首页 > 最新文献

Cancer Cell International最新文献

英文 中文
CUL2 confers ferroptosis resistance in pancreatic cancer by disrupting KEAP1-mediated NRF2 degradation. CUL2通过破坏keap1介导的NRF2降解,在胰腺癌中赋予铁凋亡抗性。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-16 DOI: 10.1186/s12935-025-04005-4
Kui Yu, LiLi Chen, Yong Zhong, Yang Wang, Na Zhu, Daoheng Wang, Wenyan Yao, Haoran Bai, Lijuan Yang, Dachuan Qi

Background: Pancreatic cancer (PC) is characterized by high chemoresistance and poor prognosis. CUL2, a scaffold protein of E3 ubiquitin ligases, has been implicated in tumor progression, but its role in PC remains unclear.

Methods: CUL2 expression was analyzed in PC tissues and cell lines using TCGA and GEO datasets, qRT-PCR, Western blot, and immunohistochemistry analysis. The biological functions of CUL2 were investigated through gain- and loss-of-function studies in vitro. The impact of CUL2 overexpression on gemcitabine sensitivity was evaluated in xenograft models. Molecular mechanisms were explored using proteasome inhibitor MG132, co-immunoprecipitation, subcellular fractionation, and ferroptosis assessment.

Results: CUL2 was significantly upregulated in PC tissues and correlated with poor prognosis. CUL2 promoted PC cell proliferation, migration, and EMT. Mechanistically, CUL2 competed with NRF2 for KEAP1 binding, thereby preventing KEAP1-mediated NRF2 degradation and promoting NRF2 nuclear translocation. CUL2 promoted oxidative stress while activating NRF2-dependent antioxidant response. The CUL2-NRF2 axis suppressed ferroptosis and conferred gemcitabine resistance in PC cells. In xenograft models, CUL2 overexpression enhanced tumor growth and attenuated gemcitabine sensitivity through NRF2-mediated ferroptosis inhibition.

Conclusions: Our findings reveal a novel mechanism whereby CUL2 promotes PC progression and ferroptosis resistance through regulation of the KEAP1-NRF2 axis. CUL2 overexpression enhances cellular antioxidant capacity and maintains mitochondrial integrity, thereby conferring broad resistance to ferroptosis-inducing conditions. This study suggests that targeting the CUL2-NRF2 axis to enhance ferroptosis sensitivity might represent a promising therapeutic strategy for PC treatment.

背景:胰腺癌(PC)具有化疗耐药高、预后差的特点。CUL2是E3泛素连接酶的一种支架蛋白,与肿瘤进展有关,但其在PC中的作用尚不清楚。方法:采用TCGA和GEO数据集、qRT-PCR、Western blot和免疫组织化学分析CUL2在PC组织和细胞系中的表达。CUL2的生物学功能通过体外功能获得和功能丧失研究进行了研究。在异种移植模型中评估CUL2过表达对吉西他滨敏感性的影响。使用蛋白酶体抑制剂MG132、共免疫沉淀、亚细胞分离和铁下垂评估来探索分子机制。结果:CUL2在PC组织中表达明显上调,且与预后不良相关。CUL2促进PC细胞增殖、迁移和EMT。在机制上,CUL2与NRF2竞争KEAP1结合,从而阻止KEAP1介导的NRF2降解,促进NRF2核易位。CUL2促进氧化应激,同时激活nrf2依赖的抗氧化反应。CUL2-NRF2轴抑制铁下垂并赋予PC细胞吉西他滨耐药性。在异种移植物模型中,CUL2过表达通过nrf2介导的铁下垂抑制促进肿瘤生长并减弱吉西他滨敏感性。结论:我们的研究结果揭示了CUL2通过调节KEAP1-NRF2轴促进PC进展和铁下垂抵抗的新机制。CUL2过表达增强细胞抗氧化能力并维持线粒体完整性,从而赋予对铁中毒诱导条件的广泛抗性。这项研究表明,靶向CUL2-NRF2轴增强铁下垂敏感性可能是一种很有前景的治疗PC的治疗策略。
{"title":"CUL2 confers ferroptosis resistance in pancreatic cancer by disrupting KEAP1-mediated NRF2 degradation.","authors":"Kui Yu, LiLi Chen, Yong Zhong, Yang Wang, Na Zhu, Daoheng Wang, Wenyan Yao, Haoran Bai, Lijuan Yang, Dachuan Qi","doi":"10.1186/s12935-025-04005-4","DOIUrl":"https://doi.org/10.1186/s12935-025-04005-4","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic cancer (PC) is characterized by high chemoresistance and poor prognosis. CUL2, a scaffold protein of E3 ubiquitin ligases, has been implicated in tumor progression, but its role in PC remains unclear.</p><p><strong>Methods: </strong>CUL2 expression was analyzed in PC tissues and cell lines using TCGA and GEO datasets, qRT-PCR, Western blot, and immunohistochemistry analysis. The biological functions of CUL2 were investigated through gain- and loss-of-function studies in vitro. The impact of CUL2 overexpression on gemcitabine sensitivity was evaluated in xenograft models. Molecular mechanisms were explored using proteasome inhibitor MG132, co-immunoprecipitation, subcellular fractionation, and ferroptosis assessment.</p><p><strong>Results: </strong>CUL2 was significantly upregulated in PC tissues and correlated with poor prognosis. CUL2 promoted PC cell proliferation, migration, and EMT. Mechanistically, CUL2 competed with NRF2 for KEAP1 binding, thereby preventing KEAP1-mediated NRF2 degradation and promoting NRF2 nuclear translocation. CUL2 promoted oxidative stress while activating NRF2-dependent antioxidant response. The CUL2-NRF2 axis suppressed ferroptosis and conferred gemcitabine resistance in PC cells. In xenograft models, CUL2 overexpression enhanced tumor growth and attenuated gemcitabine sensitivity through NRF2-mediated ferroptosis inhibition.</p><p><strong>Conclusions: </strong>Our findings reveal a novel mechanism whereby CUL2 promotes PC progression and ferroptosis resistance through regulation of the KEAP1-NRF2 axis. CUL2 overexpression enhances cellular antioxidant capacity and maintains mitochondrial integrity, thereby conferring broad resistance to ferroptosis-inducing conditions. This study suggests that targeting the CUL2-NRF2 axis to enhance ferroptosis sensitivity might represent a promising therapeutic strategy for PC treatment.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145767296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Co-delivery of camptothecin and SH3RF3-AS1 SiRNA for glioma synergistic therapy. 喜树碱与SH3RF3-AS1 SiRNA协同治疗胶质瘤。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-14 DOI: 10.1186/s12935-025-04119-9
Shun Wang, Zhaochao Nie, Lei Liang, Jing Zou, Zhiyue Zhang, Donghai Wang, Chuncheng Qu, Cheng Wang, Jun Jiang

Background: Current treatment strategies for high-grade gliomas have a limited overall patient benefit. Here, Methylated RNA immunoprecipitation sequencing (MeRIP-seq) of glioma was conducted to screen the lncRNA regulated by m6A modification in glioma, and provide an innovative chemotherapeutic strategy to improved the therapeutic efficacy.

