首页 > 最新文献

Journal of Experimental & Clinical Cancer Research最新文献

英文 中文
Correction: PSMD9 promotes the malignant progression of hepatocellular carcinoma by interacting with c-Cbl to activate EGFR signaling and recycling. 更正:PSMD9 通过与 c-Cbl 相互作用,激活表皮生长因子受体信号转导和再循环,从而促进肝细胞癌的恶性发展。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-18 DOI: 10.1186/s13046-024-03209-2
Yuting Su, Lili Meng, Chao Ge, Yuqi Liu, Chi Zhang, Yue Yang, Wei Tian, Hua Tian
{"title":"Correction: PSMD9 promotes the malignant progression of hepatocellular carcinoma by interacting with c-Cbl to activate EGFR signaling and recycling.","authors":"Yuting Su, Lili Meng, Chao Ge, Yuqi Liu, Chi Zhang, Yue Yang, Wei Tian, Hua Tian","doi":"10.1186/s13046-024-03209-2","DOIUrl":"https://doi.org/10.1186/s13046-024-03209-2","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"287"},"PeriodicalIF":11.4,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487807/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142485898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting cancer-associated fibroblasts/tumor cells cross-talk inhibits intrahepatic cholangiocarcinoma progression via cell-cycle arrest. 针对癌症相关成纤维细胞/肿瘤细胞交叉对话,通过细胞周期停滞抑制肝内胆管癌的进展。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-17 DOI: 10.1186/s13046-024-03210-9
Serena Mancarella, Isabella Gigante, Elena Pizzuto, Grazia Serino, Alberta Terzi, Francesco Dituri, Eugenio Maiorano, Leonardo Vincenti, Mario De Bellis, Francesco Ardito, Diego F Calvisi, Gianluigi Giannelli

Background: Cancer-associated fibroblasts (CAFs), mainly responsible for the desmoplastic reaction hallmark of intrahepatic Cholangiocarcinoma (iCCA), likely have a role in tumor aggressiveness and resistance to therapy, although the molecular mechanisms involved are unknown. Aim of the study is to investigate how targeting hCAF/iCCA cross-talk with a Notch1 inhibitor, namely Crenigacestat, may affect cancer progression.

Methods: We used different in vitro models in 2D and established new 3D hetero-spheroids with iCCA cells and human (h)CAFs. The results were confirmed in a xenograft model, and explanted tumoral tissues underwent transcriptomic and bioinformatic analysis.

Results: hCAFs/iCCA cross-talk sustains increased migration of both KKU-M213 and KKU-M156 cells, while Crenigacestat significantly inhibits only the cross-talk stimulated migration. Hetero-spheroids grew larger than homo-spheroids, formed by only iCCA cells. Crenigacestat significantly reduced the invasion and growth of hetero- but not of homo-spheroids. In xenograft models, hCAFs/KKU-M213 tumors grew significantly larger than KKU-M213 tumors, but were significantly reduced in volume by Crenigacestat treatment, which also significantly decreased the fibrotic reaction. Ingenuity pathway analysis revealed that genes of hCAFs/KKU-M213 but not of KKU-M213 tumors increased tumor lesions, and that Crenigacestat treatment inhibited the modulated canonical pathways. Cell cycle checkpoints were the most notably modulated pathway and Crenigacestat reduced CCNE2 gene expression, consequently inducing cell cycle arrest. In hetero-spheroids, the number of cells increased in the G2/M cell cycle phase, while Crenigacestat significantly decreased cell numbers in the G2/M phase in hetero but not in homo-spheroids.

Conclusions: The hCAFs/iCCA cross-talk is a new target for reducing cancer progression with drugs such as Crenigacestat.

