首页 > 最新文献

Cellular Oncology最新文献

英文 中文
COPB2 facilitates EDEM3-mediated mannose trimming to sustain ER homeostasis in ovarian cancer. COPB2促进edem3介导的甘露糖修剪以维持卵巢癌内质网稳态。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-07-30 DOI: 10.1007/s13402-025-01089-y
Dong-Xue Li, Ni Yang, Lan-Yu Hua, Jun-Jie Wang, Dilinazi Abudujilile, Zhi-Gang Zhang, Peng-Feng Zhu, Ting-Yan Shi, Rong Zhang

Background: Ovarian cancer (OC) is a lethal gynecologic malignancy with limited therapeutic success due to late diagnosis and therapy resistance. Endoplasmic reticulum (ER) stress and ER-associated degradation (ERAD) are key to tumor adaptation, yet the mechanisms sustaining ER homeostasis in OC remain poorly defined.

Methods: We combined multi-omics analyses, tissue microarrays, and in vitro and in vivo models. Functional assays involved COPB2 knockdown or overexpression in OC cells, xenografts in nude mice, and mechanistic studies including protein interaction and glycoproteomic analyses.

Results: COPB2 was significantly upregulated in OC and associated with poor prognosis. It promoted cell proliferation and survival by alleviating ER stress and suppressing apoptosis. Mechanistically, COPB2 interacted with EDEM3, a key ERAD enzyme, enhancing its ER localization and mannose-trimming function. COPB2 depletion impaired EDEM3 activity, resulting in glycan processing defects and ER stress accumulation. In vivo, COPB2 overexpression accelerated tumor growth.

Conclusions: This study identifies a novel COPB2-EDEM3 axis that maintains ER homeostasis and drives OC progression. Targeting this axis may offer new opportunities for therapeutic intervention and biomarker development.

背景:卵巢癌(OC)是一种致命的妇科恶性肿瘤,由于诊断较晚和治疗耐药,治疗成功率有限。内质网(ER)应激和内质网相关降解(ERAD)是肿瘤适应的关键,但在OC中维持内质网稳态的机制尚不清楚。方法:采用多组学分析、组织微阵列、体外和体内模型相结合的方法。功能分析包括在OC细胞中敲除或过表达COPB2,裸鼠异种移植,以及包括蛋白质相互作用和糖蛋白组学分析在内的机制研究。结果:COPB2在OC中显著上调,且与预后不良相关。它通过减轻内质网应激和抑制细胞凋亡促进细胞增殖和存活。在机制上,COPB2与ERAD关键酶EDEM3相互作用,增强其内质网定位和甘露糖修剪功能。COPB2缺失损害了EDEM3活性,导致多糖加工缺陷和内质网胁迫积累。在体内,COPB2过表达加速了肿瘤的生长。结论:本研究确定了一种新的COPB2-EDEM3轴,该轴维持内质网稳态并驱动OC进展。靶向这一轴可能为治疗干预和生物标志物的开发提供新的机会。
{"title":"COPB2 facilitates EDEM3-mediated mannose trimming to sustain ER homeostasis in ovarian cancer.","authors":"Dong-Xue Li, Ni Yang, Lan-Yu Hua, Jun-Jie Wang, Dilinazi Abudujilile, Zhi-Gang Zhang, Peng-Feng Zhu, Ting-Yan Shi, Rong Zhang","doi":"10.1007/s13402-025-01089-y","DOIUrl":"10.1007/s13402-025-01089-y","url":null,"abstract":"<p><strong>Background: </strong>Ovarian cancer (OC) is a lethal gynecologic malignancy with limited therapeutic success due to late diagnosis and therapy resistance. Endoplasmic reticulum (ER) stress and ER-associated degradation (ERAD) are key to tumor adaptation, yet the mechanisms sustaining ER homeostasis in OC remain poorly defined.</p><p><strong>Methods: </strong>We combined multi-omics analyses, tissue microarrays, and in vitro and in vivo models. Functional assays involved COPB2 knockdown or overexpression in OC cells, xenografts in nude mice, and mechanistic studies including protein interaction and glycoproteomic analyses.</p><p><strong>Results: </strong>COPB2 was significantly upregulated in OC and associated with poor prognosis. It promoted cell proliferation and survival by alleviating ER stress and suppressing apoptosis. Mechanistically, COPB2 interacted with EDEM3, a key ERAD enzyme, enhancing its ER localization and mannose-trimming function. COPB2 depletion impaired EDEM3 activity, resulting in glycan processing defects and ER stress accumulation. In vivo, COPB2 overexpression accelerated tumor growth.</p><p><strong>Conclusions: </strong>This study identifies a novel COPB2-EDEM3 axis that maintains ER homeostasis and drives OC progression. Targeting this axis may offer new opportunities for therapeutic intervention and biomarker development.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1465-1477"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528345/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
S100A9 promotes resistance to anti-PD-1 immunotherapy in hepatocellular carcinoma by degrading PARP1 and activating the STAT3/PD-L1 pathway. S100A9通过降解PARP1和激活STAT3/PD-L1通路,促进肝癌患者对抗pd -1免疫治疗的耐药性。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-07-14 DOI: 10.1007/s13402-025-01087-0
Xianwei Zhou, Chu Qiao, Xuehui Chu, Yajing Yang, Haoran Man, Jingxin Liu, Yunzheng Li, Zhu Xu, Huan Li, Xiaodong Shan, Zaowu Lian, Yanjun Lu, Weihong Wang, Decai Yu, Xitai Sun, Binghua Li

Background: Immune checkpoint inhibitors (ICIs), such as anti-programmed cell death protein-1 (PD-1) immunotherapy, have emerged as promising treatments for advanced hepatocellular carcinoma (HCC), significantly improving clinical outcomes. However, resistance to ICIs remains a major challenge, and the underlying mechanisms of this resistance are not yet fully understood. This study aimed to investigate the role of S100 calcium-binding protein A9 (S100A9) in mediating resistance to anti-PD-1 therapy.

Approach and results: We conducted RNA sequencing (RNA-seq) on tumor samples from anti-PD-1 responders and non-responders in HCC patients. Differential expression analysis identified S100A9 as a potential driver gene of resistance to anti-PD-1 therapy. Subcutaneous tumor models and an orthotopic HCC model established via hydrodynamic transfection were utilized to evaluate the impact of S100A9 on the efficacy of PD-1 therapy. Our findings revealed that S100A9 promotes resistance to anti-PD-1 therapy in HCC. Mechanistically, S100A9 directly interacted with PARP1 and induced its degradation via the ubiquitin-proteasome pathway. This process increased STAT3 phosphorylation at Tyr705, thereby enhancing PD-L1 transcription. Notably, treatment with the S100A9 inhibitor Tasquinimod significantly improved the efficacy of anti-PD-1 therapy in HCC.

Conclusions: Our study reveals that S100A9 facilitates immune evasion in HCC by enhancing PARP1 ubiquitination, STAT3 phosphorylation, and PD-L1 expression. Furthermore, combining S100A9 inhibitors with anti-PD-1 antibodies markedly enhances the therapeutic efficacy of ICIs in HCC. These findings highlight S100A9 as a potential therapeutic target for overcoming resistance to immunotherapy in HCC.