Methods: To elucidate the impact and the molecular mechanism of ALKBH5/SH3RF3-AS1/SH3RF3 regulation on glioma malignancy, proliferation phenotypic assays, MeRIP-qPCR, RIP-qPCR and western blotting were used. Expressions and clinical significances of ALKBH5, SH3RF3-AS1, and SH3RF3 were assessed in tissue chip. Furthermore, we designed a nanodrug (NanoPCPT/siRNA@U251) for the targeted co-delivery of CPT (targeting type Ⅰ topoisomerase) and si-SH3RF3-AS1 (inhibiting SH3RF3, a Rac1-JNK activator) in glioma treatment.

Results: SH3RF3-AS1 was demethylated and overexpressed in glioma. We elucidated the mechanistic link between ALKBH5-mediated N6-methyladenosine (m6A) modification and SH3RF3-AS1 expression, and the cis-regulation of SH3RF3-AS1 to SH3RF3. Clinically, the ALKBH5, SH3RF3-AS1, and SH3RF3 expression increased with increasing glioma grade, high SH3RF3-AS1 expression correlated with unfavorable 5-year overall survival and progression-free survival in a cohort of 148 glioma patients. SH3RF3-AS1 drived glioma progression and promoted chemoresistance to camptothecin (CPT). We designed a nanodrug (NanoPCPT/siRNA@U251) for the targeted co-delivery of CPT (targeting type Ⅰ topoisomerase) and si-SH3RF3-AS1 (inhibiting SH3RF3, a Rac1-JNK activator) in glioma treatment, and it effectively suppressed glioma cell growth both in vivo and in vitro models.

Conclusion: Our findings provide valuable insights into the roles of m6A demethylation-mediated upregulation of SH3RF3-AS1 in supporting glioma proliferation and demonstrate the potential of NanoPCPT/siRNA@U251 as an innovative chemotherapeutic strategy for glioma.

背景:目前高级别胶质瘤的治疗策略对患者的总体获益有限。本研究通过胶质瘤的甲基化RNA免疫沉淀测序(MeRIP-seq),筛选胶质瘤中m6A修饰调控的lncRNA,为提高治疗效果提供创新的化疗策略。方法:通过增殖表型分析、MeRIP-qPCR、RIP-qPCR和western blotting分析ALKBH5/SH3RF3- as1 /SH3RF3调控对胶质瘤恶性肿瘤的影响及分子机制。组织芯片检测ALKBH5、SH3RF3- as1、SH3RF3的表达及临床意义。此外,我们设计了一种纳米药物(NanoPCPT/siRNA@U251),用于靶向共递送CPT(靶向Ⅰ型拓扑异构酶)和si-SH3RF3-AS1(抑制SH3RF3,一种Rac1-JNK激活剂)治疗胶质瘤。结果:SH3RF3-AS1在胶质瘤中去甲基化并过表达。我们阐明了alkbh5介导的n6 -甲基腺苷(m6A)修饰与SH3RF3- as1表达之间的机制联系,以及SH3RF3- as1对SH3RF3的顺式调控。在临床上,ALKBH5、SH3RF3- as1和SH3RF3的表达随着胶质瘤分级的增加而增加,在148例胶质瘤患者队列中,SH3RF3- as1的高表达与不利的5年总生存率和无进展生存率相关。SH3RF3-AS1驱动胶质瘤进展并促进喜树碱(CPT)的化疗耐药。我们设计了一种纳米药物(NanoPCPT/siRNA@U251),用于靶向共递送CPT(靶向Ⅰ型拓扑异构酶)和si-SH3RF3-AS1(抑制SH3RF3,一种Rac1-JNK激活剂)治疗胶质瘤,并在体内和体外模型中有效抑制胶质瘤细胞的生长。结论:我们的研究结果为m6A去甲基化介导的SH3RF3-AS1上调在支持胶质瘤增殖中的作用提供了有价值的见解,并证明了纳米cpt /siRNA@U251作为胶质瘤创新化疗策略的潜力。
{"title":"Co-delivery of camptothecin and SH3RF3-AS1 SiRNA for glioma synergistic therapy.","authors":"Shun Wang, Zhaochao Nie, Lei Liang, Jing Zou, Zhiyue Zhang, Donghai Wang, Chuncheng Qu, Cheng Wang, Jun Jiang","doi":"10.1186/s12935-025-04119-9","DOIUrl":"https://doi.org/10.1186/s12935-025-04119-9","url":null,"abstract":"<p><strong>Background: </strong>Current treatment strategies for high-grade gliomas have a limited overall patient benefit. Here, Methylated RNA immunoprecipitation sequencing (MeRIP-seq) of glioma was conducted to screen the lncRNA regulated by m6A modification in glioma, and provide an innovative chemotherapeutic strategy to improved the therapeutic efficacy.</p><p><strong>Methods: </strong>To elucidate the impact and the molecular mechanism of ALKBH5/SH3RF3-AS1/SH3RF3 regulation on glioma malignancy, proliferation phenotypic assays, MeRIP-qPCR, RIP-qPCR and western blotting were used. Expressions and clinical significances of ALKBH5, SH3RF3-AS1, and SH3RF3 were assessed in tissue chip. Furthermore, we designed a nanodrug (Nano<sup>PCPT</sup>/siRNA@U251) for the targeted co-delivery of CPT (targeting type Ⅰ topoisomerase) and si-SH3RF3-AS1 (inhibiting SH3RF3, a Rac1-JNK activator) in glioma treatment.</p><p><strong>Results: </strong>SH3RF3-AS1 was demethylated and overexpressed in glioma. We elucidated the mechanistic link between ALKBH5-mediated N6-methyladenosine (m6A) modification and SH3RF3-AS1 expression, and the cis-regulation of SH3RF3-AS1 to SH3RF3. Clinically, the ALKBH5, SH3RF3-AS1, and SH3RF3 expression increased with increasing glioma grade, high SH3RF3-AS1 expression correlated with unfavorable 5-year overall survival and progression-free survival in a cohort of 148 glioma patients. SH3RF3-AS1 drived glioma progression and promoted chemoresistance to camptothecin (CPT). We designed a nanodrug (Nano<sup>PCPT</sup>/siRNA@U251) for the targeted co-delivery of CPT (targeting type Ⅰ topoisomerase) and si-SH3RF3-AS1 (inhibiting SH3RF3, a Rac1-JNK activator) in glioma treatment, and it effectively suppressed glioma cell growth both in vivo and in vitro models.</p><p><strong>Conclusion: </strong>Our findings provide valuable insights into the roles of m6A demethylation-mediated upregulation of SH3RF3-AS1 in supporting glioma proliferation and demonstrate the potential of Nano<sup>PCPT</sup>/siRNA@U251 as an innovative chemotherapeutic strategy for glioma.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145751561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LAMC2 promotes gallbladder carcinoma metastasis through the TGF-β pathway. LAMC2通过TGF-β途径促进胆囊癌转移。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-14 DOI: 10.1186/s12935-025-04123-z
Mengyou Xu, Yuanyuan Zhang, Huibo Feng, Taiyu Shang, Yibin Chen, Erdong Liu, Yunkai Lin, Yufei Pan, Yanan Zhou, Xiaofan Feng, Liwei Dong, Tianyi Jiang, Hongyang Wang

