penetrant kinase inhibitors, antibody–drug conjugates, and immunotherapies, are discussed alongside persistent challenges in drug delivery and resistance. Finally, we outline future directions, calling for cross-disciplinary collaboration and innovative clinical trial designs to personalize care and improve patient outcomes. Together, this review underscores the urgent need to bridge biology and therapy to transform the management of BCBrM.

乳腺癌脑转移(BCBrM)仍然是一个主要的临床挑战,治疗方案有限,预后差。尽管全身治疗取得了进展,但由于晚期乳腺癌患者的生存期延长,bbcbrm的发病率正在上升,但有效的脑靶向策略仍然缺乏,这凸显了一个关键的研究空白。这篇综述整合了最近阐明BCBrM复杂生物学基础的机制见解,并探讨了这些发现如何推动治疗创新。我们详细介绍了转移级联从局部入侵到脑定植,并检查了协调脑特异性转移的关键信号通路。重点是肿瘤细胞与脑微环境(包括星形胶质细胞、小胶质细胞和神经元)之间的动态串扰,以及代谢重编程和免疫逃避。我们批判性地评估当前的临床前模型及其翻译相关性,强调人性化和基于成像的系统的最新进展。新兴疗法,如中枢神经系统渗透激酶抑制剂、抗体-药物偶联物和免疫疗法,与药物传递和耐药性方面的持续挑战一起讨论。最后,我们概述了未来的发展方向,呼吁跨学科合作和创新的临床试验设计,以个性化护理和改善患者的结果。总之,这篇综述强调了迫切需要将生物学和治疗结合起来,以改变BCBrM的管理。
{"title":"Breast Cancer Brain Metastasis: Bridging Biological Mechanisms to Therapeutic Innovations","authors":"Jing Feng,&nbsp;Yuwei Tao,&nbsp;Fengkai Li,&nbsp;Suling Liu","doi":"10.1002/mog2.70043","DOIUrl":"https://doi.org/10.1002/mog2.70043","url":null,"abstract":"<p>Breast cancer brain metastasis (BCBrM) remains a major clinical challenge with limited therapeutic options and poor prognosis. Despite advances in systemic therapy, the incidence of BCBrM is rising due to prolonged survival of patients with advanced breast cancer, yet effective brain-targeted strategies remain scarce, underscoring a critical research gap. This review integrates recent mechanistic insights that illuminate the complex biology underpinning BCBrM and explores how these discoveries are driving therapeutic innovation. We detail the metastatic cascade from local invasion to brain colonization, and examine key signaling pathways orchestrating brain-specific metastasis. Emphasis is placed on the dynamic crosstalk between tumor cells and the brain microenvironment, including astrocytes, microglia, and neurons, as well as metabolic reprogramming and immune evasion. We critically evaluate current preclinical models and their translational relevance, highlighting recent advances in humanized and imaging-based systems. Emerging therapies, such as central nervous system-penetrant kinase inhibitors, antibody–drug conjugates, and immunotherapies, are discussed alongside persistent challenges in drug delivery and resistance. Finally, we outline future directions, calling for cross-disciplinary collaboration and innovative clinical trial designs to personalize care and improve patient outcomes. Together, this review underscores the urgent need to bridge biology and therapy to transform the management of BCBrM.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 4","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70043","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145367135","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
O-GlcNAc Transferase Promotes Metabolic Dysfunction-Associated Steatotic Liver Disease-Related Hepatocellular Carcinoma by Facilitating the Degradation of PTEN O-GlcNAc转移酶通过促进PTEN的降解促进代谢功能障碍相关的脂肪变性肝病相关的肝细胞癌
IF 2.2 Pub Date : 2025-10-14 DOI: 10.1002/mog2.70042
Haoran Liu, Suming Pu, Wenxin Zhu, Jing Huang, Jianming Li

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major risk factor for hepatocellular carcinoma (HCC), yet treatment options for advanced disease remain limited. O-GlcNAc transferase (OGT), the enzyme catalyzing O-GlcNAcylation, has been implicated in tumorigenesis, but its pro-cancer mechanism in MASLD-HCC remains poorly defined. Here, we show that OGT expression is significantly upregulated during MASLD-HCC progression and negatively regulates the tumor suppressor phosphatase and tensin homolog deleted on chromosome ten (PTEN) both in vivo and in vitro. Mechanistically, OGT catalyzes O-GlcNAcylation of PTEN at T382, which competitively inhibits the phosphorylation at the same residue. This modification promotes PTEN ubiquitination and accelerates its degradation. Importantly, O-GlcNAcylation of PTEN simultaneously impairs its intrinsic phospholipase activity. These dual effects compromise PTEN function, leading to activation of PI3K/Akt signaling pathway and enhanced tumor cell proliferation and migration. Moreover, pharmacological inhibition of OGT suppresses tumor growth and, when combined with PI3K/Akt pathway inhibitors, produces additive antitumor effects. These findings reveal a novel mechanism by which OGT-mediated O-GlcNAcylation destabilizes and inactivates PTEN, driving MASLD-HCC progression. They also highlight OGT and PTEN as promising therapeutic targets for developing novel strategies against HCC.