背景:免疫检查点抑制剂(ICIs),如抗程序性细胞死亡蛋白-1 (PD-1)免疫疗法,已经成为晚期肝细胞癌(HCC)的有希望的治疗方法,显著改善了临床结果。然而,对ICIs的耐药性仍然是一个主要挑战,而且这种耐药性的潜在机制尚未完全了解。本研究旨在探讨S100钙结合蛋白A9 (S100A9)在介导抗pd -1治疗耐药中的作用。方法和结果:我们对HCC患者抗pd -1反应者和无反应者的肿瘤样本进行了RNA测序(RNA-seq)。差异表达分析发现S100A9是抗pd -1治疗耐药的潜在驱动基因。采用皮下肿瘤模型和原位肝癌水动力转染模型评价S100A9对PD-1治疗效果的影响。我们的研究结果显示,S100A9促进肝癌患者对抗pd -1治疗的耐药。机制上,S100A9直接与PARP1相互作用,并通过泛素-蛋白酶体途径诱导其降解。这一过程增加了STAT3 Tyr705位点的磷酸化,从而增强了PD-L1的转录。值得注意的是,使用S100A9抑制剂Tasquinimod治疗可显著提高抗pd -1治疗HCC的疗效。结论:我们的研究表明,S100A9通过增强PARP1泛素化、STAT3磷酸化和PD-L1表达,促进HCC的免疫逃避。此外,S100A9抑制剂与抗pd -1抗体联合使用可显著提高ICIs在HCC中的治疗效果。这些发现强调S100A9是克服HCC免疫治疗耐药的潜在治疗靶点。
{"title":"S100A9 promotes resistance to anti-PD-1 immunotherapy in hepatocellular carcinoma by degrading PARP1 and activating the STAT3/PD-L1 pathway.","authors":"Xianwei Zhou, Chu Qiao, Xuehui Chu, Yajing Yang, Haoran Man, Jingxin Liu, Yunzheng Li, Zhu Xu, Huan Li, Xiaodong Shan, Zaowu Lian, Yanjun Lu, Weihong Wang, Decai Yu, Xitai Sun, Binghua Li","doi":"10.1007/s13402-025-01087-0","DOIUrl":"10.1007/s13402-025-01087-0","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs), such as anti-programmed cell death protein-1 (PD-1) immunotherapy, have emerged as promising treatments for advanced hepatocellular carcinoma (HCC), significantly improving clinical outcomes. However, resistance to ICIs remains a major challenge, and the underlying mechanisms of this resistance are not yet fully understood. This study aimed to investigate the role of S100 calcium-binding protein A9 (S100A9) in mediating resistance to anti-PD-1 therapy.</p><p><strong>Approach and results: </strong>We conducted RNA sequencing (RNA-seq) on tumor samples from anti-PD-1 responders and non-responders in HCC patients. Differential expression analysis identified S100A9 as a potential driver gene of resistance to anti-PD-1 therapy. Subcutaneous tumor models and an orthotopic HCC model established via hydrodynamic transfection were utilized to evaluate the impact of S100A9 on the efficacy of PD-1 therapy. Our findings revealed that S100A9 promotes resistance to anti-PD-1 therapy in HCC. Mechanistically, S100A9 directly interacted with PARP1 and induced its degradation via the ubiquitin-proteasome pathway. This process increased STAT3 phosphorylation at Tyr705, thereby enhancing PD-L1 transcription. Notably, treatment with the S100A9 inhibitor Tasquinimod significantly improved the efficacy of anti-PD-1 therapy in HCC.</p><p><strong>Conclusions: </strong>Our study reveals that S100A9 facilitates immune evasion in HCC by enhancing PARP1 ubiquitination, STAT3 phosphorylation, and PD-L1 expression. Furthermore, combining S100A9 inhibitors with anti-PD-1 antibodies markedly enhances the therapeutic efficacy of ICIs in HCC. These findings highlight S100A9 as a potential therapeutic target for overcoming resistance to immunotherapy in HCC.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1433-1447"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144625466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Apo10 and TKTL1 in blood macrophages as non-invasive biomarkers for early detection of cervical cancer. 血巨噬细胞中Apo10和TKTL1作为宫颈癌早期检测的非侵入性生物标志物
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-07-28 DOI: 10.1007/s13402-025-01092-3
Shuqing Wang, Zhiying Mao, Yuying Liu, Chunyan Lan, Lizhi Liu, Musheng Zeng, Chuanbo Xie

Purpose: Apo10 and TKTL1 are tumor-associated markers reflecting impaired apoptosis and enhanced glycolysis respectively. This study aimed to evaluate the diagnostic potential of Apo10, TKTL1, and APT (a combination of Apo10 and TKTL1) in screening early-stage cervical cancer.

Methods: A total of 152 patients with cervical cancer and 152 age-matched healthy controls were enrolled at Sun Yat-sen University Cancer Center from November 2020 to August 2023. Clinical data were collected from the Hospital Information System (HIS) and medical records, and blood samples were collected from all participants before treatment using epitope detection in monocytes (EDIM) technology 60 min after their last meal. Descriptive statistics and receiver operating characteristic (ROC) curves were used to compare the diagnostic performance of Apo10, TKTL1, and APT to those of conventional cervical cancer biomarkers (CEA, CA125, and SCC-A).

Results: Most of the enrolled patients with cervical cancer had early-stage disease (70%) and squamous cell histology (84.9%). The Apo10, TKTL1, and APT levels were significantly higher in the cervical cancer group than in the control group (Apo10:139 vs. 132, TKTL1:121 vs. 114, APT: 260 vs. 246). We also found that Apo10, TKTL1, and APT showed superior diagnostic performance (AUC: 0.864, 0.865, 0.905) compared to traditional markers (CEA: 0.690, CA125: 0.594, SCC-A: 0.806). Sensitivity analysis revealed APT maintained high diagnostic value across tumor stages and in both HPV-negative (AUC = 0.967) and TCT-negative (AUC = 0.958) subgroups.

Conclusion: Apo10, TKTL1, and APT outperform conventional biomarkers in detecting cervical cancer and may serve as reliable diagnostic indicators.