Background: Gallbladder cancer (GBC) is an aggressive malignant tumor that seriously threatens the survival of GBC patients. Laminin-γ2 (LAMC2), an important component of laminins, has been reported to facilitate cancer development and chemoresistance in several cancers. However, the biological effect of LAMC2 on GBC is still unclear.

Methods: LAMC2 expression was assessed in GBC specimens through proteomics and RNA-seq data. To evaluate the biological effects of LAMC2, we performed colony formation, sphere formation, wound healing, and transwell and invasion assays, along with an in vivo metastasis model. Additionally, we also explored the role of LAMC2 in GBC organoid. Finally, Western blot and RT-qPCR were employed to identify the molecular mechanism underlying LAMC2-mediated regulation of aggressive behavior in GBC.

Results: Here, we revealed that LAMC2 was overexpressed in GBC tissues and was positively correlated with poor patient outcomes. Furthermore, the genetic silencing of LAMC2 significantly inhibited GBC cell colony formation, sphere formation, and organoid growth. Additionally, LAMC2 deficiency impaired GBC cell metastasis in vitro and in vivo, accompanied by a reversal of the epithelial-mesenchymal transition (EMT) phenotype. Mechanistically, LAMC2 expression was induced by the canonical TGF-β/SMAD2 signaling pathway, and in turn, LAMC2 feedback enhanced the activity of TGF-β signaling by binding to TGF-β receptor II (TGF-βRII).

Conclusion: Our findings suggest that LAMC2 promotes GBC progression and metastasis, potentially by regulating the TGF-β signaling pathway. Therefore, LAMC2 could serve as a potential prognostic biomarker and therapeutic target for GBC.

背景:胆囊癌是一种侵袭性恶性肿瘤,严重威胁胆囊癌患者的生存。层粘连蛋白-γ - 2 (LAMC2)是层粘连蛋白的重要组成部分,已被报道在多种癌症中促进癌症的发展和化疗耐药。然而,LAMC2对GBC的生物学作用尚不清楚。方法:通过蛋白质组学和RNA-seq数据评估LAMC2在GBC标本中的表达。为了评估LAMC2的生物学效应,我们进行了菌落形成、球体形成、伤口愈合、转移和侵袭试验,并建立了体内转移模型。此外,我们还探讨了LAMC2在GBC类器官中的作用。最后,采用Western blot和RT-qPCR方法鉴定lamc2介导的GBC侵袭行为调控的分子机制。结果:在这里,我们发现LAMC2在GBC组织中过表达,并且与患者预后不良呈正相关。此外,LAMC2基因沉默显著抑制GBC细胞集落形成、球形成和类器官生长。此外,LAMC2缺陷在体外和体内都会损害GBC细胞转移,并伴有上皮-间质转化(EMT)表型的逆转。在机制上,LAMC2的表达是由典型的TGF-β/SMAD2信号通路诱导的,反过来,LAMC2反馈通过结合TGF-β受体II (TGF-β rii)增强TGF-β信号通路的活性。结论:本研究提示LAMC2可能通过调控TGF-β信号通路促进GBC的进展和转移。因此,LAMC2可以作为GBC的潜在预后生物标志物和治疗靶点。
{"title":"LAMC2 promotes gallbladder carcinoma metastasis through the TGF-β pathway.","authors":"Mengyou Xu, Yuanyuan Zhang, Huibo Feng, Taiyu Shang, Yibin Chen, Erdong Liu, Yunkai Lin, Yufei Pan, Yanan Zhou, Xiaofan Feng, Liwei Dong, Tianyi Jiang, Hongyang Wang","doi":"10.1186/s12935-025-04123-z","DOIUrl":"https://doi.org/10.1186/s12935-025-04123-z","url":null,"abstract":"<p><strong>Background: </strong>Gallbladder cancer (GBC) is an aggressive malignant tumor that seriously threatens the survival of GBC patients. Laminin-γ2 (LAMC2), an important component of laminins, has been reported to facilitate cancer development and chemoresistance in several cancers. However, the biological effect of LAMC2 on GBC is still unclear.</p><p><strong>Methods: </strong>LAMC2 expression was assessed in GBC specimens through proteomics and RNA-seq data. To evaluate the biological effects of LAMC2, we performed colony formation, sphere formation, wound healing, and transwell and invasion assays, along with an in vivo metastasis model. Additionally, we also explored the role of LAMC2 in GBC organoid. Finally, Western blot and RT-qPCR were employed to identify the molecular mechanism underlying LAMC2-mediated regulation of aggressive behavior in GBC.</p><p><strong>Results: </strong>Here, we revealed that LAMC2 was overexpressed in GBC tissues and was positively correlated with poor patient outcomes. Furthermore, the genetic silencing of LAMC2 significantly inhibited GBC cell colony formation, sphere formation, and organoid growth. Additionally, LAMC2 deficiency impaired GBC cell metastasis in vitro and in vivo, accompanied by a reversal of the epithelial-mesenchymal transition (EMT) phenotype. Mechanistically, LAMC2 expression was induced by the canonical TGF-β/SMAD2 signaling pathway, and in turn, LAMC2 feedback enhanced the activity of TGF-β signaling by binding to TGF-β receptor II (TGF-βRII).</p><p><strong>Conclusion: </strong>Our findings suggest that LAMC2 promotes GBC progression and metastasis, potentially by regulating the TGF-β signaling pathway. Therefore, LAMC2 could serve as a potential prognostic biomarker and therapeutic target for GBC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145755359","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cross-talk between cancer-associated fibroblasts and dormant cancer cells: current status and promising therapeutic potential. 癌症相关成纤维细胞与休眠癌细胞间的串扰:现状及治疗潜力
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-13 DOI: 10.1186/s12935-025-04120-2
Jiayue Liu, Yue Zhang, Kai Liu, Jialong Shi, Man Yan, Jing Xu, Yandi Liu, Shiwu Zhang
{"title":"Cross-talk between cancer-associated fibroblasts and dormant cancer cells: current status and promising therapeutic potential.","authors":"Jiayue Liu, Yue Zhang, Kai Liu, Jialong Shi, Man Yan, Jing Xu, Yandi Liu, Shiwu Zhang","doi":"10.1186/s12935-025-04120-2","DOIUrl":"https://doi.org/10.1186/s12935-025-04120-2","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CD9-positive tumor-associated macrophages promote renal cell carcinoma progression by activating the orphan nuclear receptor Nor1/Nr4a3. cd9阳性肿瘤相关巨噬细胞通过激活孤儿核受体Nor1/Nr4a3促进肾细胞癌进展。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-12 DOI: 10.1186/s12935-025-04060-x
Pengyu Pan, Shiyuan Li, Wanru Yin, Peng Wang, Tongtong Ma, Ziqian Bi, Junhao Song, Xinyuan Song, Xin Huang, Junfeng Hao, Guangqi Chen, Qiuyu Zhao