背景:癌症相关成纤维细胞(CAFs)主要负责肝内胆管癌(iCCA)标志性的去瘤细胞反应,可能在肿瘤侵袭性和耐药性中发挥作用,但其中涉及的分子机制尚不清楚。本研究的目的是探讨用 Notch1 抑制剂(即 Crenigacestat)靶向 hCAF/iCCA 交叉对话如何影响癌症进展:我们使用了不同的二维体外模型,并用 iCCA 细胞和人(h)CAFs 建立了新的三维异型干细胞。结果:hCAFs/iCCA 交叉配对可维持 KKU-M213 和 KKU-M156 细胞迁移的增加,而 Crenigacestat 仅能显著抑制交叉配对刺激的迁移。仅由 iCCA 细胞形成的异种球形体比同种球形体长得更大。Crenigacestat 能显著降低异型球体的侵袭和生长,但不能降低同型球体的侵袭和生长。在异种移植模型中,hCAFs/KKU-M213肿瘤的体积明显大于KKU-M213肿瘤,但经Crenigacestat处理后体积明显缩小,纤维化反应也明显减轻。Ingenuity通路分析显示,hCAFs/KKU-M213肿瘤的基因增加了肿瘤病变,而KKU-M213肿瘤的基因没有增加,Crenigacestat治疗抑制了被调控的典型通路。细胞周期检查点是最显著的调节途径,Crenigacestat 可降低 CCNE2 基因的表达,从而诱导细胞周期停滞。在异种球形体中,处于G2/M细胞周期阶段的细胞数量增加,而在异种球形体中,Crenigacestat能显著减少处于G2/M阶段的细胞数量,但在同种球形体中却没有:结论:hCAFs/iCCA交叉对话是使用克瑞格司他等药物减少癌症进展的一个新靶点。
{"title":"Targeting cancer-associated fibroblasts/tumor cells cross-talk inhibits intrahepatic cholangiocarcinoma progression via cell-cycle arrest.","authors":"Serena Mancarella, Isabella Gigante, Elena Pizzuto, Grazia Serino, Alberta Terzi, Francesco Dituri, Eugenio Maiorano, Leonardo Vincenti, Mario De Bellis, Francesco Ardito, Diego F Calvisi, Gianluigi Giannelli","doi":"10.1186/s13046-024-03210-9","DOIUrl":"https://doi.org/10.1186/s13046-024-03210-9","url":null,"abstract":"<p><strong>Background: </strong>Cancer-associated fibroblasts (CAFs), mainly responsible for the desmoplastic reaction hallmark of intrahepatic Cholangiocarcinoma (iCCA), likely have a role in tumor aggressiveness and resistance to therapy, although the molecular mechanisms involved are unknown. Aim of the study is to investigate how targeting hCAF/iCCA cross-talk with a Notch1 inhibitor, namely Crenigacestat, may affect cancer progression.</p><p><strong>Methods: </strong>We used different in vitro models in 2D and established new 3D hetero-spheroids with iCCA cells and human (h)CAFs. The results were confirmed in a xenograft model, and explanted tumoral tissues underwent transcriptomic and bioinformatic analysis.</p><p><strong>Results: </strong>hCAFs/iCCA cross-talk sustains increased migration of both KKU-M213 and KKU-M156 cells, while Crenigacestat significantly inhibits only the cross-talk stimulated migration. Hetero-spheroids grew larger than homo-spheroids, formed by only iCCA cells. Crenigacestat significantly reduced the invasion and growth of hetero- but not of homo-spheroids. In xenograft models, hCAFs/KKU-M213 tumors grew significantly larger than KKU-M213 tumors, but were significantly reduced in volume by Crenigacestat treatment, which also significantly decreased the fibrotic reaction. Ingenuity pathway analysis revealed that genes of hCAFs/KKU-M213 but not of KKU-M213 tumors increased tumor lesions, and that Crenigacestat treatment inhibited the modulated canonical pathways. Cell cycle checkpoints were the most notably modulated pathway and Crenigacestat reduced CCNE2 gene expression, consequently inducing cell cycle arrest. In hetero-spheroids, the number of cells increased in the G2/M cell cycle phase, while Crenigacestat significantly decreased cell numbers in the G2/M phase in hetero but not in homo-spheroids.</p><p><strong>Conclusions: </strong>The hCAFs/iCCA cross-talk is a new target for reducing cancer progression with drugs such as Crenigacestat.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"286"},"PeriodicalIF":11.4,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484308/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142479521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Knockdown of THOC1 reduces the proliferation of hepatocellular carcinoma and increases the sensitivity to cisplatin. 更正:敲除 THOC1 可减少肝细胞癌的增殖并增加对顺铂的敏感性。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-14 DOI: 10.1186/s13046-024-03208-3
Shijiao Cai, Yunpeng Bai, Huan Wang, Zihan Zhao, Xiujuan Ding, Heng Zhang, Xiaoyun Zhang, Yantao Liu, Yan Jia, Yinan Li, Shuang Chen, Honggang Zhou, Huijuan Liu, Cheng Yang, Tao Sun
{"title":"Correction: Knockdown of THOC1 reduces the proliferation of hepatocellular carcinoma and increases the sensitivity to cisplatin.","authors":"Shijiao Cai, Yunpeng Bai, Huan Wang, Zihan Zhao, Xiujuan Ding, Heng Zhang, Xiaoyun Zhang, Yantao Liu, Yan Jia, Yinan Li, Shuang Chen, Honggang Zhou, Huijuan Liu, Cheng Yang, Tao Sun","doi":"10.1186/s13046-024-03208-3","DOIUrl":"https://doi.org/10.1186/s13046-024-03208-3","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"285"},"PeriodicalIF":11.4,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11472587/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142485897","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanisms of neural infiltration-mediated tumor metabolic reprogramming impacting immunotherapy efficacy in non-small cell lung cancer. 神经浸润介导的肿瘤代谢重编程影响非小细胞肺癌免疫疗法疗效的机制。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-10 DOI: 10.1186/s13046-024-03202-9
Yuanyuan Zheng, Lifeng Li, Zhibo Shen, Longhao Wang, Xiaoyu Niu, Yujie Wei, Shilong Sun, Jie Zhao

Background: Current evidence underlines the active role of neural infiltration and axonogenesis within the tumor microenvironment (TME), with implications for tumor progression. Infiltrating nerves stimulate tumor growth and dissemination by secreting neurotransmitters, whereas tumor cells influence nerve growth and differentiation through complex interactions, promoting tumor progression. However, the role of neural infiltration in the progression of non-small cell lung cancer (NSCLC) remains unclear.

Methods: This study employs the techniques of immunohistochemistry, immunofluorescence, RNA sequencing, molecular biology experiments, and a murine orthotopic lung cancer model to deeply analyze the specific mechanisms behind the differential efficacy of NSCLC immunotherapy from the perspectives of neuro-tumor signal transduction, tumor metabolism, and tumor immunity.

Results: This study demonstrates that nerve growth factor (NGF) drives neural infiltration in NSCLC, and 5-hydroxytryptamine (5-HT), which is secreted by nerves, is significantly elevated in tumors with extensive neural infiltration. Transcriptome sequencing revealed that 5-HT enhanced glycolysis in NSCLC cells. Pathway analysis indicated that 5-HT activated the PI3K/Akt/mTOR pathway, promoting tumor metabolic reprogramming. This reprogramming exacerbated immunosuppression in the TME. Neutralizing 5-HT-mediated metabolic reprogramming in tumor immunity enhanced the efficacy of PD-1 monoclonal antibody treatment in mice.

Conclusions: The findings of this study provide a novel perspective on the crosstalk between nerves and lung cancer cells and provide insights into further investigations into the role of nerve infiltration in NSCLC progression.