目的:Apo10和TKTL1分别是反映细胞凋亡受损和糖酵解增强的肿瘤相关标志物。本研究旨在评估Apo10、TKTL1和APT (Apo10和TKTL1的组合)在筛查早期宫颈癌中的诊断潜力。方法:中山大学癌症中心于2020年11月至2023年8月招募了152例宫颈癌患者和152例年龄匹配的健康对照。从医院信息系统(HIS)和病历中收集临床数据,并在最后一餐后60分钟使用单核细胞表位检测(EDIM)技术采集所有参与者治疗前的血液样本。采用描述性统计和受试者工作特征(ROC)曲线比较Apo10、TKTL1和APT与常规宫颈癌生物标志物(CEA、CA125和SCC-A)的诊断效能。结果:入组的宫颈癌患者以早期病变(70%)和鳞状细胞组织学(84.9%)为主。宫颈癌组Apo10、TKTL1和APT水平明显高于对照组(Apo10:139 vs. 132, TKTL1:121 vs. 114, APT: 260 vs. 246)。我们还发现,Apo10、TKTL1和APT与传统标记(CEA: 0.690、CA125: 0.594、SCC-A: 0.806)相比,具有更好的诊断效果(AUC分别为0.864、0.865、0.905)。敏感性分析显示,APT在不同肿瘤分期、hpv阴性(AUC = 0.967)和tct阴性(AUC = 0.958)亚组均保持较高的诊断价值。结论:Apo10、TKTL1、APT在检测宫颈癌方面优于常规生物标志物,可作为可靠的诊断指标。
{"title":"Apo10 and TKTL1 in blood macrophages as non-invasive biomarkers for early detection of cervical cancer.","authors":"Shuqing Wang, Zhiying Mao, Yuying Liu, Chunyan Lan, Lizhi Liu, Musheng Zeng, Chuanbo Xie","doi":"10.1007/s13402-025-01092-3","DOIUrl":"10.1007/s13402-025-01092-3","url":null,"abstract":"<p><strong>Purpose: </strong>Apo10 and TKTL1 are tumor-associated markers reflecting impaired apoptosis and enhanced glycolysis respectively. This study aimed to evaluate the diagnostic potential of Apo10, TKTL1, and APT (a combination of Apo10 and TKTL1) in screening early-stage cervical cancer.</p><p><strong>Methods: </strong>A total of 152 patients with cervical cancer and 152 age-matched healthy controls were enrolled at Sun Yat-sen University Cancer Center from November 2020 to August 2023. Clinical data were collected from the Hospital Information System (HIS) and medical records, and blood samples were collected from all participants before treatment using epitope detection in monocytes (EDIM) technology 60 min after their last meal. Descriptive statistics and receiver operating characteristic (ROC) curves were used to compare the diagnostic performance of Apo10, TKTL1, and APT to those of conventional cervical cancer biomarkers (CEA, CA125, and SCC-A).</p><p><strong>Results: </strong>Most of the enrolled patients with cervical cancer had early-stage disease (70%) and squamous cell histology (84.9%). The Apo10, TKTL1, and APT levels were significantly higher in the cervical cancer group than in the control group (Apo10:139 vs. 132, TKTL1:121 vs. 114, APT: 260 vs. 246). We also found that Apo10, TKTL1, and APT showed superior diagnostic performance (AUC: 0.864, 0.865, 0.905) compared to traditional markers (CEA: 0.690, CA125: 0.594, SCC-A: 0.806). Sensitivity analysis revealed APT maintained high diagnostic value across tumor stages and in both HPV-negative (AUC = 0.967) and TCT-negative (AUC = 0.958) subgroups.</p><p><strong>Conclusion: </strong>Apo10, TKTL1, and APT outperform conventional biomarkers in detecting cervical cancer and may serve as reliable diagnostic indicators.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1513-1522"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528240/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Efficacy of CDK4/6 Inhibition in colorectal cancer and the role of p16 expression in predicting drug resistance. CDK4/6在结直肠癌中的抑制作用及p16表达在预测耐药中的作用
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-06-16 DOI: 10.1007/s13402-025-01080-7
Julia S Schneider, Najib Ben Khaled, Liangtao Ye, Ralf Wimmer, Linda Hammann, Alexander Weich, Christoph Suppan, Ujjwal M Mahajan, Andreas Jung, Jörg Kumbrink, Gerald Denk, Monika Rau, Volker Kunzmann, Solveig Kuss, Jens Neuman, Julia Mayerle, Andreas Geier, Heike M Hermanns, Enrico N De Toni, Florian P Reiter

Introduction: Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. The use of sequential polychemotherapies has improved the survival of patients with advanced metastatic disease. However, the survival rates achieved are low, and chemotherapy-related side effects are significant. Therefore, new, efficient, and tolerable therapies are urgently needed. In this study, we investigate the efficacy of pharmacological cyclin D-dependent kinase (CDK) 4/6 inhibition and explore the relevance of p16 as predictors of susceptibility to CDK 4/6 therapy.

Materials and methods: CDK 4/6 inhibitors were evaluated in native and FOLFOX- or ribociclib-resistant CRC, hepatocellular carcinoma (HCC), and breast cancer (BC) cell lines using viability, colony formation, and flow cytometry (FC)-based assays. Western blotting was employed to assess the expression of Rb and members of the INK4 family. SiRNA-based knockdown of CDK4/6 was utilized to gain insights into mechanisms of action or resistance. Tissue from 185 CRC patients was examined for the expression of p16 and its relevance for progression-free and overall survival. The prognostic relevance of cyclin-dependent kinase inhibitor 2 A (CDKN2A) mRNA expression data was derived from The Cancer Genome Atlas (TCGA) data.

Results: Ribociclib demonstrates significant antitumoral effects in various CRC, HCC, and BC cell lines, similar to two other approved CDK4/6 inhibitors (palbociclib and abemaciclib). Ribociclib-resistant cell lines (Hep-3B, HCC-1937, and BT-549) exhibited higher p16 expression compared to ribociclib-sensitive cell lines. In ribociclib-sensitive cell lines, CDK4/6 inhibition led to G1 phase arrest, whereas resistant cells did not exhibit such effects. A similar phenotype could be observed upon dual siRNA based CDK4/6 knockdown in ribociclib-sensitive HuH-7 and ribociclib-resistant Hep-3B cell lines. All CRC cell lines tested showed sensitivity to ribociclib, including the FOLFOX-resistant SW620 cell line. Low mRNA expression of CDKN2A (p16) was associated with favorable prognosis in CRC patients. No prognostic significance was found for p16 protein expression in an early-stage CRC cohort (n = 185).

Conclusion: Ribociclib demonstrates significant antitumoral effects across a large panel of cancer cell lines and chemoresistant models, especially in CRC. Resistance towards ribociclib is associated with high p16 expression, which is a negative prognostic marker for patients with CRC. Our findings underscore p16 as a promising biomarker for predicting ribociclib responsiveness and emphasize the need for further mechanistic studies and combination therapy approaches to overcome resistance in p16high patients.