Tumor growth, metastasis, and treatment response in renal cell carcinoma (RCC) are believed to be regulated by the tumor microenvironment (TME). However, the mechanisms underlying the genomic, transcriptomic, and epigenetic alterations in clear cell RCC progression remain incompletely understood. This study analyzed single-cell RNA sequencing data from public databases, including tissue samples from three patients with RCC and three matched pairs of immune cells from tumor tissue and peripheral blood to screen macrophages with differential expression in the TME. We found that a CD9+ macrophage population was identified in the RCC tumor tissue, suggesting a role for CD9-expressing macrophages in RCC progression. The function of these macrophages was further investigated through in vitro and in vivo experiments involving CD9 knockdown or overexpression. CD9 expression was elevated in RCC tissues compared to adjacent non-cancerous tissues. CD9 knockdown considerably inhibited the TME-induced macrophages (M2), while CD9-overexpressing macrophages markedly promoted RCC cell growth in vitro and in vivo. Moreover, this phenomenon may be associated with the orphan nuclear receptor NOR1//Nr4a3 activation by CD9. These findings indicated that macrophages in the TME exhibited distinct expression patterns during RCC progression and that CD9+ macrophages promoted RCC development, suggesting that CD9 may be a potential therapeutic target for inhibiting RCC progression.

肾细胞癌(RCC)的肿瘤生长、转移和治疗反应被认为是由肿瘤微环境(TME)调节的。然而,透明细胞RCC进展中基因组、转录组学和表观遗传学改变的机制仍不完全清楚。本研究分析了来自公共数据库的单细胞RNA测序数据,包括来自3名RCC患者的组织样本和来自肿瘤组织和外周血的3对匹配的免疫细胞,以筛选TME中差异表达的巨噬细胞。我们发现在RCC肿瘤组织中发现了CD9+巨噬细胞群,这表明表达CD9的巨噬细胞在RCC进展中起作用。通过体外和体内实验进一步研究这些巨噬细胞的功能,包括CD9敲低或过表达。与邻近非癌组织相比,RCC组织中CD9表达升高。CD9敲低显著抑制tme诱导的巨噬细胞(M2),而CD9过表达的巨噬细胞在体外和体内均显著促进RCC细胞的生长。此外,这种现象可能与孤儿核受体NOR1//Nr4a3被CD9激活有关。这些发现表明,TME中的巨噬细胞在RCC进展过程中表现出不同的表达模式,CD9+巨噬细胞促进RCC的发展,提示CD9可能是抑制RCC进展的潜在治疗靶点。
{"title":"CD9-positive tumor-associated macrophages promote renal cell carcinoma progression by activating the orphan nuclear receptor Nor1/Nr4a3.","authors":"Pengyu Pan, Shiyuan Li, Wanru Yin, Peng Wang, Tongtong Ma, Ziqian Bi, Junhao Song, Xinyuan Song, Xin Huang, Junfeng Hao, Guangqi Chen, Qiuyu Zhao","doi":"10.1186/s12935-025-04060-x","DOIUrl":"https://doi.org/10.1186/s12935-025-04060-x","url":null,"abstract":"<p><p>Tumor growth, metastasis, and treatment response in renal cell carcinoma (RCC) are believed to be regulated by the tumor microenvironment (TME). However, the mechanisms underlying the genomic, transcriptomic, and epigenetic alterations in clear cell RCC progression remain incompletely understood. This study analyzed single-cell RNA sequencing data from public databases, including tissue samples from three patients with RCC and three matched pairs of immune cells from tumor tissue and peripheral blood to screen macrophages with differential expression in the TME. We found that a CD9<sup>+</sup> macrophage population was identified in the RCC tumor tissue, suggesting a role for CD9-expressing macrophages in RCC progression. The function of these macrophages was further investigated through in vitro and in vivo experiments involving CD9 knockdown or overexpression. CD9 expression was elevated in RCC tissues compared to adjacent non-cancerous tissues. CD9 knockdown considerably inhibited the TME-induced macrophages (M2), while CD9-overexpressing macrophages markedly promoted RCC cell growth in vitro and in vivo. Moreover, this phenomenon may be associated with the orphan nuclear receptor NOR1//Nr4a3 activation by CD9. These findings indicated that macrophages in the TME exhibited distinct expression patterns during RCC progression and that CD9<sup>+</sup> macrophages promoted RCC development, suggesting that CD9 may be a potential therapeutic target for inhibiting RCC progression.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741291","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting the LINC01272-FUS signal axis inhibits the migration and invasion of testicular germ cell tumors. 靶向LINC01272-FUS信号轴抑制睾丸生殖细胞肿瘤的迁移和侵袭。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-12 DOI: 10.1186/s12935-025-04095-0
Xueheng Zhao, Jian Cao, Lei Xue, Liqing Fan, Fang Zhu, Zailong Qin, Ziyi Chen, Zhizhong Liu, Jinhui Shu, Hao Bo

Objective: Testicular germ cell tumors (TGCTs), the most prevalent malignancy in males aged 15-40 years, exhibit suboptimal prognosis despite therapeutic advances. This study aimed to identify novel diagnostic biomarkers and elucidate their molecular mechanisms in TGCTs pathogenesis.