背景:目前有证据表明,神经浸润和轴突生长在肿瘤微环境(TME)中发挥着积极作用,并对肿瘤进展产生影响。浸润神经通过分泌神经递质刺激肿瘤生长和扩散,而肿瘤细胞则通过复杂的相互作用影响神经生长和分化,从而促进肿瘤进展。然而,神经浸润在非小细胞肺癌(NSCLC)进展中的作用仍不清楚:本研究采用免疫组化、免疫荧光、RNA测序、分子生物学实验和小鼠正位肺癌模型等技术,从神经-肿瘤信号转导、肿瘤代谢和肿瘤免疫等角度深入分析了NSCLC免疫治疗不同疗效背后的具体机制:该研究表明,神经生长因子(NGF)驱动NSCLC的神经浸润,而神经分泌的5-羟色胺(5-HT)在有广泛神经浸润的肿瘤中显著升高。转录组测序显示,5-羟色胺促进了 NSCLC 细胞的糖酵解。通路分析表明,5-羟色胺激活了PI3K/Akt/mTOR通路,促进了肿瘤代谢的重编程。这种重编程加剧了TME中的免疫抑制。中和5-HT介导的肿瘤免疫代谢重编程可增强PD-1单克隆抗体治疗小鼠的疗效:本研究的发现为神经与肺癌细胞之间的串扰提供了一个新的视角,并为进一步研究神经浸润在 NSCLC 进展中的作用提供了启示。
{"title":"Mechanisms of neural infiltration-mediated tumor metabolic reprogramming impacting immunotherapy efficacy in non-small cell lung cancer.","authors":"Yuanyuan Zheng, Lifeng Li, Zhibo Shen, Longhao Wang, Xiaoyu Niu, Yujie Wei, Shilong Sun, Jie Zhao","doi":"10.1186/s13046-024-03202-9","DOIUrl":"10.1186/s13046-024-03202-9","url":null,"abstract":"<p><strong>Background: </strong>Current evidence underlines the active role of neural infiltration and axonogenesis within the tumor microenvironment (TME), with implications for tumor progression. Infiltrating nerves stimulate tumor growth and dissemination by secreting neurotransmitters, whereas tumor cells influence nerve growth and differentiation through complex interactions, promoting tumor progression. However, the role of neural infiltration in the progression of non-small cell lung cancer (NSCLC) remains unclear.</p><p><strong>Methods: </strong>This study employs the techniques of immunohistochemistry, immunofluorescence, RNA sequencing, molecular biology experiments, and a murine orthotopic lung cancer model to deeply analyze the specific mechanisms behind the differential efficacy of NSCLC immunotherapy from the perspectives of neuro-tumor signal transduction, tumor metabolism, and tumor immunity.</p><p><strong>Results: </strong>This study demonstrates that nerve growth factor (NGF) drives neural infiltration in NSCLC, and 5-hydroxytryptamine (5-HT), which is secreted by nerves, is significantly elevated in tumors with extensive neural infiltration. Transcriptome sequencing revealed that 5-HT enhanced glycolysis in NSCLC cells. Pathway analysis indicated that 5-HT activated the PI3K/Akt/mTOR pathway, promoting tumor metabolic reprogramming. This reprogramming exacerbated immunosuppression in the TME. Neutralizing 5-HT-mediated metabolic reprogramming in tumor immunity enhanced the efficacy of PD-1 monoclonal antibody treatment in mice.</p><p><strong>Conclusions: </strong>The findings of this study provide a novel perspective on the crosstalk between nerves and lung cancer cells and provide insights into further investigations into the role of nerve infiltration in NSCLC progression.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"284"},"PeriodicalIF":11.4,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11465581/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhancer looping protein LDB1 modulates MYB expression in T-ALL cell lines in vitro by cooperating with master transcription factors. 增强子循环蛋白 LDB1 通过与主转录因子合作调节体外 T-ALL 细胞系中 MYB 的表达。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1186/s13046-024-03199-1
Yan Li, Zimu Zhang, Juanjuan Yu, Hongli Yin, Xinran Chu, Haibo Cao, Yanfang Tao, Yongping Zhang, Zhiheng Li, Shuiyan Wu, Yizhou Hu, Frank Zhu, Jizhao Gao, Xiaodong Wang, Bi Zhou, Wanyan Jiao, Yumeng Wu, Yang Yang, Yanling Chen, Ran Zhuo, Ying Yang, Fenli Zhang, Lei Shi, Yixin Hu, Jian Pan, Shaoyan Hu

Background: Despite significant progress in the prognosis of pediatric T-cell acute lymphoblastic leukemia (T-ALL) in recent decades, a notable portion of children still confronts challenges such as treatment resistance and recurrence, leading to limited options and a poor prognosis. LIM domain-binding protein 1 (LDB1) has been confirmed to exert a crucial role in various physiological and pathological processes. In our research, we aim to elucidate the underlying function and mechanisms of LDB1 within the background of T-ALL.

Methods: Employing short hairpin RNA (shRNA) techniques, we delineated the functional impact of LDB1 in T-ALL cell lines. Through the application of RNA-Seq, CUT&Tag, and immunoprecipitation assays, we scrutinized master transcription factors cooperating with LDB1 and identified downstream targets under LDB1 regulation.

Results: LDB1 emerges as a critical transcription factor co-activator in cell lines derived from T-ALL. It primarily collaborates with master transcription factors (ERG, ETV6, IRF1) to cooperatively regulate the transcription of downstream target genes. Both in vitro and in vivo experiments affirm the essential fuction of LDB1 in the proliferation and survival of cell lines derived from T-ALL, with MYB identified as a significant downstream target of LDB1.

Conclusions: To sum up, our research establishes the pivotal fuction of LDB1 in the tumorigenesis and progression of T-ALL cell lines. Mechanistic insights reveal that LDB1 cooperates with ERG, ETV6, and IRF1 to modulate the expression of downstream effector genes. Furthermore, LDB1 controls MYB through remote enhancer modulation, providing valuable mechanistic insights into its involvement in the progression of T-ALL.