导读:结直肠癌(CRC)是全球癌症相关死亡的主要原因。序贯多药化疗的使用提高了晚期转移性疾病患者的生存率。然而,获得的存活率很低,化疗相关的副作用也很明显。因此,迫切需要新的、有效的、可耐受的治疗方法。在这项研究中,我们研究了药物周期蛋白d依赖性激酶(CDK) 4/6抑制的有效性,并探讨了p16作为CDK 4/6治疗易感性预测因子的相关性。材料和方法:cdk4 /6抑制剂在原生和FOLFOX或核糖素耐药的结直肠癌、肝细胞癌(HCC)和乳腺癌(BC)细胞系中使用活力、集落形成和基于流式细胞术(FC)的检测进行评估。Western blotting检测Rb及INK4家族成员的表达。利用sirna敲低CDK4/6来深入了解作用或抗性机制。我们检测了185例结直肠癌患者的组织中p16的表达及其与无进展生存期和总生存期的相关性。细胞周期蛋白依赖性激酶抑制剂2a (CDKN2A) mRNA表达数据与预后的相关性来源于癌症基因组图谱(TCGA)数据。结果:Ribociclib在各种CRC、HCC和BC细胞系中显示出显著的抗肿瘤作用,类似于另外两种已批准的CDK4/6抑制剂(palbociclib和abemaciclib)。与核糖素敏感细胞系相比,核糖素耐药细胞系(Hep-3B、HCC-1937和BT-549)表现出更高的p16表达。在核糖素敏感细胞系中,CDK4/6抑制导致G1期阻滞,而耐药细胞没有表现出这种作用。在核糖素敏感的HuH-7和核糖素耐药的Hep-3B细胞系中,基于双siRNA的CDK4/6敲低也可以观察到类似的表型。所有CRC细胞系均显示对核糖环尼的敏感性,包括耐folfox的SW620细胞系。CDKN2A (p16) mRNA低表达与CRC患者预后良好相关。在早期CRC队列(n = 185)中,未发现p16蛋白表达的预后意义。结论:Ribociclib在大量的癌细胞系和化疗耐药模型中显示出显著的抗肿瘤作用,特别是在结直肠癌中。对ribociclib的耐药与p16的高表达有关,p16是结直肠癌患者的阴性预后标志物。我们的研究结果强调了p16作为预测核糖环尼反应性的有希望的生物标志物,并强调需要进一步的机制研究和联合治疗方法来克服p16高患者的耐药。
{"title":"Efficacy of CDK4/6 Inhibition in colorectal cancer and the role of p16 expression in predicting drug resistance.","authors":"Julia S Schneider, Najib Ben Khaled, Liangtao Ye, Ralf Wimmer, Linda Hammann, Alexander Weich, Christoph Suppan, Ujjwal M Mahajan, Andreas Jung, Jörg Kumbrink, Gerald Denk, Monika Rau, Volker Kunzmann, Solveig Kuss, Jens Neuman, Julia Mayerle, Andreas Geier, Heike M Hermanns, Enrico N De Toni, Florian P Reiter","doi":"10.1007/s13402-025-01080-7","DOIUrl":"10.1007/s13402-025-01080-7","url":null,"abstract":"<p><strong>Introduction: </strong>Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. The use of sequential polychemotherapies has improved the survival of patients with advanced metastatic disease. However, the survival rates achieved are low, and chemotherapy-related side effects are significant. Therefore, new, efficient, and tolerable therapies are urgently needed. In this study, we investigate the efficacy of pharmacological cyclin D-dependent kinase (CDK) 4/6 inhibition and explore the relevance of p16 as predictors of susceptibility to CDK 4/6 therapy.</p><p><strong>Materials and methods: </strong>CDK 4/6 inhibitors were evaluated in native and FOLFOX- or ribociclib-resistant CRC, hepatocellular carcinoma (HCC), and breast cancer (BC) cell lines using viability, colony formation, and flow cytometry (FC)-based assays. Western blotting was employed to assess the expression of Rb and members of the INK4 family. SiRNA-based knockdown of CDK4/6 was utilized to gain insights into mechanisms of action or resistance. Tissue from 185 CRC patients was examined for the expression of p16 and its relevance for progression-free and overall survival. The prognostic relevance of cyclin-dependent kinase inhibitor 2 A (CDKN2A) mRNA expression data was derived from The Cancer Genome Atlas (TCGA) data.</p><p><strong>Results: </strong>Ribociclib demonstrates significant antitumoral effects in various CRC, HCC, and BC cell lines, similar to two other approved CDK4/6 inhibitors (palbociclib and abemaciclib). Ribociclib-resistant cell lines (Hep-3B, HCC-1937, and BT-549) exhibited higher p16 expression compared to ribociclib-sensitive cell lines. In ribociclib-sensitive cell lines, CDK4/6 inhibition led to G1 phase arrest, whereas resistant cells did not exhibit such effects. A similar phenotype could be observed upon dual siRNA based CDK4/6 knockdown in ribociclib-sensitive HuH-7 and ribociclib-resistant Hep-3B cell lines. All CRC cell lines tested showed sensitivity to ribociclib, including the FOLFOX-resistant SW620 cell line. Low mRNA expression of CDKN2A (p16) was associated with favorable prognosis in CRC patients. No prognostic significance was found for p16 protein expression in an early-stage CRC cohort (n = 185).</p><p><strong>Conclusion: </strong>Ribociclib demonstrates significant antitumoral effects across a large panel of cancer cell lines and chemoresistant models, especially in CRC. Resistance towards ribociclib is associated with high p16 expression, which is a negative prognostic marker for patients with CRC. Our findings underscore p16 as a promising biomarker for predicting ribociclib responsiveness and emphasize the need for further mechanistic studies and combination therapy approaches to overcome resistance in p16<sup>high</sup> patients.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1363-1375"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528337/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144301189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
SOX4 mediates cancer-associated fibroblasts related radioresistance in hepatocellular carcinoma. SOX4介导肝癌中癌相关成纤维细胞相关放射耐药
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-07-30 DOI: 10.1007/s13402-025-01088-z
Ke Jiang, Botian Huang, Shasha He, Meiyan Zhu, Xiao Zhao, Miaowen Liu, Zhenwei Peng, Yan Wang, Yong Chen

Purpose: Cancer-associated fibroblasts (CAFs), a crucial component of tumor microenvironment, play a critical role in tumorigenesis, progression, and conferring resistance to radiotherapy and chemotherapy. This study aimed to investigate the association between CAFs, CAF- related genes, and radioresistance in hepatocellular carcinoma (HCC).

Methods: CAFs were isolated from HCC tissues and subsequently utilized for co-culturing with HCC cells using CAFs-conditioned medium. An orthotopic HCC mouse model was established by co-implanting CAFs and HCC cells. Through integrative analysis of three RNA-sequencing datasets (TCGA-LIHC tumor vs. normal tissues, Huh7 radioresistant vs. parent cells, and CAF vs. control group), CAF-associated prognostic genes were identified using comprehensive bioinformatics approaches. Experimental validation was performed by real-time quantitative PCR, western blot, immunohistochemistry, cell viability assays, and colony formation assays.

Results: Our findings demonstrated that CAFs significantly enhance radioresistance in HCC. Based on 13 CAF-related prognostic genes, TCGA-LIHC patients were stratified into two distinct clusters via consensus clustering, exhibiting significant differences in overall survival, immune cell infiltration, and therapeutic response. A prognostic nomogram incorporating three hub genes and clinical characteristics was developed. Notably, SOX4 was upregulated in tumor tissues, radioresistant cells, and CAF-exposed HCC cells, correlating with poor prognosis. SOX4 knockdown suppressed HCC proliferation and reversed CAF-induced radioresistance. Additionally, a competitive endogenous RNA (ceRNA) network of LINC00665/miR-122-5p/SOX4 was constructed.

Conclusion: CAFs serve as crucial mediators of radioresistance in HCC, and CAF-related genes provide valuable prognostic and therapeutic insights. SOX4 emerges as a promising therapeutic target to improve radiotherapy efficacy in HCC.

Clinical trial number: Not applicable.