Methods: Through integrated bioinformatics analysis of public TCGA(The Cancer Genome Atlas)datasets and clinical specimen validation (n = 14), we screened LINC01272 as a candidate biomarker. RNA immunoprecipitation (RIP) assays were employed to verify the interaction between LINC01272 and fused in sarcoma (FUS). Functional experiments including siRNA-mediated gene silencing, transwell migration/invasion assays, and CCK-8 proliferation tests were performed in TGCTs cell lines (Tcam-2 and NCCIT).

Results: LINC01272 is significantly highly expressed in TGCT tissues. High LINC01272 levels indicate shorter overall and recurrence-free survival periods for patients; the predictive value is particularly significant in the subgroup with low tumor mutation burden. Silencing LINC01272 can inhibit the migration, invasion and proliferation of Tcam-2 and NCCIT cells, and down-regulate N-cadherin and Vimentin, while up-regulating LAMA1. RIP assays confirmed the direct binding of LINC01272 to FUS. FUS silencing can replicate the tumor suppressive effect of LINC01272 deletion, while FUS overexpression can reverse the inhibition of migration, invasion and proliferation caused by LINC01272 silencing.

Conclusion: We identified the LINC01272-FUS axis as a critical regulatory pathway in TGCTs progression, providing mechanistic insights for developing liquid biopsy biomarkers and RNA-targeted therapies.

目的:睾丸生殖细胞肿瘤(tgct)是15-40岁男性中最常见的恶性肿瘤,尽管治疗进展,但预后不佳。本研究旨在寻找新的诊断性生物标志物,并阐明其在tgct发病机制中的分子机制。方法:通过对TCGA(The Cancer Genome Atlas)公共数据集的综合生物信息学分析和临床标本验证(n = 14),筛选LINC01272作为候选生物标志物。采用RNA免疫沉淀法(RIP)验证LINC01272与融合肉瘤(FUS)之间的相互作用。在tgct细胞系(Tcam-2和NCCIT)中进行功能实验,包括sirna介导的基因沉默、跨井迁移/侵袭试验和CCK-8增殖试验。结果:LINC01272在TGCT组织中显著高表达。高LINC01272水平表明患者的总生存期和无复发生存期较短;在肿瘤突变负荷低的亚组中,预测值尤其显著。沉默LINC01272可抑制Tcam-2和NCCIT细胞的迁移、侵袭和增殖,下调N-cadherin和Vimentin,上调LAMA1。RIP实验证实LINC01272与FUS直接结合。FUS沉默可以复制LINC01272缺失对肿瘤的抑制作用,而FUS过表达可以逆转LINC01272沉默对迁移、侵袭和增殖的抑制作用。结论:我们确定了LINC01272-FUS轴是tgct进展的关键调控途径,为开发液体活检生物标志物和rna靶向治疗提供了机制见解。
{"title":"Targeting the LINC01272-FUS signal axis inhibits the migration and invasion of testicular germ cell tumors.","authors":"Xueheng Zhao, Jian Cao, Lei Xue, Liqing Fan, Fang Zhu, Zailong Qin, Ziyi Chen, Zhizhong Liu, Jinhui Shu, Hao Bo","doi":"10.1186/s12935-025-04095-0","DOIUrl":"https://doi.org/10.1186/s12935-025-04095-0","url":null,"abstract":"<p><strong>Objective: </strong>Testicular germ cell tumors (TGCTs), the most prevalent malignancy in males aged 15-40 years, exhibit suboptimal prognosis despite therapeutic advances. This study aimed to identify novel diagnostic biomarkers and elucidate their molecular mechanisms in TGCTs pathogenesis.</p><p><strong>Methods: </strong>Through integrated bioinformatics analysis of public TCGA(The Cancer Genome Atlas)datasets and clinical specimen validation (n = 14), we screened LINC01272 as a candidate biomarker. RNA immunoprecipitation (RIP) assays were employed to verify the interaction between LINC01272 and fused in sarcoma (FUS). Functional experiments including siRNA-mediated gene silencing, transwell migration/invasion assays, and CCK-8 proliferation tests were performed in TGCTs cell lines (Tcam-2 and NCCIT).</p><p><strong>Results: </strong>LINC01272 is significantly highly expressed in TGCT tissues. High LINC01272 levels indicate shorter overall and recurrence-free survival periods for patients; the predictive value is particularly significant in the subgroup with low tumor mutation burden. Silencing LINC01272 can inhibit the migration, invasion and proliferation of Tcam-2 and NCCIT cells, and down-regulate N-cadherin and Vimentin, while up-regulating LAMA1. RIP assays confirmed the direct binding of LINC01272 to FUS. FUS silencing can replicate the tumor suppressive effect of LINC01272 deletion, while FUS overexpression can reverse the inhibition of migration, invasion and proliferation caused by LINC01272 silencing.</p><p><strong>Conclusion: </strong>We identified the LINC01272-FUS axis as a critical regulatory pathway in TGCTs progression, providing mechanistic insights for developing liquid biopsy biomarkers and RNA-targeted therapies.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741270","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabolic-transcriptional rewiring by NFE2L2 promotes M2 macrophage polarization and anti-PD-L1 resistance in glioma. NFE2L2代谢-转录重接线促进胶质瘤中M2巨噬细胞极化和抗pd - l1抗性。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-12 DOI: 10.1186/s12935-025-04113-1
Qiusi Tian, Qun Zhang, Rongzhen Deng, Shengnan Dong, Zhijun Bao, Qingjuan Chen

Glioma is a highly malignant brain tumor, and its characteristic immunosuppressive microenvironment is closely associated with poor patient prognosis. In this study, we aimed to investigate the regulatory role of the transcription factor NFE2L2 (nuclear factor erythroid 2-related factor 2) in glioma and its underlying molecular mechanisms. Analysis based on the TCGA database and clinical samples revealed that NFE2L2 is significantly overexpressed in glioma tissues, and its expression level is closely correlated with tumor immune infiltration, particularly showing a positive association with M2-like tumor-associated macrophage (TAM) infiltration. By establishing an in vitro TAM induction model, we observed that NFE2L2 expression was upregulated during M2-like macrophage polarization. Further gene overexpression experiments demonstrated that NFE2L2 significantly enhances the migratory and invasive capabilities of glioma cells. To explore its in vivo function, we constructed an NFE2L2-deficient mouse model. The results showed that NFE2L2 deficiency led to the remodeling of the tumor immune microenvironment, thereby significantly improving the therapeutic response to PD-1 immune checkpoint inhibitors and suppressing glioma progression. Combined transcriptomic and metabolomic analyses identified differential genes and metabolites associated with NFE2L2, revealing its critical role in promoting M2-like macrophage polarization and modulating the tumor microenvironment. In summary, NFE2L2 not only plays a pivotal role in glioma development but may also serve as a potential therapeutic target. Future studies will further explore the specific mechanisms by which NFE2L2 regulates macrophage polarization and its clinical application prospects in immunotherapy.