代谢功能障碍相关脂肪变性肝病(MASLD)是肝细胞癌(HCC)的主要危险因素,但晚期疾病的治疗选择仍然有限。O-GlcNAc转移酶(OGT)是一种催化o- glcn酰化的酶,与肿瘤发生有关,但其在MASLD-HCC中的致癌机制仍不明确。在这里,我们发现在体内和体外,在MASLD-HCC进展过程中,OGT的表达显著上调,并负调控第十号染色体上缺失的肿瘤抑制磷酸酶和紧张素同源物(PTEN)。从机制上讲,OGT催化PTEN在T382位点的o - glcn酰化,从而竞争性地抑制相同残基上的磷酸化。这种修饰促进PTEN泛素化并加速其降解。重要的是,PTEN的o - glcn酰化同时损害了其内在磷脂酶活性。这些双重作用损害PTEN功能,激活PI3K/Akt信号通路,增强肿瘤细胞的增殖和迁移。此外,OGT的药理抑制可抑制肿瘤生长,当与PI3K/Akt通路抑制剂联合使用时,可产生附加的抗肿瘤作用。这些发现揭示了一种新的机制,通过这种机制,ogt介导的o - glcn酰化使PTEN失稳和失活,从而推动MASLD-HCC的进展。他们还强调了OGT和PTEN是开发针对HCC的新策略的有希望的治疗靶点。
{"title":"O-GlcNAc Transferase Promotes Metabolic Dysfunction-Associated Steatotic Liver Disease-Related Hepatocellular Carcinoma by Facilitating the Degradation of PTEN","authors":"Haoran Liu,&nbsp;Suming Pu,&nbsp;Wenxin Zhu,&nbsp;Jing Huang,&nbsp;Jianming Li","doi":"10.1002/mog2.70042","DOIUrl":"https://doi.org/10.1002/mog2.70042","url":null,"abstract":"<p>Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major risk factor for hepatocellular carcinoma (HCC), yet treatment options for advanced disease remain limited. <i>O</i>-GlcNAc transferase (OGT), the enzyme catalyzing <i>O</i>-GlcNAcylation, has been implicated in tumorigenesis, but its pro-cancer mechanism in MASLD-HCC remains poorly defined. Here, we show that OGT expression is significantly upregulated during MASLD-HCC progression and negatively regulates the tumor suppressor phosphatase and tensin homolog deleted on chromosome ten (PTEN) both <i>in vivo</i> and <i>in vitro</i>. Mechanistically, OGT catalyzes <i>O</i>-GlcNAcylation of PTEN at T382, which competitively inhibits the phosphorylation at the same residue. This modification promotes PTEN ubiquitination and accelerates its degradation. Importantly, <i>O</i>-GlcNAcylation of PTEN simultaneously impairs its intrinsic phospholipase activity. These dual effects compromise PTEN function, leading to activation of PI3K/Akt signaling pathway and enhanced tumor cell proliferation and migration. Moreover, pharmacological inhibition of OGT suppresses tumor growth and, when combined with PI3K/Akt pathway inhibitors, produces additive antitumor effects. These findings reveal a novel mechanism by which OGT-mediated <i>O</i>-GlcNAcylation destabilizes and inactivates PTEN, driving MASLD-HCC progression. They also highlight OGT and PTEN as promising therapeutic targets for developing novel strategies against HCC.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 4","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70042","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145316903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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 Interplay of Aging and Cancer: Mechanisms, Implications, and Therapeutic Strategies 衰老和癌症的相互作用:机制、意义和治疗策略
IF 2.2 Pub Date : 2025-09-17 DOI: 10.1002/mog2.70041
Liangce Wang, Yaru Luo, Xin Chen, Yuan Wang, Yan Zhang

Aging is a complex biological process that significantly influences human health, including susceptibility to cancer. Although aging and cancer are distinct phenomena, they intersect through shared molecular mechanisms such as genomic instability, telomere attrition, epigenetic alterations, and chronic inflammation. Despite increasing recognition of these connections, how aging-related changes influence cancer development and treatment remains poorly understood. This review explores the intricate relationship between aging and cancer, highlighting how age-related changes in the tumor microenvironment, systemic inflammation and cellular senescence contribute to oncogenesis and tumor progression. We also assess the impact of aging on cancer treatment outcomes, as well as how cancer and its therapies may contribute to the acceleration of biological aging. Furthermore, we discuss potential intervention strategies that target the aging-related mechanisms that drive cancer development and progression. We review current progress and future directions in aging and cancer research, emphasizing that, with continuous technological advances and deepening insights, incorporating aging biology into oncology is both timely and necessary. By integrating recent advances in cancer biology and geroscience, this review offers insights critical for designing age-adapted therapeutic strategies. It underscores the need to shift toward personalized oncology approaches that account for the biological and clinical heterogeneity of aging.