目的:肿瘤相关成纤维细胞(Cancer-associated fibroblasts, CAFs)是肿瘤微环境的重要组成部分,在肿瘤的发生、发展和对放疗和化疗的耐药性中起着关键作用。本研究旨在探讨CAF、CAF相关基因与肝细胞癌(HCC)放射耐药之间的关系。方法:从HCC组织中分离出CAFs,随后使用CAFs条件培养基与HCC细胞共培养。采用人工肝细胞与肝癌细胞共植的方法建立原位肝癌小鼠模型。通过对三个rna测序数据集(TCGA-LIHC肿瘤与正常组织,Huh7放射耐药与亲本细胞,CAF与对照组)的综合分析,利用综合生物信息学方法确定了CAF相关的预后基因。通过实时定量PCR、western blot、免疫组织化学、细胞活力测定和菌落形成测定进行实验验证。结果:我们的研究结果表明,CAFs可显著增强HCC的放射耐药。基于13个ca相关的预后基因,通过共识聚类将TCGA-LIHC患者分为两个不同的簇,在总生存、免疫细胞浸润和治疗反应方面存在显著差异。结合三个中心基因和临床特征的预后图被开发。值得注意的是,SOX4在肿瘤组织、放射耐药细胞和ca暴露的HCC细胞中表达上调,与预后不良相关。SOX4敲除抑制HCC增殖并逆转钙诱导的放射耐药。此外,我们构建了LINC00665/miR-122-5p/SOX4竞争性内源性RNA (ceRNA)网络。结论:肝细胞癌是肝细胞癌放射耐药的重要介质,肝细胞癌相关基因为预后和治疗提供了有价值的见解。SOX4有望成为提高肝癌放疗疗效的治疗靶点。临床试验号:不适用。
{"title":"SOX4 mediates cancer-associated fibroblasts related radioresistance in hepatocellular carcinoma.","authors":"Ke Jiang, Botian Huang, Shasha He, Meiyan Zhu, Xiao Zhao, Miaowen Liu, Zhenwei Peng, Yan Wang, Yong Chen","doi":"10.1007/s13402-025-01088-z","DOIUrl":"10.1007/s13402-025-01088-z","url":null,"abstract":"<p><strong>Purpose: </strong>Cancer-associated fibroblasts (CAFs), a crucial component of tumor microenvironment, play a critical role in tumorigenesis, progression, and conferring resistance to radiotherapy and chemotherapy. This study aimed to investigate the association between CAFs, CAF- related genes, and radioresistance in hepatocellular carcinoma (HCC).</p><p><strong>Methods: </strong>CAFs were isolated from HCC tissues and subsequently utilized for co-culturing with HCC cells using CAFs-conditioned medium. An orthotopic HCC mouse model was established by co-implanting CAFs and HCC cells. Through integrative analysis of three RNA-sequencing datasets (TCGA-LIHC tumor vs. normal tissues, Huh7 radioresistant vs. parent cells, and CAF vs. control group), CAF-associated prognostic genes were identified using comprehensive bioinformatics approaches. Experimental validation was performed by real-time quantitative PCR, western blot, immunohistochemistry, cell viability assays, and colony formation assays.</p><p><strong>Results: </strong>Our findings demonstrated that CAFs significantly enhance radioresistance in HCC. Based on 13 CAF-related prognostic genes, TCGA-LIHC patients were stratified into two distinct clusters via consensus clustering, exhibiting significant differences in overall survival, immune cell infiltration, and therapeutic response. A prognostic nomogram incorporating three hub genes and clinical characteristics was developed. Notably, SOX4 was upregulated in tumor tissues, radioresistant cells, and CAF-exposed HCC cells, correlating with poor prognosis. SOX4 knockdown suppressed HCC proliferation and reversed CAF-induced radioresistance. Additionally, a competitive endogenous RNA (ceRNA) network of LINC00665/miR-122-5p/SOX4 was constructed.</p><p><strong>Conclusion: </strong>CAFs serve as crucial mediators of radioresistance in HCC, and CAF-related genes provide valuable prognostic and therapeutic insights. SOX4 emerges as a promising therapeutic target to improve radiotherapy efficacy in HCC.</p><p><strong>Clinical trial number: </strong>Not applicable.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1449-1463"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528248/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741309","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Molecular stratification of prostate cancer through sensory perception-related multi-omics analysis reveals chemoresistant mechanisms. 通过感觉感知相关的多组学分析,前列腺癌分子分层揭示了化疗耐药机制。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-08-19 DOI: 10.1007/s13402-025-01099-w
Shi Li, Ye Tian, Yu Sun, Tian Xu, Wei Wang

Background: Advanced prostate cancer (PCa) displays significant genetic heterogeneity and therapy resistance, yet the role of sensory perception pathways in its progression remains unclear.

Methods: We performed an integrative multi-omics analysis of sensory perception-linked mRNAs and lncRNAs from TCGA and scRNA-seq data. Unsupervised consensus clustering defined three molecular subtypes (CS1-CS3). Key biomarkers were validated in patient tissues and serum. Immune and stromal infiltration were quantified using TIDE and ESTIMATE. Single-cell trajectories characterized TSC22D3-positive T cells, and NicheNet mapped ligand-receptor interactions.

Results: Three subtypes emerged, with CS1 showing the poorest prognosis, marked chemotherapy resistance, and pronounced stromal-immune crosstalk. CS1 tumors exhibited elevated B- and T-cell infiltration and increased oxidative phosphorylation in TSC22D3-positive T cells. NicheNet analysis identified the TNF-CCL20 axis as a central mediator of immunosuppressive signaling and chemoresistance in CS1.

Conclusions: This study establishes sensory perception-associated molecular subtypes in PCa and links CS1 chemoresistance to immune microenvironment reprogramming via TNF-CCL20 signaling. These findings offer mechanistic insights into PCa progression and suggest actionable targets to overcome therapeutic resistance.

背景:晚期前列腺癌(PCa)表现出明显的遗传异质性和治疗耐药性,但感觉知觉通路在其进展中的作用尚不清楚。方法:我们对来自TCGA和scRNA-seq数据的感觉感知相关mrna和lncrna进行了综合多组学分析。无监督一致聚类定义了三种分子亚型(CS1-CS3)。在患者组织和血清中验证了关键生物标志物。采用TIDE和ESTIMATE定量免疫和间质浸润。单细胞轨迹表征了tsc22d3阳性T细胞,NicheNet绘制了配体与受体的相互作用。结果:出现三种亚型,其中CS1预后最差,化疗耐药明显,基质免疫串扰明显。CS1肿瘤表现为B细胞和T细胞浸润升高,tsc22d3阳性T细胞氧化磷酸化升高。NicheNet分析发现TNF-CCL20轴是CS1免疫抑制信号传导和化疗耐药的中心介质。结论:本研究在前列腺癌中建立了感觉感知相关的分子亚型,并通过TNF-CCL20信号传导将CS1化疗耐药与免疫微环境重编程联系起来。这些发现提供了前列腺癌进展的机制见解,并提出了克服治疗耐药的可行靶点。
{"title":"Molecular stratification of prostate cancer through sensory perception-related multi-omics analysis reveals chemoresistant mechanisms.","authors":"Shi Li, Ye Tian, Yu Sun, Tian Xu, Wei Wang","doi":"10.1007/s13402-025-01099-w","DOIUrl":"10.1007/s13402-025-01099-w","url":null,"abstract":"<p><strong>Background: </strong>Advanced prostate cancer (PCa) displays significant genetic heterogeneity and therapy resistance, yet the role of sensory perception pathways in its progression remains unclear.</p><p><strong>Methods: </strong>We performed an integrative multi-omics analysis of sensory perception-linked mRNAs and lncRNAs from TCGA and scRNA-seq data. Unsupervised consensus clustering defined three molecular subtypes (CS1-CS3). Key biomarkers were validated in patient tissues and serum. Immune and stromal infiltration were quantified using TIDE and ESTIMATE. Single-cell trajectories characterized TSC22D3-positive T cells, and NicheNet mapped ligand-receptor interactions.</p><p><strong>Results: </strong>Three subtypes emerged, with CS1 showing the poorest prognosis, marked chemotherapy resistance, and pronounced stromal-immune crosstalk. CS1 tumors exhibited elevated B- and T-cell infiltration and increased oxidative phosphorylation in TSC22D3-positive T cells. NicheNet analysis identified the TNF-CCL20 axis as a central mediator of immunosuppressive signaling and chemoresistance in CS1.</p><p><strong>Conclusions: </strong>This study establishes sensory perception-associated molecular subtypes in PCa and links CS1 chemoresistance to immune microenvironment reprogramming via TNF-CCL20 signaling. These findings offer mechanistic insights into PCa progression and suggest actionable targets to overcome therapeutic resistance.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1609-1614"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528230/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144871677","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Revealing genetic drivers of ovarian cancer and chemoresistance: insights from whole-genome CRISPR-knockout library screens. 揭示卵巢癌和化疗耐药的遗传驱动因素:来自全基因组crispr敲除文库筛选的见解。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-08-28 DOI: 10.1007/s13402-025-01102-4
Tali S Skipper, Kristie-Ann Dickson, Christopher E Denes, Matthew A Waller, Tian Y Du, G Gregory Neely, Nikola A Bowden, Alen Faiz, Deborah J Marsh

Understanding genetic dependencies in cancer is key to identifying novel actionable drug targets to advance precision medicine. Whole-genome CRISPR-knockout library screening methods have facilitated this goal. Pooled libraries of single guide RNAs (sgRNAs) targeting over 90% of the annotated protein coding genome are used to induce gene knockouts in pre-clinical cancer models. Novel genes of interest are identified by evaluating sgRNA dropout or enrichment following selection pressure application. This method is particularly beneficial for researching cancers where effective treatment strategies are limited. One example of a commonly chemoresistant cancer, particularly at relapse, is the low survival malignancy epithelial ovarian cancer (EOC), made up of multiple histotypes with distinct molecular profiles. CRISPR-knockout library screens in pre-clinical EOC models have demonstrated the ability to predict biomarkers of treatment response, identify targets synergistic with standard-of-care chemotherapy, and determine novel actionable targets which are synthetic lethal with cancer-associated mutations. Robust experimental design of CRISPR-knockout library screens, including the selection of strong pre-clinical cell line models, allows for meaningful conclusions to be made. We discuss essential design criteria for the use of CRISPR-knockout library screens to discover genetic dependencies in cancer and draw attention to discoveries with translational potential for EOC.