胶质瘤是一种高度恶性的脑肿瘤,其特有的免疫抑制微环境与患者预后不良密切相关。在这项研究中,我们旨在探讨转录因子NFE2L2(核因子红系2相关因子2)在胶质瘤中的调节作用及其潜在的分子机制。基于TCGA数据库和临床样本的分析显示,NFE2L2在胶质瘤组织中显著过表达,其表达水平与肿瘤免疫浸润密切相关,特别是与m2样肿瘤相关巨噬细胞(TAM)浸润呈正相关。通过建立TAM体外诱导模型,我们观察到在m2样巨噬细胞极化过程中,NFE2L2表达上调。进一步的基因过表达实验表明,NFE2L2显著增强了胶质瘤细胞的迁移和侵袭能力。为了探索其在体内的功能,我们构建了nfe2l2缺陷小鼠模型。结果表明,NFE2L2缺失导致肿瘤免疫微环境重塑,从而显著改善PD-1免疫检查点抑制剂的治疗反应,抑制胶质瘤进展。结合转录组学和代谢组学分析发现了与NFE2L2相关的差异基因和代谢物,揭示了其在促进m2样巨噬细胞极化和调节肿瘤微环境中的关键作用。综上所述,NFE2L2不仅在胶质瘤的发展中起着关键作用,而且可能作为潜在的治疗靶点。未来的研究将进一步探索NFE2L2调控巨噬细胞极化的具体机制及其在免疫治疗中的临床应用前景。
{"title":"Metabolic-transcriptional rewiring by NFE2L2 promotes M2 macrophage polarization and anti-PD-L1 resistance in glioma.","authors":"Qiusi Tian, Qun Zhang, Rongzhen Deng, Shengnan Dong, Zhijun Bao, Qingjuan Chen","doi":"10.1186/s12935-025-04113-1","DOIUrl":"https://doi.org/10.1186/s12935-025-04113-1","url":null,"abstract":"<p><p>Glioma is a highly malignant brain tumor, and its characteristic immunosuppressive microenvironment is closely associated with poor patient prognosis. In this study, we aimed to investigate the regulatory role of the transcription factor NFE2L2 (nuclear factor erythroid 2-related factor 2) in glioma and its underlying molecular mechanisms. Analysis based on the TCGA database and clinical samples revealed that NFE2L2 is significantly overexpressed in glioma tissues, and its expression level is closely correlated with tumor immune infiltration, particularly showing a positive association with M2-like tumor-associated macrophage (TAM) infiltration. By establishing an in vitro TAM induction model, we observed that NFE2L2 expression was upregulated during M2-like macrophage polarization. Further gene overexpression experiments demonstrated that NFE2L2 significantly enhances the migratory and invasive capabilities of glioma cells. To explore its in vivo function, we constructed an NFE2L2-deficient mouse model. The results showed that NFE2L2 deficiency led to the remodeling of the tumor immune microenvironment, thereby significantly improving the therapeutic response to PD-1 immune checkpoint inhibitors and suppressing glioma progression. Combined transcriptomic and metabolomic analyses identified differential genes and metabolites associated with NFE2L2, revealing its critical role in promoting M2-like macrophage polarization and modulating the tumor microenvironment. In summary, NFE2L2 not only plays a pivotal role in glioma development but may also serve as a potential therapeutic target. Future studies will further explore the specific mechanisms by which NFE2L2 regulates macrophage polarization and its clinical application prospects in immunotherapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741325","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Theranostic potential of MXene-based platforms for dual MiRNA targeting in metastatic bladder cancer. 基于mxene的双MiRNA靶向平台在转移性膀胱癌中的治疗潜力
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-11 DOI: 10.1186/s12935-025-04084-3
Negar Nayerain Jazi, Sajad Alavimanesh, Asma Vafadar, Maedeh Choubani, Shayan Khalili Alashti, Farhad Moradi, Amir Savardashtaki

Metastatic bladder cancer (mBC) continues to resist current treatment approaches, largely because aberrant microRNAs (miRNAs) simultaneously fuel epithelial-mesenchymal transition, enable immune system evasion, and promote resistance to therapies. Mounting evidence pinpoints miR-21 as a key oncomiR that amplifies PI3K/AKT signaling, while miR-200c plays a counterbalancing role by maintaining epithelial traits and promoting apoptosis. This makes the combined strategy of suppressing miR-21 while restoring miR-200c an especially compelling, though technically intricate, therapeutic target. Recently, two-dimensional transition-metal carbides and nitrides-known as MXenes-have emerged as promising tools, thanks to their exceptionally high surface area, excellent conductivity, and customizable surface chemistry, all of which make them ideal for shielding miRNAs, delivering them selectively to tumors, and enabling real-time photothermal monitoring. In this review, we weave together insights from epidemiology, molecular oncology, and nanotechnology to chart a translational pathway for applying MXene-based theranostic systems in mBC. We compile mechanistic data regarding miR-21 and miR-200c, detail the MXene physicochemical traits crucial for RNA loading and biosensing, and critically assess polymer-, ligand-, and ion-intercalation methods that improve biocompatibility without compromising electrical performance. Notably, preclinical studies show that MXene scaffolds can co-deliver anti-miR-21 and miR-200c mimics, destroy tumors via near-infrared photothermal therapy, and allow for electrochemical tracking of miRNA activity inside tumors-together achieving a potent, combined blockade of metastatic processes. Persistent challenges, including oxidative degradation, dose-dependent toxicity, and large-scale manufacturing, are addressed alongside promising innovations such as biodegradable surface coatings and machine-learning-assisted material design. Altogether, dual-miRNA theranostics using MXenes hold immense promise as transformative tools for precision treatment of metastatic bladder cancer.