衰老是一个复杂的生物过程,显著影响人类健康,包括对癌症的易感性。尽管衰老和癌症是截然不同的现象,但它们通过基因组不稳定、端粒磨损、表观遗传改变和慢性炎症等共同的分子机制相互交叉。尽管人们越来越认识到这些联系,但与衰老相关的变化如何影响癌症的发展和治疗仍然知之甚少。本文探讨了衰老与癌症之间的复杂关系,重点介绍了肿瘤微环境、全身炎症和细胞衰老中与年龄相关的变化如何促进肿瘤的发生和进展。我们还评估了衰老对癌症治疗结果的影响,以及癌症及其治疗如何加速生物衰老。此外,我们还讨论了针对驱动癌症发展和进展的衰老相关机制的潜在干预策略。我们回顾了衰老与癌症研究的现状和未来方向,强调随着技术的不断进步和认识的不断深入,将衰老生物学纳入肿瘤学是及时和必要的。通过整合癌症生物学和老年科学的最新进展,本综述为设计适合年龄的治疗策略提供了关键的见解。它强调需要转向个性化的肿瘤学方法,以解释衰老的生物学和临床异质性。
{"title":"The Interplay of Aging and Cancer: Mechanisms, Implications, and Therapeutic Strategies","authors":"Liangce Wang,&nbsp;Yaru Luo,&nbsp;Xin Chen,&nbsp;Yuan Wang,&nbsp;Yan Zhang","doi":"10.1002/mog2.70041","DOIUrl":"https://doi.org/10.1002/mog2.70041","url":null,"abstract":"<p>Aging is a complex biological process that significantly influences human health, including susceptibility to cancer. Although aging and cancer are distinct phenomena, they intersect through shared molecular mechanisms such as genomic instability, telomere attrition, epigenetic alterations, and chronic inflammation. Despite increasing recognition of these connections, how aging-related changes influence cancer development and treatment remains poorly understood. This review explores the intricate relationship between aging and cancer, highlighting how age-related changes in the tumor microenvironment, systemic inflammation and cellular senescence contribute to oncogenesis and tumor progression. We also assess the impact of aging on cancer treatment outcomes, as well as how cancer and its therapies may contribute to the acceleration of biological aging. Furthermore, we discuss potential intervention strategies that target the aging-related mechanisms that drive cancer development and progression. We review current progress and future directions in aging and cancer research, emphasizing that, with continuous technological advances and deepening insights, incorporating aging biology into oncology is both timely and necessary. By integrating recent advances in cancer biology and geroscience, this review offers insights critical for designing age-adapted therapeutic strategies. It underscores the need to shift toward personalized oncology approaches that account for the biological and clinical heterogeneity of aging.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 3","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70041","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145101748","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
β-Catenin/TCF4 Is Required for DDX17-Induced Epithelial–Mesenchymal Transition and Metastasis in Hepatocellular Carcinoma β-Catenin/TCF4在ddx17诱导的肝细胞癌上皮-间质转移和转移中是必需的
IF 2.2 Pub Date : 2025-09-14 DOI: 10.1002/mog2.70039
Chaoxiang Lv, Na Luo, Yuanning Luo, Qiqi Zhang, Xiuhua Cao, Jingliang Cheng, Chunli Wei, Qingxi Guo, Kan Guo, Zhiqiang Mei, Xiaoyan Liu, Junjiang Fu

The DEAD-box RNA helicase 17 (DDX17) is strongly linked to the occurrence and development of specific human cancers, emphasizing its previously unrecognized biological roles in cancer progression and metastasis. However, the precise mechanisms by which DDX17 regulates liver cancer metastasis have not been thoroughly explored. In this study, increased DDX17 expression levels showed a robust association with the invasive potential of hepatocellular carcinoma (HCC) cells. Silencing DDX17 expression resulted in substantial reduction of HCC cell migration and invasion potentials, while DDX17 overexpression had the opposite effect. Silencing DDX17 also attenuated epithelial–mesenchymal transition (EMT) in HCC cells and significantly reduced metastatic lesions in an orthotopic HCC nude mouse model. Mechanistically, chromatin immunoprecipitation assays revealed that TCF4 physically interacts with the DDX17 promoter, activating its transcriptional expression. Immunoprecipitation results demonstrated that DDX17-mediated nuclear input of β-catenin is dependent on its helicase functional domain. Furthermore, we demonstrated that β-catenin/TCF4 is essential for DDX17-induced migration and invasion in HCC cells. Taken together, these findings emphasize the significance of DDX17 in the malignant progression and metastasis of HCC, revealing a novel mechanism involving the β-catenin/TCF4/DDX17 pathway.