背景:尽管近几十年来小儿T细胞急性淋巴细胞白血病(T-ALL)的预后取得了重大进展,但仍有相当一部分患儿面临耐药和复发等挑战,导致治疗选择有限且预后不佳。LIM结构域结合蛋白1(LDB1)已被证实在各种生理和病理过程中发挥着至关重要的作用。在我们的研究中,我们的目标是在T-ALL的背景下阐明LDB1的潜在功能和机制:方法:利用短发夹 RNA(shRNA)技术,我们研究了 LDB1 在 T-ALL 细胞系中的功能影响。通过应用 RNA-Seq、CUT&Tag 和免疫沉淀实验,我们仔细研究了与 LDB1 合作的主转录因子,并确定了受 LDB1 调控的下游靶标:结果:在T-ALL细胞系中,LDB1是一个关键的转录因子共激活因子。它主要与主转录因子(ERG、ETV6、IRF1)合作,共同调控下游靶基因的转录。体外和体内实验都证实了 LDB1 在 T-ALL 细胞系的增殖和存活过程中起着至关重要的作用,而 MYB 被确定为 LDB1 的一个重要下游靶标:综上所述,我们的研究证实了 LDB1 在 T-ALL 细胞系的肿瘤发生和发展过程中的关键作用。机理揭示了 LDB1 与 ERG、ETV6 和 IRF1 合作调节下游效应基因的表达。此外,LDB1 还通过远程增强子调控来控制 MYB,为其参与 T-ALL 的进展提供了宝贵的机理启示。
{"title":"Enhancer looping protein LDB1 modulates MYB expression in T-ALL cell lines in vitro by cooperating with master transcription factors.","authors":"Yan Li, Zimu Zhang, Juanjuan Yu, Hongli Yin, Xinran Chu, Haibo Cao, Yanfang Tao, Yongping Zhang, Zhiheng Li, Shuiyan Wu, Yizhou Hu, Frank Zhu, Jizhao Gao, Xiaodong Wang, Bi Zhou, Wanyan Jiao, Yumeng Wu, Yang Yang, Yanling Chen, Ran Zhuo, Ying Yang, Fenli Zhang, Lei Shi, Yixin Hu, Jian Pan, Shaoyan Hu","doi":"10.1186/s13046-024-03199-1","DOIUrl":"10.1186/s13046-024-03199-1","url":null,"abstract":"<p><strong>Background: </strong>Despite significant progress in the prognosis of pediatric T-cell acute lymphoblastic leukemia (T-ALL) in recent decades, a notable portion of children still confronts challenges such as treatment resistance and recurrence, leading to limited options and a poor prognosis. LIM domain-binding protein 1 (LDB1) has been confirmed to exert a crucial role in various physiological and pathological processes. In our research, we aim to elucidate the underlying function and mechanisms of LDB1 within the background of T-ALL.</p><p><strong>Methods: </strong>Employing short hairpin RNA (shRNA) techniques, we delineated the functional impact of LDB1 in T-ALL cell lines. Through the application of RNA-Seq, CUT&Tag, and immunoprecipitation assays, we scrutinized master transcription factors cooperating with LDB1 and identified downstream targets under LDB1 regulation.</p><p><strong>Results: </strong>LDB1 emerges as a critical transcription factor co-activator in cell lines derived from T-ALL. It primarily collaborates with master transcription factors (ERG, ETV6, IRF1) to cooperatively regulate the transcription of downstream target genes. Both in vitro and in vivo experiments affirm the essential fuction of LDB1 in the proliferation and survival of cell lines derived from T-ALL, with MYB identified as a significant downstream target of LDB1.</p><p><strong>Conclusions: </strong>To sum up, our research establishes the pivotal fuction of LDB1 in the tumorigenesis and progression of T-ALL cell lines. Mechanistic insights reveal that LDB1 cooperates with ERG, ETV6, and IRF1 to modulate the expression of downstream effector genes. Furthermore, LDB1 controls MYB through remote enhancer modulation, providing valuable mechanistic insights into its involvement in the progression of T-ALL.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"283"},"PeriodicalIF":11.4,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462673/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exosomal miR-106a-5p from highly metastatic colorectal cancer cells drives liver metastasis by inducing macrophage M2 polarization in the tumor microenvironment. 高度转移性结直肠癌细胞的外泌体 miR-106a-5p 通过诱导肿瘤微环境中的巨噬细胞 M2 极化来驱动肝转移。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1186/s13046-024-03204-7
Yahang Liang, Junyu Li, Yuli Yuan, Houqiong Ju, Hualin Liao, Mingming Li, Yang Liu, Yao Yao, Lingling Yang, Taiyuan Li, Xiong Lei

Background: The tumor microenvironment (TME) is a dynamic system orchestrated by intricate cell-to-cell crosstalk. Specifically, macrophages within the TME play a crucial role in driving tumor progression. Exosomes are key mediators of communication between tumor cells and the TME. However, the mechanisms underlying exosome-driven crosstalk between tumor cells and macrophages during colorectal cancer (CRC) progression remain incompletely elucidated.

Methods: Single-cell RNA sequencing were analyzed using the Seurat package. Exosomes were isolated using ultracentrifugation and characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blot. miRNAs differentially expressed in exosomes were analyzed using the limma package. CD206 expression in CRC tissues, exosomes tracing, and exosomal miR-106a-5p transport were observed through immunofluorescence. Macrophage polarization was assessed via qRT-PCR, ELISA, and flow cytometry. The interactions between miR-106a-5p, hnRNPA1, and SOCS6 were evaluated using miRNA pull-down, RIP, and dual-luciferase reporter assays. Transwell assays and liver metastasis model explored the role of exosomal miR-106a-5p-induced M2 macrophages in promoting CRC liver metastasis.

Result: The proportion of M2 macrophages is increased in CRC with liver metastasis compared to those without. Highly metastatic CRC cells release exosomes enriched with miR-106a-5p, which promote macrophages M2 polarization by suppressing SOCS6 and activating JAK2/STAT3 pathway. These M2 macrophages reciprocally enhance CRC liver metastasis. hnRNPA1 regulate the transport of miR-106a-5p into exosomes. Clinically, elevated miR-106a-5p in plasma exosomes correlated with liver metastasis and poor prognosis.

Conclusion: CRC-derived exosomal miR-106a-5p plays a critical role in promoting liver metastasis and is a potential biomarker for the prevention and treatment of CRC liver metastasis.