了解癌症的遗传依赖性是确定新的可操作药物靶点以推进精准医疗的关键。全基因组crispr敲除文库筛选方法促进了这一目标。针对超过90%的注释蛋白编码基因组的单导rna (sgRNAs)汇集文库用于在临床前癌症模型中诱导基因敲除。新的感兴趣的基因是通过评估sgRNA辍学或富集后的选择压力应用鉴定。这种方法特别有利于研究有效治疗策略有限的癌症。低生存率恶性上皮性卵巢癌(EOC)是一种常见的化疗耐药癌症,特别是在复发时,它由具有不同分子谱的多种组织型组成。临床前EOC模型中的crispr敲除文库筛选已经证明能够预测治疗反应的生物标志物,识别与标准治疗化疗协同作用的靶标,并确定具有癌症相关突变的合成致死的新型可操作靶标。crispr敲除文库筛选的稳健实验设计,包括选择强大的临床前细胞系模型,可以得出有意义的结论。我们讨论了使用crispr敲除文库筛选来发现癌症中的遗传依赖性的基本设计标准,并提请注意具有EOC翻译潜力的发现。
{"title":"Revealing genetic drivers of ovarian cancer and chemoresistance: insights from whole-genome CRISPR-knockout library screens.","authors":"Tali S Skipper, Kristie-Ann Dickson, Christopher E Denes, Matthew A Waller, Tian Y Du, G Gregory Neely, Nikola A Bowden, Alen Faiz, Deborah J Marsh","doi":"10.1007/s13402-025-01102-4","DOIUrl":"10.1007/s13402-025-01102-4","url":null,"abstract":"<p><p>Understanding genetic dependencies in cancer is key to identifying novel actionable drug targets to advance precision medicine. Whole-genome CRISPR-knockout library screening methods have facilitated this goal. Pooled libraries of single guide RNAs (sgRNAs) targeting over 90% of the annotated protein coding genome are used to induce gene knockouts in pre-clinical cancer models. Novel genes of interest are identified by evaluating sgRNA dropout or enrichment following selection pressure application. This method is particularly beneficial for researching cancers where effective treatment strategies are limited. One example of a commonly chemoresistant cancer, particularly at relapse, is the low survival malignancy epithelial ovarian cancer (EOC), made up of multiple histotypes with distinct molecular profiles. CRISPR-knockout library screens in pre-clinical EOC models have demonstrated the ability to predict biomarkers of treatment response, identify targets synergistic with standard-of-care chemotherapy, and determine novel actionable targets which are synthetic lethal with cancer-associated mutations. Robust experimental design of CRISPR-knockout library screens, including the selection of strong pre-clinical cell line models, allows for meaningful conclusions to be made. We discuss essential design criteria for the use of CRISPR-knockout library screens to discover genetic dependencies in cancer and draw attention to discoveries with translational potential for EOC.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1245-1265"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528352/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944370","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
RACK1 attenuates pancreatic tumorigenesis by suppressing acinar-to-ductal metaplasia through inflammatory signaling modulation. RACK1通过炎症信号调节抑制腺泡到导管的化生,从而减轻胰腺肿瘤的发生。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-09-01 DOI: 10.1007/s13402-025-01084-3
Wei Zhang, Tingting Jiang, Huiqing Zhang, Fang Wei, Xiaojia Li, Keping Xie

Purpose: Pancreatic ductal adenocarcinoma (PDA) remains one of the most lethal malignancies with limited early diagnostic and therapeutic options. Although receptor for activated C kinase 1 (RACK1) is an evolutionarily conserved scaffold protein, its functional role and mechanistic involvement in PDA pathogenesis remain elusive.

Methods: Using multimodal approaches including: (1) genetically engineered mouse models of pancreatitis and carcinogenesis, (2) patient-derived PDA tissues with matched normal specimens, (3) primary acinar cell 3D cultures, and (4) orthogonal gain/loss-of-function assays in PDA cell lines, we systematically investigated RACK1's spatiotemporal expression patterns and functional impacts. Mechanistic dissection was performed through gene expression profiling and pathway enrichment analyses with functional validation.

Results: RACK1 exhibited progressive silencing across pancreatic lesion progression: acinar cells (normal) > ADM > pancreatic intraepithelial neoplasia (PanIN) > PDA. This acinar-specific protein was undetectable in ductal/islet lineages and was further suppressed under inflammatory challenge. Functionally, RACK1 depletion accelerated ADM initiation and enhanced PDA cell motility and metastatic dissemination in vivo, whereas its overexpression exerted tumor-suppressive effects. Mechanistically, caerulein/TGF-α stimulation and KrasG12D activation converged to inhibit RACK1 while activating MAP2K3-SRC-RELA(p65) signaling nodes, establishing a pro-inflammatory feedforward loop.

Conclusions: RACK1 serves as a gatekeeper restraining inflammation-driven ADM transformation, with its downregulation constituting an early molecular event in PDA pathogenesis. The RACK1-MAP2K3 axis orchestrates malignant transition through simultaneous NF-κB activation (inflammatory priming) and MAPK hyperactivation (proliferative drive). Our findings nominate RACK1 as both a stratification biomarker for high-risk pancreatic lesions and a druggable node for intercepting preneoplastic progression.