DEAD-box RNA解旋酶17 (DDX17)与特定人类癌症的发生和发展密切相关,强调了其在癌症进展和转移中以前未被认识到的生物学作用。然而,DDX17调控肝癌转移的确切机制尚未被充分探讨。在这项研究中,DDX17表达水平的升高与肝细胞癌(HCC)细胞的侵袭潜力密切相关。沉默DDX17表达可显著降低HCC细胞的迁移和侵袭潜能,而过表达DDX17则具有相反的效果。在原位肝癌裸鼠模型中,沉默DDX17还能减弱肝癌细胞的上皮-间质转化(EMT),并显著减少转移灶。在机制上,染色质免疫沉淀试验显示TCF4与DDX17启动子物理相互作用,激活其转录表达。免疫沉淀结果表明,ddx17介导的β-catenin的核输入依赖于其解旋酶功能域。此外,我们证明β-catenin/TCF4对于ddx17诱导的HCC细胞迁移和侵袭至关重要。综上所述,这些发现强调了DDX17在HCC恶性进展和转移中的重要性,揭示了一种涉及β-catenin/TCF4/DDX17通路的新机制。
{"title":"β-Catenin/TCF4 Is Required for DDX17-Induced Epithelial–Mesenchymal Transition and Metastasis in Hepatocellular Carcinoma","authors":"Chaoxiang Lv,&nbsp;Na Luo,&nbsp;Yuanning Luo,&nbsp;Qiqi Zhang,&nbsp;Xiuhua Cao,&nbsp;Jingliang Cheng,&nbsp;Chunli Wei,&nbsp;Qingxi Guo,&nbsp;Kan Guo,&nbsp;Zhiqiang Mei,&nbsp;Xiaoyan Liu,&nbsp;Junjiang Fu","doi":"10.1002/mog2.70039","DOIUrl":"https://doi.org/10.1002/mog2.70039","url":null,"abstract":"<p>The DEAD-box RNA helicase 17 (DDX17) is strongly linked to the occurrence and development of specific human cancers, emphasizing its previously unrecognized biological roles in cancer progression and metastasis. However, the precise mechanisms by which DDX17 regulates liver cancer metastasis have not been thoroughly explored. In this study, increased DDX17 expression levels showed a robust association with the invasive potential of hepatocellular carcinoma (HCC) cells. Silencing DDX17 expression resulted in substantial reduction of HCC cell migration and invasion potentials, while DDX17 overexpression had the opposite effect. Silencing DDX17 also attenuated epithelial–mesenchymal transition (EMT) in HCC cells and significantly reduced metastatic lesions in an orthotopic HCC nude mouse model. Mechanistically, chromatin immunoprecipitation assays revealed that TCF4 physically interacts with the DDX17 promoter, activating its transcriptional expression. Immunoprecipitation results demonstrated that DDX17-mediated nuclear input of β-catenin is dependent on its helicase functional domain. Furthermore, we demonstrated that β-catenin/TCF4 is essential for DDX17-induced migration and invasion in HCC cells. Taken together, these findings emphasize the significance of DDX17 in the malignant progression and metastasis of HCC, revealing a novel mechanism involving the β-catenin/TCF4/DDX17 pathway.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 3","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70039","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145062415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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 Epstein–Barr Virus Envelope Glycoprotein BALF4 Promotes the Ubiquitination and Degradation of RNA Acetyltransferase NAT10 Suppressing Gastric Cancer Progression eb病毒包膜糖蛋白BALF4促进RNA乙酰转移酶NAT10的泛素化和降解,抑制胃癌进展
IF 2.2 Pub Date : 2025-09-14 DOI: 10.1002/mog2.70040
Tianle Qiu, Chenbin Chen, Xiangwei Sun, Yuanbo Hu, Bujian Pan, Jun Xu, Jian Wen, Xian Shen, Xiangyang Xue, Xiaodong Chen

Epstein–Barr virus-associated gastric cancer (EBVaGC) is a unique subtype of gastric cancer (GC) with distinct molecular characteristics that generally has a better prognosis. BamHI-A leftward frame 4 (BALF4), an envelope glycoprotein encoded by the Epstein-Barr virus (EBV), plays an important role in EBV infection. However, its biological function and potential molecular mechanisms in EBVaGC remain unclear. This study aimed to investigate the impact of the highly expressed viral gene BALF4 on the progression of EBVaGC. Here, we detected the expression of BALF4 in GC tissue chips and validated that the presence of BALF4 might be associated with a favorable prognosis in EBVaGC. The results showed that BALF4 inhibited the proliferation, migration, and invasion of GC cells in vitro and in vivo. In addition, we discovered that BALF4 interacts with N-acetyltransferase 10 (NAT10). High expression of NAT10 in GC tissues promotes the malignant phenotype of GC cells. We discovered that BALF4 could inhibit the malignant progression of GC by promoting the ubiquitination and degradation of NAT10. In summary, our study revealed a possible mechanism explaining the favorable prognosis of the EBVaGC subtype, which contributes to a better understanding of this special type of GC.