背景:肿瘤微环境(TME)是一个动态系统,由细胞间错综复杂的串联作用协调而成。具体来说,肿瘤微环境中的巨噬细胞在推动肿瘤进展方面发挥着至关重要的作用。外泌体是肿瘤细胞与 TME 之间沟通的关键媒介。然而,在结直肠癌(CRC)进展过程中,外泌体驱动的肿瘤细胞与巨噬细胞之间的串联机制仍未完全阐明:方法:使用Seurat软件包分析单细胞RNA测序。采用超速离心法分离外泌体,并通过透射电子显微镜、纳米颗粒追踪分析和免疫印迹进行表征。通过免疫荧光观察了 CD206 在 CRC 组织中的表达、外泌体追踪和外泌体 miR-106a-5p 转运。通过 qRT-PCR、ELISA 和流式细胞术评估了巨噬细胞的极化。使用 miRNA pull-down、RIP 和双荧光素酶报告实验评估了 miR-106a-5p、hnRNPA1 和 SOCS6 之间的相互作用。Transwell试验和肝转移模型探讨了外泌体miR-106a-5p诱导的M2巨噬细胞在促进CRC肝转移中的作用:结果:与无肝转移的CRC相比,有肝转移的CRC中M2巨噬细胞的比例增加。高度转移的 CRC 细胞释放富含 miR-106a-5p 的外泌体,通过抑制 SOCS6 和激活 JAK2/STAT3 通路促进巨噬细胞 M2 极化。hnRNPA1 可调控 miR-106a-5p 向外泌体的转运。在临床上,血浆外泌体中miR-106a-5p的升高与肝转移和预后不良有关:结论:CRC外泌体miR-106a-5p在促进肝转移中起着关键作用,是预防和治疗CRC肝转移的潜在生物标志物。
{"title":"Exosomal miR-106a-5p from highly metastatic colorectal cancer cells drives liver metastasis by inducing macrophage M2 polarization in the tumor microenvironment.","authors":"Yahang Liang, Junyu Li, Yuli Yuan, Houqiong Ju, Hualin Liao, Mingming Li, Yang Liu, Yao Yao, Lingling Yang, Taiyuan Li, Xiong Lei","doi":"10.1186/s13046-024-03204-7","DOIUrl":"10.1186/s13046-024-03204-7","url":null,"abstract":"<p><strong>Background: </strong>The tumor microenvironment (TME) is a dynamic system orchestrated by intricate cell-to-cell crosstalk. Specifically, macrophages within the TME play a crucial role in driving tumor progression. Exosomes are key mediators of communication between tumor cells and the TME. However, the mechanisms underlying exosome-driven crosstalk between tumor cells and macrophages during colorectal cancer (CRC) progression remain incompletely elucidated.</p><p><strong>Methods: </strong>Single-cell RNA sequencing were analyzed using the Seurat package. Exosomes were isolated using ultracentrifugation and characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blot. miRNAs differentially expressed in exosomes were analyzed using the limma package. CD206 expression in CRC tissues, exosomes tracing, and exosomal miR-106a-5p transport were observed through immunofluorescence. Macrophage polarization was assessed via qRT-PCR, ELISA, and flow cytometry. The interactions between miR-106a-5p, hnRNPA1, and SOCS6 were evaluated using miRNA pull-down, RIP, and dual-luciferase reporter assays. Transwell assays and liver metastasis model explored the role of exosomal miR-106a-5p-induced M2 macrophages in promoting CRC liver metastasis.</p><p><strong>Result: </strong>The proportion of M2 macrophages is increased in CRC with liver metastasis compared to those without. Highly metastatic CRC cells release exosomes enriched with miR-106a-5p, which promote macrophages M2 polarization by suppressing SOCS6 and activating JAK2/STAT3 pathway. These M2 macrophages reciprocally enhance CRC liver metastasis. hnRNPA1 regulate the transport of miR-106a-5p into exosomes. Clinically, elevated miR-106a-5p in plasma exosomes correlated with liver metastasis and poor prognosis.</p><p><strong>Conclusion: </strong>CRC-derived exosomal miR-106a-5p plays a critical role in promoting liver metastasis and is a potential biomarker for the prevention and treatment of CRC liver metastasis.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"281"},"PeriodicalIF":11.4,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462797/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hepatocyte-derived Igκ promotes HCC progression by stabilizing electron transfer flavoprotein subunit α to facilitate fatty acid β-oxidation. 肝细胞衍生的Igκ通过稳定电子传递黄蛋白亚基α促进脂肪酸β氧化,从而促进肝癌的进展。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1186/s13046-024-03203-8
Jingjing Guo, Huining Gu, Sha Yin, Jiongming Yang, Qianqian Wang, Weiyan Xu, Yifan Wang, Shenghua Zhang, Xiaofeng Liu, Xunde Xian, Xiaoyan Qiu, Jing Huang

Background: Lipid metabolism dysregulation is a key characteristic of hepatocellular carcinoma (HCC) onset and progression. Elevated expression of immunoglobulin (Ig), especially the Igκ free light chain with a unique Vκ4-1/Jκ3 rearrangement in cancer cells, is linked to increased malignancy and has been implicated in colon cancer tumorigenesis. However, the role of Igκ in HCC carcinogenesis remains unclear. The aim of this study was to elucidate the pivotal roles of hepatocyte-derived Igκ in HCC development.

Methods: The rearrangement sequence and expression level of hepatocyte-derived Igκ in HCC cells were determined via RT-PCR, Sanger sequencing, immunohistochemistry, and western blot analysis. The function of Igκ in HCC tumorigenesis was assessed by silencing Igκ using siRNA or gRNA in various HCC cell lines. To assess the role of Igκ in HCC pathogenesis in vivo, a mouse model with hepatocyte-specific Igκ knockout and diethylnitrosamine (DEN) and carbon tetrachloride (CCL4)-induced HCC was utilized. The molecular mechanism by which Igκ affects HCC tumorigenesis was investigated through multiomics analyses, quantitative real-time PCR, immunoprecipitation, mass spectrometry, immunofluorescence, and metabolite detection.

Results: We confirmed that Igκ, especially Vκ4-1/Jκ3-Igκ, is highly expressed in human HCC cells. Igκ depletion inhibited HCC cell proliferation and migration in vitro, and hepatocyte-specific Igκ deficiency ameliorated HCC progression in mice with DEN and CCL4-induced HCC in vivo. Mechanistically, Vκ4-1/Jκ3-Igκ interacts with electron transfer flavoprotein subunit α (ETFA), delaying its protein degradation. Loss of Igκ led to a decrease in the expression of mitochondrial respiratory chain complexes III and IV, resulting in aberrant fatty acid β-oxidation (FAO) and lipid accumulation, which in turn inhibited HCC cell proliferation and migration.

Conclusion: Our findings indicate that the Igκ/ETFA axis deregulates fatty acid β-oxidation, contributing to HCC progression, which suggests that targeting fatty acid metabolism may be an effective HCC treatment strategy. The results of this study suggest that hepatocyte-derived Vκ4-1/Jκ3-Igκ may serve as a promising therapeutic target for HCC.