转移性膀胱癌(mBC)持续抵抗目前的治疗方法,主要是因为异常的microrna (mirna)同时促进上皮-间质转化,使免疫系统逃避,并促进对治疗的抵抗。越来越多的证据表明,miR-21是放大PI3K/AKT信号的关键oncomiR,而miR-200c通过维持上皮性状和促进细胞凋亡发挥平衡作用。这使得抑制miR-21同时恢复miR-200c的联合策略成为一个特别引人注目的治疗靶点,尽管技术上很复杂。最近,二维过渡金属碳化物和氮化物(称为mxenes)因其极高的表面积、优异的导电性和可定制的表面化学特性而成为一种很有前途的工具,所有这些都使它们成为屏蔽mirna的理想选择,可以选择性地将它们输送到肿瘤中,并实现实时光热监测。在这篇综述中,我们将流行病学、分子肿瘤学和纳米技术的见解结合起来,绘制了基于mxene的治疗系统在mBC中的应用的转化途径。我们汇编了关于miR-21和miR-200c的机制数据,详细介绍了对RNA装载和生物传感至关重要的MXene物理化学特性,并批判性地评估了在不影响电性能的情况下提高生物相容性的聚合物、配体和离子插入方法。值得注意的是,临床前研究表明,MXene支架可以共同递送抗mir -21和miR-200c模拟物,通过近红外光热疗法破坏肿瘤,并允许电化学跟踪肿瘤内的miRNA活性,共同实现对转移过程的有效联合阻断。持续的挑战,包括氧化降解、剂量依赖性毒性和大规模生产,以及有前途的创新,如可生物降解表面涂层和机器学习辅助材料设计,都得到了解决。总之,使用MXenes的双mirna疗法作为精确治疗转移性膀胱癌的变革性工具具有巨大的前景。
{"title":"Theranostic potential of MXene-based platforms for dual MiRNA targeting in metastatic bladder cancer.","authors":"Negar Nayerain Jazi, Sajad Alavimanesh, Asma Vafadar, Maedeh Choubani, Shayan Khalili Alashti, Farhad Moradi, Amir Savardashtaki","doi":"10.1186/s12935-025-04084-3","DOIUrl":"10.1186/s12935-025-04084-3","url":null,"abstract":"<p><p>Metastatic bladder cancer (mBC) continues to resist current treatment approaches, largely because aberrant microRNAs (miRNAs) simultaneously fuel epithelial-mesenchymal transition, enable immune system evasion, and promote resistance to therapies. Mounting evidence pinpoints miR-21 as a key oncomiR that amplifies PI3K/AKT signaling, while miR-200c plays a counterbalancing role by maintaining epithelial traits and promoting apoptosis. This makes the combined strategy of suppressing miR-21 while restoring miR-200c an especially compelling, though technically intricate, therapeutic target. Recently, two-dimensional transition-metal carbides and nitrides-known as MXenes-have emerged as promising tools, thanks to their exceptionally high surface area, excellent conductivity, and customizable surface chemistry, all of which make them ideal for shielding miRNAs, delivering them selectively to tumors, and enabling real-time photothermal monitoring. In this review, we weave together insights from epidemiology, molecular oncology, and nanotechnology to chart a translational pathway for applying MXene-based theranostic systems in mBC. We compile mechanistic data regarding miR-21 and miR-200c, detail the MXene physicochemical traits crucial for RNA loading and biosensing, and critically assess polymer-, ligand-, and ion-intercalation methods that improve biocompatibility without compromising electrical performance. Notably, preclinical studies show that MXene scaffolds can co-deliver anti-miR-21 and miR-200c mimics, destroy tumors via near-infrared photothermal therapy, and allow for electrochemical tracking of miRNA activity inside tumors-together achieving a potent, combined blockade of metastatic processes. Persistent challenges, including oxidative degradation, dose-dependent toxicity, and large-scale manufacturing, are addressed alongside promising innovations such as biodegradable surface coatings and machine-learning-assisted material design. Altogether, dual-miRNA theranostics using MXenes hold immense promise as transformative tools for precision treatment of metastatic bladder cancer.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":"19"},"PeriodicalIF":6.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12801870/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145740301","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
Methylated CfDNA may distinguish between high- and intermediate-risk uveal melanoma: a pilot study. 甲基化CfDNA可以区分高风险和中度葡萄膜黑色素瘤:一项初步研究。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-11 DOI: 10.1186/s12935-025-04093-2
Mike Wu, Daniël P de Bruyn, Ruben G Boers, Aaron B Beasley, Daan M Hazelaar, Stavros Makrodimitris, Joachim B Boers, Jolanda Vaarwater, Ronald O B de Keizer, Robert M Verdijk, Nicole C Naus, Dion Paridaens, Saskia M Wilting, Elin S Gray, Wilfred F J van IJcken, Joost Gribnau, Annelies de Klein, Erwin Brosens, Emine Kiliç

Background: Uveal melanoma (UM) is a highly aggressive malignancy with a metastatic risk that depends on the molecular subclass. This subclass can be determined through molecular characterization of tumor-derived tissue. With eye-sparing treatments, tumor tissue is rarely available for molecular testing. We hypothesized that minimal invasive biomarkers such as methylated cell-free DNA (cfDNA) or circulating tumor DNA (ctDNA) can be used for prognosis and monitoring of patients.

Methods: Plasma cfDNA was isolated from healthy blood donors (HBDs, N = 19) and UM patients (N = 22). Plasma was collected at baseline (localized disease, N = 13) and during follow-up (metastatic disease, N = 9) from independent patients with high metastatic risk (HR, N = 11) (monosomy 3 and/or BAP1-mutated tumor) or intermediate metastatic risk (IR, N = 11) (disomy 3 and/or SF3B1-mutated tumor). Methylation signatures were determined using genome-wide LpnPI-based methylated DNA sequencing (MeD-seq). Samples with a CpG/reads ratio < 20% (N = 3) were excluded. IchorCNA was used to estimate the tumor fraction. cfDNA samples with detectable tumor fraction (N = 2) were analyzed separately from the other cfDNA samples without detectable tumor fraction (N = 18) to reduce noise in downstream analyses. Differentially methylated regions (DMRs) were identified between the following predefined subgroups: UM (N = 11) vs. HBDs (N = 19), and HR (N = 10) vs. IR (N = 7). To visualize clustering, principal component analysis (PCA) and hierarchical clustering was performed on the DMRs with fold change > 2.0. Gene set enrichment analysis (GSEA, Z-score > 2.0 and p < 0.05) was performed to evaluate biological relevance.