Epstein-Barr病毒相关胃癌(EBVaGC)是一种独特的胃癌(GC)亚型,具有独特的分子特征,通常预后较好。BamHI-A左框4 (BALF4)是eb病毒(EBV)编码的包膜糖蛋白,在EBV感染中起重要作用。然而,其在EBVaGC中的生物学功能和潜在的分子机制尚不清楚。本研究旨在探讨高表达的病毒基因BALF4对EBVaGC进展的影响。我们在GC组织芯片中检测了BALF4的表达,证实了BALF4的存在可能与EBVaGC良好的预后有关。结果表明,BALF4在体外和体内均能抑制GC细胞的增殖、迁移和侵袭。此外,我们发现BALF4与n -乙酰转移酶10 (NAT10)相互作用。胃癌组织中NAT10的高表达促进胃癌细胞的恶性表型。我们发现BALF4可以通过促进NAT10的泛素化和降解来抑制GC的恶性进展。总之,我们的研究揭示了EBVaGC亚型预后良好的可能机制,有助于更好地理解这种特殊类型的GC。
{"title":"The Epstein–Barr Virus Envelope Glycoprotein BALF4 Promotes the Ubiquitination and Degradation of RNA Acetyltransferase NAT10 Suppressing Gastric Cancer Progression","authors":"Tianle Qiu,&nbsp;Chenbin Chen,&nbsp;Xiangwei Sun,&nbsp;Yuanbo Hu,&nbsp;Bujian Pan,&nbsp;Jun Xu,&nbsp;Jian Wen,&nbsp;Xian Shen,&nbsp;Xiangyang Xue,&nbsp;Xiaodong Chen","doi":"10.1002/mog2.70040","DOIUrl":"https://doi.org/10.1002/mog2.70040","url":null,"abstract":"<p>Epstein–Barr virus-associated gastric cancer (EBVaGC) is a unique subtype of gastric cancer (GC) with distinct molecular characteristics that generally has a better prognosis. <i>Bam</i>HI-A leftward frame 4 (<i>BALF4</i>), an envelope glycoprotein encoded by the Epstein-Barr virus (EBV), plays an important role in EBV infection. However, its biological function and potential molecular mechanisms in EBVaGC remain unclear. This study aimed to investigate the impact of the highly expressed viral gene <i>BALF4</i> on the progression of EBVaGC. Here, we detected the expression of BALF4 in GC tissue chips and validated that the presence of BALF4 might be associated with a favorable prognosis in EBVaGC. The results showed that BALF4 inhibited the proliferation, migration, and invasion of GC cells in vitro and in vivo. In addition, we discovered that BALF4 interacts with N-acetyltransferase 10 (NAT10). High expression of NAT10 in GC tissues promotes the malignant phenotype of GC cells. We discovered that BALF4 could inhibit the malignant progression of GC by promoting the ubiquitination and degradation of NAT10. In summary, our study revealed a possible mechanism explaining the favorable prognosis of the EBVaGC subtype, which contributes to a better understanding of this special type of GC.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 3","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70040","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145062416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lactate: A Key Promoter of Cancer Stem Cells and Implications for Cancer Therapy 乳酸:癌症干细胞的关键启动子及其对癌症治疗的影响
IF 2.2 Pub Date : 2025-09-04 DOI: 10.1002/mog2.70034
Yan Xue, Lin-Zhu Zhang

A groundbreaking study published in Cell Metabolism reveals how lactate promotes cancer stemness and therapy resistance by reshaping epigenetic landscapes [1]. By employing advanced single-cell tracking and machine learning in tumor organoids, the authors demonstrate that lactate suppresses differentiation and induces dedifferentiation of cancer cells into stem-like states through MYC activation. This discovery underscores lactate as a key regulator of tumor dynamics and identifies bromodomain protein 4 (BRD4) inhibitors as a promising therapeutic strategy to prevent relapse.