背景:脂质代谢失调是肝细胞癌(HCC)发病和进展的一个主要特征。免疫球蛋白(Ig),尤其是癌细胞中具有独特 Vκ4-1/Jκ3 重排的 Igκ 游离轻链的高表达与恶性程度的增加有关,并与结肠癌的肿瘤发生有关。然而,Igκ 在 HCC 癌变中的作用仍不清楚。本研究旨在阐明肝细胞来源的 Igκ 在 HCC 发生中的关键作用:方法:通过 RT-PCR、Sanger 测序、免疫组织化学和 Western 印迹分析确定肝细胞源 Igκ 在 HCC 细胞中的重排序列和表达水平。通过使用 siRNA 或 gRNA 在各种 HCC 细胞系中沉默 Igκ 来评估 Igκ 在 HCC 肿瘤发生中的功能。为了评估 Igκ 在 HCC 体内发病机制中的作用,研究人员利用肝细胞特异性 Igκ 基因敲除和二乙基亚硝胺(DEN)及四氯化碳(CCL4)诱导的 HCC 小鼠模型。通过多组学分析、定量实时 PCR、免疫沉淀、质谱、免疫荧光和代谢物检测等方法,研究了 Igκ 影响 HCC 肿瘤发生的分子机制:结果:我们证实了 Igκ,尤其是 Vκ4-1/Jκ3-Igκ 在人 HCC 细胞中的高表达。体外 Igκ 缺失可抑制 HCC 细胞的增殖和迁移,肝细胞特异性 Igκ 缺乏可改善 DEN 和 CCL4 诱导的 HCC 小鼠体内的 HCC 进展。从机理上讲,Vκ4-1/Jκ3-Igκ与电子传递黄蛋白亚基α(ETFA)相互作用,延迟其蛋白质降解。Igκ的缺失导致线粒体呼吸链复合物III和IV的表达减少,导致脂肪酸β氧化(FAO)异常和脂质积累,进而抑制了HCC细胞的增殖和迁移:结论:我们的研究结果表明,Igκ/ETF 轴可抑制脂肪酸的β-氧化,从而导致 HCC 的恶化,这表明靶向脂肪酸代谢可能是一种有效的 HCC 治疗策略。本研究的结果表明,肝细胞衍生的Vκ4-1/Jκ3-Igκ可作为HCC的治疗靶点。
{"title":"Hepatocyte-derived Igκ promotes HCC progression by stabilizing electron transfer flavoprotein subunit α to facilitate fatty acid β-oxidation.","authors":"Jingjing Guo, Huining Gu, Sha Yin, Jiongming Yang, Qianqian Wang, Weiyan Xu, Yifan Wang, Shenghua Zhang, Xiaofeng Liu, Xunde Xian, Xiaoyan Qiu, Jing Huang","doi":"10.1186/s13046-024-03203-8","DOIUrl":"10.1186/s13046-024-03203-8","url":null,"abstract":"<p><strong>Background: </strong>Lipid metabolism dysregulation is a key characteristic of hepatocellular carcinoma (HCC) onset and progression. Elevated expression of immunoglobulin (Ig), especially the Igκ free light chain with a unique Vκ4-1/Jκ3 rearrangement in cancer cells, is linked to increased malignancy and has been implicated in colon cancer tumorigenesis. However, the role of Igκ in HCC carcinogenesis remains unclear. The aim of this study was to elucidate the pivotal roles of hepatocyte-derived Igκ in HCC development.</p><p><strong>Methods: </strong>The rearrangement sequence and expression level of hepatocyte-derived Igκ in HCC cells were determined via RT-PCR, Sanger sequencing, immunohistochemistry, and western blot analysis. The function of Igκ in HCC tumorigenesis was assessed by silencing Igκ using siRNA or gRNA in various HCC cell lines. To assess the role of Igκ in HCC pathogenesis in vivo, a mouse model with hepatocyte-specific Igκ knockout and diethylnitrosamine (DEN) and carbon tetrachloride (CCL4)-induced HCC was utilized. The molecular mechanism by which Igκ affects HCC tumorigenesis was investigated through multiomics analyses, quantitative real-time PCR, immunoprecipitation, mass spectrometry, immunofluorescence, and metabolite detection.</p><p><strong>Results: </strong>We confirmed that Igκ, especially Vκ4-1/Jκ3-Igκ, is highly expressed in human HCC cells. Igκ depletion inhibited HCC cell proliferation and migration in vitro, and hepatocyte-specific Igκ deficiency ameliorated HCC progression in mice with DEN and CCL4-induced HCC in vivo. Mechanistically, Vκ4-1/Jκ3-Igκ interacts with electron transfer flavoprotein subunit α (ETFA), delaying its protein degradation. Loss of Igκ led to a decrease in the expression of mitochondrial respiratory chain complexes III and IV, resulting in aberrant fatty acid β-oxidation (FAO) and lipid accumulation, which in turn inhibited HCC cell proliferation and migration.</p><p><strong>Conclusion: </strong>Our findings indicate that the Igκ/ETFA axis deregulates fatty acid β-oxidation, contributing to HCC progression, which suggests that targeting fatty acid metabolism may be an effective HCC treatment strategy. The results of this study suggest that hepatocyte-derived Vκ4-1/Jκ3-Igκ may serve as a promising therapeutic target for HCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"280"},"PeriodicalIF":11.4,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462706/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Plasma-derived extracellular vesicles miR-335-5p as potential diagnostic biomarkers for fusion-positive rhabdomyosarcoma. 血浆源性细胞外囊泡 miR-335-5p 作为融合阳性横纹肌肉瘤的潜在诊断生物标记物
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1186/s13046-024-03197-3
Virginia Di Paolo, Alessandro Paolini, Angela Galardi, Patrizia Gasparini, Loris De Cecco, Marta Colletti, Silvia Lampis, Salvatore Raieli, Cristiano De Stefanis, Evelina Miele, Ida Russo, Valentina Di Ruscio, Michela Casanova, Rita Alaggio, Andrea Masotti, Giuseppe Maria Milano, Franco Locatelli, Angela Di Giannatale

Background: Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma, with embryonal (ERMS) and alveolar (ARMS) representing the two most common histological subtypes. ARMS shows poor prognosis, being often metastatic at diagnosis. Thus, the discovery of novel biomarkers predictive of tumor aggressiveness represents one of the most important challenges to overcome and may help the development of tailored therapies. In the last years, miRNAs carried in extracellular vesicles (EVs), small vesicles of endocytic origin, have emerged as ideal candidate biomarkers due to their stability in plasma and their tissue specificity.

Methods: EVs miRNAs were isolated from plasma of 21 patients affected by RMS and 13 healthy childrens (HC). We performed a miRNA profile using the Serum/Plasma Focus microRNA PCR panels (Qiagen), and RT-qPCR for validation analysis. Statistically significant (p < 0.05) miRNAs were obtained by ANOVA test.

Results: We identified nine EVs miRNAs (miR-483-5p, miR-132-3p, miR-766-3p, miR-454-3p miR-197-3p, miR-335-3p, miR-17-5p, miR-486-5p and miR-484) highly upregulated in RMS patients compared to HCs. Interestingly, 4 miRNAs (miR-335-5p, miR-17-5p, miR-486-5p and miR-484) were significantly upregulated in ARMS samples compared to ERMS. In the validation analysis performed in a larger group of patients only three miRNAs (miR-483-5p, miR-335-5p and miR-484) were differentially significantly expressed in RMS patients compared to HC. Among these, mir-335-5p was significant also when compared ARMS to ERMS patients. MiR-335-5p was upregulated in RMS tumor tissues respect to normal tissues (p = 0.00202) and upregulated significantly between ARMS and ERMS (p = 0.04). Furthermore, the miRNA expression correlated with the Intergroup Rhabdomyosarcoma Study (IRS) grouping system, (p = 0.0234), and survival (OS, p = 0.044; PFS, p = 0.025). By performing in situ hybridization, we observed that miR-335-5p signal was exclusively in the cytoplasm of cancer cells.