目的:胰腺导管腺癌(PDA)仍然是最致命的恶性肿瘤之一,早期诊断和治疗选择有限。尽管活化C激酶1受体(RACK1)是一种进化上保守的支架蛋白,但其在PDA发病机制中的功能作用和机制参与尚不清楚。方法:采用多模式方法,包括:(1)基因工程小鼠胰腺炎和癌变模型,(2)患者来源的PDA组织与匹配的正常标本,(3)原代腺泡细胞3D培养,(4)PDA细胞系的正交增益/功能丧失试验,系统地研究了RACK1的时空表达模式和功能影响。通过基因表达谱和途径富集分析进行机制解剖,并进行功能验证。结果:RACK1在胰腺病变进程中表现出进行性沉默:腺泡细胞(正常)> ADM >胰腺上皮内瘤变(PanIN) > PDA。这种腺泡特异性蛋白在导管/胰岛谱系中检测不到,并且在炎症挑战下进一步抑制。在功能上,RACK1缺失加速了ADM的启动,增强了PDA细胞在体内的运动和转移传播,而其过表达则具有肿瘤抑制作用。机制上,caerulein/TGF-α刺激和KrasG12D激活融合抑制RACK1,同时激活MAP2K3-SRC-RELA(p65)信号节点,建立促炎前馈回路。结论:RACK1在抑制炎症驱动的ADM转化中起着守门人的作用,其下调是PDA发病的早期分子事件。RACK1-MAP2K3轴通过同时激活NF-κB(炎症启动)和MAPK过度激活(增殖驱动)来协调恶性转移。我们的研究结果表明,RACK1既是高风险胰腺病变的分层生物标志物,也是阻断肿瘤前进展的可用药节点。
{"title":"RACK1 attenuates pancreatic tumorigenesis by suppressing acinar-to-ductal metaplasia through inflammatory signaling modulation.","authors":"Wei Zhang, Tingting Jiang, Huiqing Zhang, Fang Wei, Xiaojia Li, Keping Xie","doi":"10.1007/s13402-025-01084-3","DOIUrl":"10.1007/s13402-025-01084-3","url":null,"abstract":"<p><strong>Purpose: </strong>Pancreatic ductal adenocarcinoma (PDA) remains one of the most lethal malignancies with limited early diagnostic and therapeutic options. Although receptor for activated C kinase 1 (RACK1) is an evolutionarily conserved scaffold protein, its functional role and mechanistic involvement in PDA pathogenesis remain elusive.</p><p><strong>Methods: </strong>Using multimodal approaches including: (1) genetically engineered mouse models of pancreatitis and carcinogenesis, (2) patient-derived PDA tissues with matched normal specimens, (3) primary acinar cell 3D cultures, and (4) orthogonal gain/loss-of-function assays in PDA cell lines, we systematically investigated RACK1's spatiotemporal expression patterns and functional impacts. Mechanistic dissection was performed through gene expression profiling and pathway enrichment analyses with functional validation.</p><p><strong>Results: </strong>RACK1 exhibited progressive silencing across pancreatic lesion progression: acinar cells (normal) > ADM > pancreatic intraepithelial neoplasia (PanIN) > PDA. This acinar-specific protein was undetectable in ductal/islet lineages and was further suppressed under inflammatory challenge. Functionally, RACK1 depletion accelerated ADM initiation and enhanced PDA cell motility and metastatic dissemination in vivo, whereas its overexpression exerted tumor-suppressive effects. Mechanistically, caerulein/TGF-α stimulation and Kras<sup>G12D</sup> activation converged to inhibit RACK1 while activating MAP2K3-SRC-RELA(p65) signaling nodes, establishing a pro-inflammatory feedforward loop.</p><p><strong>Conclusions: </strong>RACK1 serves as a gatekeeper restraining inflammation-driven ADM transformation, with its downregulation constituting an early molecular event in PDA pathogenesis. The RACK1-MAP2K3 axis orchestrates malignant transition through simultaneous NF-κB activation (inflammatory priming) and MAPK hyperactivation (proliferative drive). Our findings nominate RACK1 as both a stratification biomarker for high-risk pancreatic lesions and a druggable node for intercepting preneoplastic progression.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1395-1411"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528279/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944286","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
OTUD1 downregulates PD-L1 expression by deubiquitinating STAT3 and promotes the immune response in ccRCC. OTUD1通过去泛素化STAT3下调PD-L1表达,促进CcRCC的免疫应答。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-06-12 DOI: 10.1007/s13402-025-01079-0
Huaiyuan Liang, Xinlin Liu, Wanyang Guo, Wei Xiong, Da Ren, Wentao Liu

Background: Clear cell renal cell carcinoma (ccRCC) is the most common malignant tumor of the urinary system and has the highest mortality rate. In addition to surgical tumor reduction, systemic drug therapy is the most important treatment method for metastatic renal cancer. In recent years, immunotherapeutic drugs represented by PD-1 antibodies have been used in the treatment of metastatic ccRCC and achieved good therapeutic effects. However, due to the occurrence of immune escape, only about 50-60% of patients with advanced renal cancer can significantly benefit from immunotherapy. The mechanism of immune escape is extremely complex and has not been clarified. We intend to delve into the driving mechanisms of immune evasion in ccRCC and explore potential targets for intervention.

Methods: In this study, we analyzed the expression of OTUD1 and related pathways in ccRCC through TCGA, GEO dataset, and cBioPortal web tool. At the same time, a mouse model of allogeneic transplanted clear cell renal cell carcinoma was constructed, and the effect of OTUD1 on anti-PD1 antibody therapy was discussed. Experiments such as co-IP, flow cytometry, and RNA-seq analysis were used to investigate the mechanism by which OTUD1 regulates immunity through STAT3.

Results: This study reveals that OTUD1 suppresses PD-L1 expression and enhances antitumor immunity in clear cell renal cell carcinoma (ccRCC) by deubiquitinating and stabilizing STAT3, thereby inhibiting its nuclear translocation and transcriptional activity. As a key regulator of the JAK-STAT pathway, OTUD1 disrupts PD-1/PD-L1-mediated immune evasion, offering a potential therapeutic strategy to improve immunotherapy responses in ccRCC. These findings highlight the OTUD1-STAT3-PD-L1 axis as a novel mechanism for overcoming immune checkpoint resistance.

Conclusion: Overall, we demonstrate that OTUD1 interacts with STAT3 and deubiquitinates, inhibits its nuclear translocation and activity, and ultimately inhibits immune evasion of ccRCC by downregulating PD-L1.

背景:透明细胞肾细胞癌(ccRCC)是泌尿系统最常见的恶性肿瘤,死亡率最高。除手术减瘤外,全身药物治疗是转移性肾癌最重要的治疗方法。近年来,以PD-1抗体为代表的免疫治疗药物已被用于转移性ccRCC的治疗,并取得了良好的治疗效果。然而,由于免疫逃逸的发生,只有约50-60%的晚期肾癌患者能从免疫治疗中明显获益。免疫逃逸的机制极其复杂,目前尚未明确。我们打算深入研究ccRCC中免疫逃避的驱动机制,并探索潜在的干预靶点。方法:本研究通过TCGA、GEO数据集和cBioPortal网络工具分析OTUD1及其相关通路在ccRCC中的表达。同时构建小鼠同种异体移植透明细胞肾细胞癌模型,探讨OTUD1对抗pd1抗体治疗的作用。通过co-IP、流式细胞术、RNA-seq分析等实验,探讨OTUD1通过STAT3调控免疫的机制。结果:本研究发现OTUD1通过去泛素化和稳定STAT3,抑制透明细胞肾细胞癌(clear cell renal cell carcinoma, ccRCC)中PD-L1的表达,增强抗肿瘤免疫,从而抑制STAT3的核易位和转录活性。作为JAK-STAT通路的关键调节因子,OTUD1破坏PD-1/ pd - l1介导的免疫逃避,为改善ccRCC的免疫治疗反应提供了一种潜在的治疗策略。这些发现突出了OTUD1-STAT3-PD-L1轴作为克服免疫检查点抵抗的新机制。结论:总的来说,我们证明OTUD1与STAT3和去泛素化相互作用,抑制其核易位和活性,最终通过下调PD-L1抑制ccRCC的免疫逃避。
{"title":"OTUD1 downregulates PD-L1 expression by deubiquitinating STAT3 and promotes the immune response in ccRCC.","authors":"Huaiyuan Liang, Xinlin Liu, Wanyang Guo, Wei Xiong, Da Ren, Wentao Liu","doi":"10.1007/s13402-025-01079-0","DOIUrl":"10.1007/s13402-025-01079-0","url":null,"abstract":"<p><strong>Background: </strong>Clear cell renal cell carcinoma (ccRCC) is the most common malignant tumor of the urinary system and has the highest mortality rate. In addition to surgical tumor reduction, systemic drug therapy is the most important treatment method for metastatic renal cancer. In recent years, immunotherapeutic drugs represented by PD-1 antibodies have been used in the treatment of metastatic ccRCC and achieved good therapeutic effects. However, due to the occurrence of immune escape, only about 50-60% of patients with advanced renal cancer can significantly benefit from immunotherapy. The mechanism of immune escape is extremely complex and has not been clarified. We intend to delve into the driving mechanisms of immune evasion in ccRCC and explore potential targets for intervention.</p><p><strong>Methods: </strong>In this study, we analyzed the expression of OTUD1 and related pathways in ccRCC through TCGA, GEO dataset, and cBioPortal web tool. At the same time, a mouse model of allogeneic transplanted clear cell renal cell carcinoma was constructed, and the effect of OTUD1 on anti-PD1 antibody therapy was discussed. Experiments such as co-IP, flow cytometry, and RNA-seq analysis were used to investigate the mechanism by which OTUD1 regulates immunity through STAT3.</p><p><strong>Results: </strong>This study reveals that OTUD1 suppresses PD-L1 expression and enhances antitumor immunity in clear cell renal cell carcinoma (ccRCC) by deubiquitinating and stabilizing STAT3, thereby inhibiting its nuclear translocation and transcriptional activity. As a key regulator of the JAK-STAT pathway, OTUD1 disrupts PD-1/PD-L1-mediated immune evasion, offering a potential therapeutic strategy to improve immunotherapy responses in ccRCC. These findings highlight the OTUD1-STAT3-PD-L1 axis as a novel mechanism for overcoming immune checkpoint resistance.</p><p><strong>Conclusion: </strong>Overall, we demonstrate that OTUD1 interacts with STAT3 and deubiquitinates, inhibits its nuclear translocation and activity, and ultimately inhibits immune evasion of ccRCC by downregulating PD-L1.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1349-1361"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528312/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282551","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
METTL1-WDR4 promotes the migration and proliferation of gastric cancer through N7-methylguanosine. METTL1-WDR4通过n7 -甲基鸟苷促进胃癌的迁移和增殖。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-10-01 Epub Date: 2025-08-05 DOI: 10.1007/s13402-025-01094-1
Jiaqi Wang, Kai Zhou, Tao He, Qihang Hu, Jianquan Liu, Pingang Li, Yan Shi, Jun Song, Peiwu Yu