Tumor cells typically rely on glycolysis (the Warburg effect) for rapid energy supply, producing a large amount of lactate. Traditionally viewed as a metabolic byproduct, lactate is now recognized as a signaling molecule with multiple roles in the tumor microenvironment. The accumulation of lactate is closely associated with tumor proliferation, invasion, and drug resistance [2]. In gastric cancer, lactate promotes NBS1 K388 lactylation, enhancing MRN complex formation and recruitment to DNA double-strand breaks, thereby improving homologous recombination repair and mediating chemoradiotherapy resistance [3]. Lactate metabolism has thus emerged as a key focus in cancer research.

Cancer recurrence and metastasis remain major clinical challenges, driven largely by cancer stem cells (CSCs). CSCs exhibit self-renewal, multi-directional differentiation, and drug resistance, enabling survival post-therapy and tumor regeneration. Cancer differentiated cells (CDCs), as an important component of the tumor cell population, exhibit significant functional differences from CSCs. Research by the Rodríguez Colman team at the University Medical Center Utrecht revealed that murine intestinal stem cells and differentiated cells display distinct metabolic profiles and interact via lactate [4]. This suggests similar metabolic heterogeneity may exist between CSCs and CDCs in human intestinal tumors, with lactate playing a potential role in tumor progression. Further investigation into lactate's functional impact in these interactions is warranted.

Immunostaining of clinical samples showed that the expression level of monocarboxylate transporter 4 (MCT4), a lactate transporter, was significantly higher in CDCs than in CSCs. This indicates that CDCs have a higher glycolysis level. This finding demonstrates that, although tumor metabolism is generally characterized by the Warburg effect, there are notable differences in aerobic glycolysis levels among different tumor cell populations. Single-cell analysis further confirmed that the lactate level was elevated in CDCs relative to CSCs, indicating a higher glycolytic rate in CDCs. However, no significant differences were observed in basal glucose levels or glucose uptake between CDCs and CSCs. Interestingly, CSCs exhibited a greater capacity for lactate uptake compared to

发表在《细胞代谢》杂志上的一项突破性研究揭示了乳酸如何通过重塑表观遗传景观来促进癌症的发生和治疗抵抗。通过在肿瘤类器官中使用先进的单细胞跟踪和机器学习,作者证明乳酸抑制分化,并通过MYC激活诱导癌细胞去分化为干细胞样状态。这一发现强调了乳酸是肿瘤动力学的关键调节因子,并确定了溴结构域蛋白4 (BRD4)抑制剂是预防复发的有希望的治疗策略。肿瘤细胞通常依靠糖酵解(Warburg效应)快速提供能量,产生大量乳酸。传统上,乳酸被认为是一种代谢副产物,现在被认为是肿瘤微环境中具有多种作用的信号分子。乳酸的积累与肿瘤的增殖、侵袭和耐药密切相关。在胃癌中,乳酸促进NBS1 K388的乳酸化,增强MRN复合物的形成和DNA双链断裂的募集,从而改善同源重组修复,介导放化疗耐药[3]。因此,乳酸代谢已成为癌症研究的一个关键焦点。癌症复发和转移仍然是主要的临床挑战,主要由癌症干细胞(CSCs)驱动。CSCs表现出自我更新、多向分化和耐药,使其能够在治疗后存活和肿瘤再生。癌分化细胞(Cancer differentiated cells, cdc)作为肿瘤细胞群的重要组成部分,与csc在功能上存在显著差异。乌得勒支大学医学中心Rodríguez科尔曼团队的研究表明,小鼠肠道干细胞和分化细胞表现出不同的代谢谱,并通过乳酸[4]相互作用。