Conclusion: We identified miR-335-5p as significantly upregulated in plasma derived EVs and tumor tissue of patients affected by ARMS. Its expression correlates to stage and survival in patients. Future studies are needed to validate miR-335-5p as prognostic biomarker and to deeply elucidate its biological role.

背景:横纹肌肉瘤(RMS)是最常见的儿科软组织肉瘤,胚胎型(ERMS)和肺泡型(ARMS)是两种最常见的组织学亚型。ARMS预后较差,确诊时往往已发生转移。因此,发现可预测肿瘤侵袭性的新型生物标志物是需要克服的最重要挑战之一,这可能有助于开发有针对性的疗法。过去几年中,细胞外囊泡(EVs)中携带的 miRNA 因其在血浆中的稳定性和组织特异性而成为理想的候选生物标志物:从 21 名 RMS 患者和 13 名健康儿童(HC)的血浆中分离出 EVs miRNA。我们使用血清/血浆Focus microRNA PCR板(Qiagen)进行了miRNA分析,并使用RT-qPCR进行了验证分析。结果表明,这些数据具有统计学意义(p与 HCs 相比,我们在 RMS 患者中发现了 9 种 EVs miRNA(miR-483-5p、miR-132-3p、miR-766-3p、miR-454-3p miR-197-3p、miR-335-3p、miR-17-5p、miR-486-5p 和 miR-484)高度上调。有趣的是,与ERMS相比,4个miRNA(miR-335-5p、miR-17-5p、miR-486-5p和miR-484)在ARMS样本中显著上调。在对更多患者进行的验证分析中,只有三个 miRNA(miR-483-5p、miR-335-5p 和 miR-484)在 RMS 患者中的表达与 HC 患者相比有明显差异。其中,mir-335-5p在ARMS与ERMS患者的比较中也有显著差异。与正常组织相比,MiR-335-5p在RMS肿瘤组织中上调(p = 0.00202),并且在ARMS和ERMS之间明显上调(p = 0.04)。此外,miRNA表达与组间横纹肌肉瘤研究(IRS)分组系统(p = 0.0234)和生存期(OS,p = 0.044;PFS,p = 0.025)相关。通过原位杂交,我们观察到 miR-335-5p 信号只存在于癌细胞的细胞质中:结论:我们发现 miR-335-5p 在 ARMS 患者的血浆衍生 EVs 和肿瘤组织中明显上调。其表达与患者的分期和生存期相关。未来的研究需要验证 miR-335-5p 作为预后生物标志物的有效性,并深入阐明其生物学作用。
{"title":"Plasma-derived extracellular vesicles miR-335-5p as potential diagnostic biomarkers for fusion-positive rhabdomyosarcoma.","authors":"Virginia Di Paolo, Alessandro Paolini, Angela Galardi, Patrizia Gasparini, Loris De Cecco, Marta Colletti, Silvia Lampis, Salvatore Raieli, Cristiano De Stefanis, Evelina Miele, Ida Russo, Valentina Di Ruscio, Michela Casanova, Rita Alaggio, Andrea Masotti, Giuseppe Maria Milano, Franco Locatelli, Angela Di Giannatale","doi":"10.1186/s13046-024-03197-3","DOIUrl":"10.1186/s13046-024-03197-3","url":null,"abstract":"<p><strong>Background: </strong>Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma, with embryonal (ERMS) and alveolar (ARMS) representing the two most common histological subtypes. ARMS shows poor prognosis, being often metastatic at diagnosis. Thus, the discovery of novel biomarkers predictive of tumor aggressiveness represents one of the most important challenges to overcome and may help the development of tailored therapies. In the last years, miRNAs carried in extracellular vesicles (EVs), small vesicles of endocytic origin, have emerged as ideal candidate biomarkers due to their stability in plasma and their tissue specificity.</p><p><strong>Methods: </strong>EVs miRNAs were isolated from plasma of 21 patients affected by RMS and 13 healthy childrens (HC). We performed a miRNA profile using the Serum/Plasma Focus microRNA PCR panels (Qiagen), and RT-qPCR for validation analysis. Statistically significant (p < 0.05) miRNAs were obtained by ANOVA test.</p><p><strong>Results: </strong>We identified nine EVs miRNAs (miR-483-5p, miR-132-3p, miR-766-3p, miR-454-3p miR-197-3p, miR-335-3p, miR-17-5p, miR-486-5p and miR-484) highly upregulated in RMS patients compared to HCs. Interestingly, 4 miRNAs (miR-335-5p, miR-17-5p, miR-486-5p and miR-484) were significantly upregulated in ARMS samples compared to ERMS. In the validation analysis performed in a larger group of patients only three miRNAs (miR-483-5p, miR-335-5p and miR-484) were differentially significantly expressed in RMS patients compared to HC. Among these, mir-335-5p was significant also when compared ARMS to ERMS patients. MiR-335-5p was upregulated in RMS tumor tissues respect to normal tissues (p = 0.00202) and upregulated significantly between ARMS and ERMS (p = 0.04). Furthermore, the miRNA expression correlated with the Intergroup Rhabdomyosarcoma Study (IRS) grouping system, (p = 0.0234), and survival (OS, p = 0.044; PFS, p = 0.025). By performing in situ hybridization, we observed that miR-335-5p signal was exclusively in the cytoplasm of cancer cells.</p><p><strong>Conclusion: </strong>We identified miR-335-5p as significantly upregulated in plasma derived EVs and tumor tissue of patients affected by ARMS. Its expression correlates to stage and survival in patients. Future studies are needed to validate miR-335-5p as prognostic biomarker and to deeply elucidate its biological role.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"282"},"PeriodicalIF":11.4,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11463097/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: STUB1-mediated K63-linked ubiquitination of UHRF1 promotes the progression of cholangiocarcinoma by maintaining DNA hypermethylation of PLA2G2A. 更正:STUB1 介导的 K63 链接泛素化 UHRF1 通过维持 PLA2G2A 的 DNA 高甲基化促进胆管癌的进展。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-08 DOI: 10.1186/s13046-024-03198-2
Junsheng Chen, Da Wang, Guanhua Wu, Fei Xiong, Wenzheng Liu, Qi Wang, Yiyang Kuai, Wenhua Huang, Yongqiang Qi, Bing Wang, Yongjun Chen
{"title":"Correction: STUB1-mediated K63-linked ubiquitination of UHRF1 promotes the progression of cholangiocarcinoma by maintaining DNA hypermethylation of PLA2G2A.","authors":"Junsheng Chen, Da Wang, Guanhua Wu, Fei Xiong, Wenzheng Liu, Qi Wang, Yiyang Kuai, Wenhua Huang, Yongqiang Qi, Bing Wang, Yongjun Chen","doi":"10.1186/s13046-024-03198-2","DOIUrl":"https://doi.org/10.1186/s13046-024-03198-2","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"279"},"PeriodicalIF":11.4,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11460208/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NAT10/ac4C/JunB facilitates TNBC malignant progression and immunosuppression by driving glycolysis addiction. NAT10/ac4C/JunB通过驱动糖酵解成瘾促进TNBC恶性进展和免疫抑制。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-04 DOI: 10.1186/s13046-024-03200-x
Guozheng Li, Xin Ma, Shiyao Sui, Yihai Chen, Hui Li, Lei Liu, Xin Zhang, Lei Zhang, Yi Hao, Zihan Yang, Shuai Yang, Xu He, Qin Wang, Weiyang Tao, Shouping Xu