Background: Gastric cancer (GC) is one of the most common malignant tumor worldwide. Metastasis is leading cases of cancer-related death of GC. It has been found that N7-methylguanosine (m7G) modifications play an important role in cancer. However, the role of m7G modifications within mRNA and its "writer" METTL1 and WDR4 in tumors, particularly GC, has not been revealed.

Methods: RT-qPCR, WB and IHC were used to detect the expression of METTL1 and WDR4 in GC cells and tissues. Function-based experiments were performed using METTL1-WDR4 knockdown and overexpression cell lines in vitro and in vivo, including CCK8, colony formation, transwell and nude mice models. Mechanistically, RNA-seq, MeRIP-seq, MeRIP-qPCR, western blot, dot blot, co-IP, ChIP and IHC stainings were performed.

Results: METTL1 and WDR4 were upregulated in GC patients. High expression of METTL1 and WDR4 were associated with poor prognosis. Silencing METTL1-WDR4 inhibited GC cell migration and proliferation in vitro and vivo. Mechanistically, METTL1-WDR4 can enhance the mRNA stability of PIK3C2B and AKT by promoting their m7G levels, which leading the overexpression of p-AKT. Interestingly, we also found that on the one hand, the transcription factor YY1 can promote the mRNA transcription expression of METTL1 and WDR4 at the same time, and on the other hand, METTL1-WDR4 can promote YY1 expression by increasing the level of m7G. This regulation presents positive feedback. Above all, METTL1 and WDR4 ultimately up-regulate the level of m7G and promote the malignant progression of GC.

Conclusions: These findings suggest that METTL1-WDR4 might serve as a potential diagnostic and prognostic biomarker and a therapeutic target for GC treatment by regulating m7G level.

背景:胃癌是世界范围内最常见的恶性肿瘤之一。转移是胃癌相关死亡的主要原因。已经发现n7 -甲基鸟苷(m7G)修饰在癌症中起重要作用。然而,m7G修饰及其“撰写者”METTL1和WDR4在肿瘤,特别是胃癌中的作用尚未被揭示。方法:采用RT-qPCR、WB和IHC检测胃癌细胞和组织中METTL1和WDR4的表达。基于功能的实验采用METTL1-WDR4敲除和过表达细胞系进行体外和体内实验,包括CCK8、集落形成、transwell和裸鼠模型。机械染色:RNA-seq、MeRIP-seq、MeRIP-qPCR、western blot、dot blot、co-IP、ChIP和IHC染色。结果:METTL1和WDR4在胃癌患者中表达上调。METTL1和WDR4的高表达与预后不良相关。沉默METTL1-WDR4可抑制GC细胞的迁移和增殖。从机制上讲,METTL1-WDR4可以通过提高PIK3C2B和AKT的m7G水平来增强其mRNA的稳定性,从而导致p-AKT的过表达。有趣的是,我们还发现转录因子YY1一方面可以同时促进METTL1和WDR4的mRNA转录表达,另一方面,METTL1-WDR4可以通过增加m7G的水平来促进YY1的表达。这一规定带来了积极的反馈。综上所述,METTL1和WDR4最终上调m7G水平,促进GC的恶性进展。结论:这些发现提示METTL1-WDR4可能通过调节m7G水平作为胃癌的潜在诊断和预后生物标志物和治疗靶点。
{"title":"METTL1-WDR4 promotes the migration and proliferation of gastric cancer through N<sup>7</sup>-methylguanosine.","authors":"Jiaqi Wang, Kai Zhou, Tao He, Qihang Hu, Jianquan Liu, Pingang Li, Yan Shi, Jun Song, Peiwu Yu","doi":"10.1007/s13402-025-01094-1","DOIUrl":"10.1007/s13402-025-01094-1","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) is one of the most common malignant tumor worldwide. Metastasis is leading cases of cancer-related death of GC. It has been found that N<sup>7</sup>-methylguanosine (m7G) modifications play an important role in cancer. However, the role of m7G modifications within mRNA and its \"writer\" METTL1 and WDR4 in tumors, particularly GC, has not been revealed.</p><p><strong>Methods: </strong>RT-qPCR, WB and IHC were used to detect the expression of METTL1 and WDR4 in GC cells and tissues. Function-based experiments were performed using METTL1-WDR4 knockdown and overexpression cell lines in vitro and in vivo, including CCK8, colony formation, transwell and nude mice models. Mechanistically, RNA-seq, MeRIP-seq, MeRIP-qPCR, western blot, dot blot, co-IP, ChIP and IHC stainings were performed.</p><p><strong>Results: </strong>METTL1 and WDR4 were upregulated in GC patients. High expression of METTL1 and WDR4 were associated with poor prognosis. Silencing METTL1-WDR4 inhibited GC cell migration and proliferation in vitro and vivo. Mechanistically, METTL1-WDR4 can enhance the mRNA stability of PIK3C2B and AKT by promoting their m7G levels, which leading the overexpression of p-AKT. Interestingly, we also found that on the one hand, the transcription factor YY1 can promote the mRNA transcription expression of METTL1 and WDR4 at the same time, and on the other hand, METTL1-WDR4 can promote YY1 expression by increasing the level of m7G. This regulation presents positive feedback. Above all, METTL1 and WDR4 ultimately up-regulate the level of m7G and promote the malignant progression of GC.</p><p><strong>Conclusions: </strong>These findings suggest that METTL1-WDR4 might serve as a potential diagnostic and prognostic biomarker and a therapeutic target for GC treatment by regulating m7G level.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1523-1537"},"PeriodicalIF":4.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12528236/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144788370","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
期刊
Cellular 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