这表明人类肠道肿瘤中CSCs和cdc之间可能存在类似的代谢异质性,乳酸盐在肿瘤进展中发挥潜在作用。进一步研究乳酸在这些相互作用中的功能影响是必要的。临床样品免疫染色显示,乳酸转运蛋白MCT4在CDCs中的表达水平明显高于CSCs。这表明cdc具有较高的糖酵解水平。这一发现表明,尽管肿瘤代谢通常以Warburg效应为特征,但不同肿瘤细胞群的有氧糖酵解水平存在显著差异。单细胞分析进一步证实,相对于CSCs, CDCs中的乳酸水平升高,表明CDCs的糖酵解速率更高。然而,cdc和CSCs在基础葡萄糖水平或葡萄糖摄取方面没有观察到显著差异。有趣的是,与cdc相比,CSCs表现出更大的乳酸摄取能力。基于这些观察,研究人员提出cdc产生和释放的乳酸被CSCs吸收,这代表了两种细胞类型之间的代谢相互作用。综上所述,cdc表现出比csc更高的糖酵解活性。基于这些代谢差异,研究人员进一步探讨了乳酸盐的影响。为了进一步研究CSCs和cdc之间的代谢差异,以及它们之间的相互作用,Rodríguez科尔曼的团队开发了一个人类类器官模型。他们植入了一个干细胞报告细胞(STAR)和三个代谢物传感器(分别报告NAD+/NADH、葡萄糖和乳酸)。分析得出了令人惊讶的结果:高乳酸浓度增加了CSC的数量。为了阐明这一现象背后的机制,研究人员对肿瘤类器官发育进行了4D成像。结果显示,在对照条件下,由于细胞分化,类器官组织中干细胞的比例随着时间的推移而下降,36%的细胞分化,不到1%的细胞进行去分化。相比之下,在乳酸处理下,细胞分化减少了50%,27%的细胞去分化。这些发现表明,乳酸诱导的CSC增加是由于抑制CSC分化和诱导CDC去分化进入CSC状态。为了进一步探讨乳酸如何抑制CSC分化并促进CDC去分化,研究人员对肠道干细胞的分裂模式进行了研究。通常,肠道干细胞分裂后,它们会产生两个具有相同命运的子细胞——要么都是已分化的细胞,要么都是干细胞。只有4.42%的干细胞分裂产生一个分化细胞和一个干细胞。然而,乳酸处理破坏了这种平衡,使不对称分裂的频率增加到18.3%,增加了三倍多。 值得注意的是,这种不对称分裂的增加主要是由两个分化细胞中的一个去分化驱动的,从CDC过渡到CSC。总之,乳酸通过将cdc转化为CSC而增加肿瘤内CSC的数量。这一过程的关键机制是乳酸重塑CDC代谢和诱导干性的能力。为了进一步研究这种去分化现象背后的机制,Rodríguez科尔曼的团队进行了多组学分析。他们发现乳酸增强了组蛋白乙酰化,增加了关键基因的染色质可及性,包括致癌基因MYC。同时,BRD4识别乙酰化赖氨酸残基,促进MYC基因转录,从而提高MYC蛋白水平。MYC作为WNT通路中众所周知的转录因子,在调节结直肠癌细胞的干细胞、增殖和迁移中起着至关重要的作用。这阐明了乳酸维持肿瘤中csc数量的机制。最后,研究人员证实BRD4抑制剂和MYC抑制剂都能有效抑制CSCs的增加(图1)。肠道肿瘤中CSC和CDC的代谢分工和动态相互作用机制为肿瘤异质性和治疗耐药提供了新的认识。本研究发现CDC通过高表达MCT4分泌乳酸,而CSC则通过高线粒体活性和mct1介导的乳酸摄取形成“乳酸循环”,形成代谢共生网络。这种代谢耦合不仅维持了CSC的干性,而且通过乳酸介导的表观遗传重编程促进了细胞的可塑性。具体来说,乳酸通过表观遗传机制(如组蛋白乙酰化)激活BRD4,并增强MYC转录。MYC作为转录因子,可以进一步调控WNT相关基因,形成正反馈回路,放大WNT信号。WNT/β-catenin通路(特别是结直肠癌APC突变过度激活)通过细胞核β-catenin/TCF/LEF复合物直接上调MYC转录,维持干细胞特性和增殖。这些发现揭示了肿瘤生态系统中代谢物-表观遗传偶联调节干细胞动力学的新范式,表明靶向乳酸- myc轴可能会破坏肿瘤细胞的可塑性,并为克服治疗耐药性提供突破。尽管本研究利用类器官模型和单细胞跟踪技术系统地阐明了乳酸代谢在肿瘤动力学中的核心作用,但也应承认某些局限性。目前的模型没有纳入体内微环境因素,如血管化和免疫相互作用。例如,肿瘤相关巨噬细胞(tumor associated macrophages, tam)在缺氧条件下分泌乳酸,通过旁分泌信号进一步增强CSC的可塑性。相反,乳酸可能会抑制细胞毒性t细胞的活性,创造一个免疫逃避生态位。此外,MYC抑制仅部分逆转乳酸效应,表明参与其他表观遗传调控机制,如乳酸化修饰[5]。未来的研究应通过动物模型和临床样本验证乳酸盐在转移和耐药中的调节作用。将代谢通量分析与蛋白质翻译后修饰组学相结合对于系统地破译代谢-表观遗传相互作用网络至关重要。从转化医学的角度来看,开发针对乳酸转运蛋白(MCT1/4)或组蛋白乙酰转移酶(如p300/CBP)的新型抑制剂,并探索其与现有BRD4抑制剂的协同作用,可能为克服肿瘤代谢适应性提供有效策略。此外,基于乳酸水平和CSC丰度的诊断标志物的发展,结合代谢成像技术,将为个性化治疗策略提供关键见解。推进这些多维度的研究方向,有望实现临床转化的突破,从“代谢重编程”向“治疗重编程”转变。严雪:写作—原稿,写作—审稿、编辑。张林竹:构思、资金获取、撰写、审稿、编辑。两位作者已经阅读并批准了最终的手稿。作者没有什么可报告的。作者声明无利益冲突。
{"title":"Lactate: A Key Promoter of Cancer Stem Cells and Implications for Cancer Therapy","authors":"Yan Xue,&nbsp;Lin-Zhu Zhang","doi":"10.1002/mog2.70034","DOIUrl":"https://doi.org/10.1002/mog2.70034","url":null,"abstract":"<p>A groundbreaking study published in <i>Cell Metabolism</i> reveals how lactate promotes cancer stemness and therapy resistance by reshaping epigenetic landscapes [<span>1</span>]. By employing advanced single-cell tracking and machine learning in tumor organoids, the authors demonstrate that lactate suppresses differentiation and induces dedifferentiation of cancer cells into stem-like states through MYC activation. This discovery underscores lactate as a key regulator of tumor dynamics and identifies bromodomain protein 4 (BRD4) inhibitors as a promising therapeutic strategy to prevent relapse.