Background: N4-Acetylcytidine (ac4C), a highly conserved post-transcriptional mechanism, plays a pivotal role in RNA modification and tumor progression. However, the molecular mechanism by which ac4C modification mediates tumor immunosuppression remains elusive in triple-negative breast cancer (TNBC).

Methods: NAT10 expression was analyzed in TNBC samples in the level of mRNA and protein, and compared with the corresponding normal tissues. ac4C modification levels also measured in the TNBC samples. The effects of NAT10 on immune microenvironment and tumor metabolism were investigated. NAT10-mediated ac4C and its downstream regulatory mechanisms were determined in vitro and in vivo. The combination therapy of targeting NAT10 in TNBC was further explored.

Results: The results revealed that the loss of NAT10 inhibited TNBC development and promoted T cell activation. Mechanistically, NAT10 upregulated JunB expression by increasing ac4C modification levels on its mRNA. Moreover, JunB further up-regulated LDHA expression and facilitated glycolysis. By deeply digging, remodelin, a NAT10 inhibitor, elevated the surface expression of CTLA-4 on T cells. The combination of remodelin and CTLA-4 mAb can further activate T cells and inhibite tumor progression.

Conclusion: Taken together, our study demonstrated that the NAT10-ac4C-JunB-LDHA pathway increases glycolysis levels and creates an immunosuppressive tumor microenvironment (TME). Consequently, targeting this pathway may assist in the identification of novel therapeutic strategies to improve the efficacy of cancer immunotherapy.

背景:N4-乙酰胞苷(ac4C)是一种高度保守的转录后机制,在RNA修饰和肿瘤进展中发挥着关键作用。然而,在三阴性乳腺癌(TNBC)中,ac4C修饰介导肿瘤免疫抑制的分子机制仍然难以捉摸:方法:分析 TNBC 样本中 NAT10 的 mRNA 和蛋白表达水平,并与相应的正常组织进行比较。研究还探讨了NAT10对免疫微环境和肿瘤代谢的影响。在体外和体内确定了 NAT10 介导的 ac4C 及其下游调控机制。进一步探讨了靶向 NAT10 的联合疗法在 TNBC 中的应用:结果表明:NAT10的缺失抑制了TNBC的发展并促进了T细胞的活化。从机理上讲,NAT10通过增加JunB mRNA上的ac4C修饰水平来上调JunB的表达。此外,JunB还能进一步上调LDHA的表达,促进糖酵解。通过深度挖掘,NAT10抑制剂remodelin可提高T细胞表面CTLA-4的表达。remodelin与CTLA-4 mAb的结合可进一步激活T细胞,抑制肿瘤进展:综上所述,我们的研究表明,NAT10-ac4C-JunB-LDHA通路会增加糖酵解水平,并形成免疫抑制性肿瘤微环境(TME)。因此,靶向这一途径可能有助于确定新型治疗策略,提高癌症免疫疗法的疗效。
{"title":"NAT10/ac4C/JunB facilitates TNBC malignant progression and immunosuppression by driving glycolysis addiction.","authors":"Guozheng Li, Xin Ma, Shiyao Sui, Yihai Chen, Hui Li, Lei Liu, Xin Zhang, Lei Zhang, Yi Hao, Zihan Yang, Shuai Yang, Xu He, Qin Wang, Weiyang Tao, Shouping Xu","doi":"10.1186/s13046-024-03200-x","DOIUrl":"10.1186/s13046-024-03200-x","url":null,"abstract":"<p><strong>Background: </strong>N4-Acetylcytidine (ac4C), a highly conserved post-transcriptional mechanism, plays a pivotal role in RNA modification and tumor progression. However, the molecular mechanism by which ac4C modification mediates tumor immunosuppression remains elusive in triple-negative breast cancer (TNBC).</p><p><strong>Methods: </strong>NAT10 expression was analyzed in TNBC samples in the level of mRNA and protein, and compared with the corresponding normal tissues. ac4C modification levels also measured in the TNBC samples. The effects of NAT10 on immune microenvironment and tumor metabolism were investigated. NAT10-mediated ac4C and its downstream regulatory mechanisms were determined in vitro and in vivo. The combination therapy of targeting NAT10 in TNBC was further explored.</p><p><strong>Results: </strong>The results revealed that the loss of NAT10 inhibited TNBC development and promoted T cell activation. Mechanistically, NAT10 upregulated JunB expression by increasing ac4C modification levels on its mRNA. Moreover, JunB further up-regulated LDHA expression and facilitated glycolysis. By deeply digging, remodelin, a NAT10 inhibitor, elevated the surface expression of CTLA-4 on T cells. The combination of remodelin and CTLA-4 mAb can further activate T cells and inhibite tumor progression.</p><p><strong>Conclusion: </strong>Taken together, our study demonstrated that the NAT10-ac4C-JunB-LDHA pathway increases glycolysis levels and creates an immunosuppressive tumor microenvironment (TME). Consequently, targeting this pathway may assist in the identification of novel therapeutic strategies to improve the efficacy of cancer immunotherapy.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"278"},"PeriodicalIF":11.4,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11451012/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373422","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Experimental & Clinical Cancer Research
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:481959085
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