Results: Distinct clustering was observed between UM and HBDs samples, and between HR and IR samples, although outliers were present in the latter comparison. GSEA implicated eight canonical pathways including the S100 Family Signaling Pathway and RAF/MAP kinase cascade, which are linked to tumorigenesis and immune processes.

Conclusion: This pilot study reports on cfDNA methylation signatures that differentiates UM patients from HBDs, and may distinguish between intermediate and high risk UM subgroups, supporting its prognostic potential. However, its role in monitoring disease progression requires further validation. Independent replication studies are warranted to confirm our findings and evaluate the clinical applicability in UM.

背景:葡萄膜黑色素瘤(UM)是一种高度侵袭性的恶性肿瘤,其转移风险取决于分子亚类。这个亚类可以通过肿瘤来源组织的分子特征来确定。在保眼治疗中,肿瘤组织很少能用于分子检测。我们假设微创生物标志物,如甲基化无细胞DNA (cfDNA)或循环肿瘤DNA (ctDNA)可用于患者的预后和监测。方法:分别从健康献血者(HBDs, N = 19)和UM患者(N = 22)中分离血浆cfDNA。在基线(局限性疾病,N = 13)和随访期间(转移性疾病,N = 9)收集具有高转移风险(HR, N = 11)(3号单体和/或bap1突变肿瘤)或中度转移风险(IR, N = 11)(3号二体和/或sf3b1突变肿瘤)的独立患者的血浆。甲基化特征是使用基于全基因组lpnpi的甲基化DNA测序(MeD-seq)来确定的。CpG/reads比值为2.0的样品。基因集富集分析(GSEA, Z-score > 2.0和p)结果:在UM和HBDs样本之间,以及HR和IR样本之间观察到明显的聚类,尽管后者比较中存在异常值。GSEA涉及8个典型通路,包括S100家族信号通路和RAF/MAP激酶级联,它们与肿瘤发生和免疫过程有关。结论:这项初步研究报告了cfDNA甲基化特征,可以区分UM患者和hbd患者,并可能区分中度和高风险UM亚组,支持其预后潜力。然而,它在监测疾病进展中的作用需要进一步验证。有必要进行独立的重复性研究来证实我们的发现并评估其在UM中的临床适用性。
{"title":"Methylated CfDNA may distinguish between high- and intermediate-risk uveal melanoma: a pilot study.","authors":"Mike Wu, Daniël P de Bruyn, Ruben G Boers, Aaron B Beasley, Daan M Hazelaar, Stavros Makrodimitris, Joachim B Boers, Jolanda Vaarwater, Ronald O B de Keizer, Robert M Verdijk, Nicole C Naus, Dion Paridaens, Saskia M Wilting, Elin S Gray, Wilfred F J van IJcken, Joost Gribnau, Annelies de Klein, Erwin Brosens, Emine Kiliç","doi":"10.1186/s12935-025-04093-2","DOIUrl":"10.1186/s12935-025-04093-2","url":null,"abstract":"<p><strong>Background: </strong>Uveal melanoma (UM) is a highly aggressive malignancy with a metastatic risk that depends on the molecular subclass. This subclass can be determined through molecular characterization of tumor-derived tissue. With eye-sparing treatments, tumor tissue is rarely available for molecular testing. We hypothesized that minimal invasive biomarkers such as methylated cell-free DNA (cfDNA) or circulating tumor DNA (ctDNA) can be used for prognosis and monitoring of patients.</p><p><strong>Methods: </strong>Plasma cfDNA was isolated from healthy blood donors (HBDs, N = 19) and UM patients (N = 22). Plasma was collected at baseline (localized disease, N = 13) and during follow-up (metastatic disease, N = 9) from independent patients with high metastatic risk (HR, N = 11) (monosomy 3 and/or BAP1-mutated tumor) or intermediate metastatic risk (IR, N = 11) (disomy 3 and/or SF3B1-mutated tumor). Methylation signatures were determined using genome-wide LpnPI-based methylated DNA sequencing (MeD-seq). Samples with a CpG/reads ratio < 20% (N = 3) were excluded. IchorCNA was used to estimate the tumor fraction. cfDNA samples with detectable tumor fraction (N = 2) were analyzed separately from the other cfDNA samples without detectable tumor fraction (N = 18) to reduce noise in downstream analyses. Differentially methylated regions (DMRs) were identified between the following predefined subgroups: UM (N = 11) vs. HBDs (N = 19), and HR (N = 10) vs. IR (N = 7). To visualize clustering, principal component analysis (PCA) and hierarchical clustering was performed on the DMRs with fold change > 2.0. Gene set enrichment analysis (GSEA, Z-score > 2.0 and p < 0.05) was performed to evaluate biological relevance.</p><p><strong>Results: </strong>Distinct clustering was observed between UM and HBDs samples, and between HR and IR samples, although outliers were present in the latter comparison. GSEA implicated eight canonical pathways including the S100 Family Signaling Pathway and RAF/MAP kinase cascade, which are linked to tumorigenesis and immune processes.</p><p><strong>Conclusion: </strong>This pilot study reports on cfDNA methylation signatures that differentiates UM patients from HBDs, and may distinguish between intermediate and high risk UM subgroups, supporting its prognostic potential. However, its role in monitoring disease progression requires further validation. Independent replication studies are warranted to confirm our findings and evaluate the clinical applicability in UM.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":"18"},"PeriodicalIF":6.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12801463/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741261","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
THOC1 binds to U2AF2 and regulates ovarian cancer progression through the beta-catenin / c-myc / cyclinD1 signaling pathway. THOC1与U2AF2结合,通过β -catenin / c-myc / cyclinD1信号通路调控卵巢癌进展。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-11 DOI: 10.1186/s12935-025-04034-z
Yifan Feng, Jiaqi Liu, Shuping Zhang, Zhe Zhang, Dong Zhang, Yunxia Cao, Min Li
{"title":"THOC1 binds to U2AF2 and regulates ovarian cancer progression through the beta-catenin / c-myc / cyclinD1 signaling pathway.","authors":"Yifan Feng, Jiaqi Liu, Shuping Zhang, Zhe Zhang, Dong Zhang, Yunxia Cao, Min Li","doi":"10.1186/s12935-025-04034-z","DOIUrl":"10.1186/s12935-025-04034-z","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":"17"},"PeriodicalIF":6.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12801630/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721259","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
Cancer Cell International
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