</p><p>Tumor cells typically rely on glycolysis (the Warburg effect) for rapid energy supply, producing a large amount of lactate. Traditionally viewed as a metabolic byproduct, lactate is now recognized as a signaling molecule with multiple roles in the tumor microenvironment. The accumulation of lactate is closely associated with tumor proliferation, invasion, and drug resistance [<span>2</span>]. In gastric cancer, lactate promotes NBS1 K388 lactylation, enhancing MRN complex formation and recruitment to DNA double-strand breaks, thereby improving homologous recombination repair and mediating chemoradiotherapy resistance [<span>3</span>]. Lactate metabolism has thus emerged as a key focus in cancer research.</p><p>Cancer recurrence and metastasis remain major clinical challenges, driven largely by cancer stem cells (CSCs). CSCs exhibit self-renewal, multi-directional differentiation, and drug resistance, enabling survival post-therapy and tumor regeneration. Cancer differentiated cells (CDCs), as an important component of the tumor cell population, exhibit significant functional differences from CSCs. Research by the Rodríguez Colman team at the University Medical Center Utrecht revealed that murine intestinal stem cells and differentiated cells display distinct metabolic profiles and interact via lactate [<span>4</span>]. This suggests similar metabolic heterogeneity may exist between CSCs and CDCs in human intestinal tumors, with lactate playing a potential role in tumor progression. Further investigation into lactate's functional impact in these interactions is warranted.</p><p>Immunostaining of clinical samples showed that the expression level of monocarboxylate transporter 4 (MCT4), a lactate transporter, was significantly higher in CDCs than in CSCs. This indicates that CDCs have a higher glycolysis level. This finding demonstrates that, although tumor metabolism is generally characterized by the Warburg effect, there are notable differences in aerobic glycolysis levels among different tumor cell populations. Single-cell analysis further confirmed that the lactate level was elevated in CDCs relative to CSCs, indicating a higher glycolytic rate in CDCs. However, no significant differences were observed in basal glucose levels or glucose uptake between CDCs and CSCs. Interestingly, CSCs exhibited a greater capacity for lactate uptake compared to ","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 3","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70034","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144998988","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
MedComm – Oncology
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