Journal of Experimental & Clinical Cancer Research最新文献

筛选
英文 中文
CC48 a new CB2R agonist/FAAH inhibitor dual drug blocks gastric cancer progression and overcomes paclitaxel resistance. 新型CB2R激动剂/FAAH抑制剂双药CC48阻断胃癌进展,克服紫杉醇耐药。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-16 DOI: 10.1186/s13046-025-03476-7
Annalisa Schirizzi, Natasha Renna, Giampiero De Leonardis, Rosangela Montanaro, Francesco Mastropasqua, Giovanni Graziano, Chiara Riganti, Isabella Pisano, Antonio Laghezza, Carmen Abate, Angela Stefanachi, Nicola Antonio Colabufo, Cristina Caccioppoli, Giusy Bianco, Anna Maria Valentini, Raffaele Armentano, Gianluigi Giannelli, Marialessandra Contino, Rosalba D'Alessandro
{"title":"CC48 a new CB2R agonist/FAAH inhibitor dual drug blocks gastric cancer progression and overcomes paclitaxel resistance.","authors":"Annalisa Schirizzi, Natasha Renna, Giampiero De Leonardis, Rosangela Montanaro, Francesco Mastropasqua, Giovanni Graziano, Chiara Riganti, Isabella Pisano, Antonio Laghezza, Carmen Abate, Angela Stefanachi, Nicola Antonio Colabufo, Cristina Caccioppoli, Giusy Bianco, Anna Maria Valentini, Raffaele Armentano, Gianluigi Giannelli, Marialessandra Contino, Rosalba D'Alessandro","doi":"10.1186/s13046-025-03476-7","DOIUrl":"10.1186/s13046-025-03476-7","url":null,"abstract":"<p><p>Gastric cancer (GC) has poor survival in advanced stages, with limited treatment options. Paclitaxel (PTX) is commonly used, but resistance often arises, highlighting the need for targeted therapies. Cannabinoid receptor type 2 (CB2R) is overexpressed in several cancers and its activation has been associated with reduced tumor growth and metastasis. This study evaluated the antitumor activity of selected CB2R agonists with dual activity (CC48 and Fi9) compared to single-target compounds (ASF151), a reference agonist (compound 1), and an antagonist (AM630). The compounds' cytotoxicity was determined in GC lines, including PTX-resistant cells, with different levels of CB2R expression. Firstly, were ported that the addition of CB2R ligands to PTX significantly reduces the actively proliferating cells (Ki67+) even in chemotherapy-resistant GC cells. Concentrations below the IC50 of all compounds were used to minimise toxicity. Activation of Akt/mTORC1 and MAPK cascades were found to be related to antiproliferative activity, which was found to be independent of CB2R expression in the different cell lines. Surprisingly, both agonist and antagonist compounds inhibited cell growth. The interaction of CC48 and the reference compounds 1 and AM630, with P-glycoprotein (P-gp) could explain their greater effectiveness in overcoming PTX resistance. Furthermore, CC48 was particularly effective among the agonists in inducing the expression of key autophagy proteins and activating the apoptotic pathway via caspase 3/7 (p < 0.05). The combination of CC48 with PTX further amplified this effect in both sensitive and resistant cells (p < 0.01). CC48 significantly reduced GC cells migration and epithelial-mesenchymal transition (EMT) by modulating the vimentin protein (p < 0.05). In an orthotopic mouse model, CC48 inhibits tumor volume (p < 0.01)and also reduces the number of Ki67 + cells (p < 0.05), without cytotoxic effects. Histological analysis revealed widespread necrosis with inflammatory and apoptotic features, including pyknotic nuclei and fibrotic replacement in CC48-treatedtumors. Moreover, CC48 treatment reduced circulating levels of G-CSF, IL-12 (p40), and eotaxin (p < 0.05), suggesting an immunomodulatory role. In conclusion CC48, a novel multi-target ligand (MTDL), activating CB2R and inhibiting Fatty Acid Amide Hydrolase (FAAH), effectively blocks GC progression modulating the immune response and overcoming PTX resistance.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"209"},"PeriodicalIF":11.4,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12265377/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144644032","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
Tumor-intrinsic ENO1 inhibition promotes antitumor immune response and facilitates the efficacy of anti-PD-L1 immunotherapy in bladder cancer. 肿瘤内源性ENO1抑制可促进抗肿瘤免疫反应,促进抗pd - l1免疫治疗在膀胱癌中的疗效。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-15 DOI: 10.1186/s13046-025-03464-x
Chengquan Shen, Jing Liu, Ding Hu, Changxue Liu, Fei Xie, Yonghua Wang
{"title":"Tumor-intrinsic ENO1 inhibition promotes antitumor immune response and facilitates the efficacy of anti-PD-L1 immunotherapy in bladder cancer.","authors":"Chengquan Shen, Jing Liu, Ding Hu, Changxue Liu, Fei Xie, Yonghua Wang","doi":"10.1186/s13046-025-03464-x","DOIUrl":"10.1186/s13046-025-03464-x","url":null,"abstract":"<p><p>Immunotherapy has revolutionized cancer treatment, yet understanding immunotherapy resistance mechanisms remains challenging. Here, a CRISPR cas9 screening in vivo and an RNA-sequencing for clinical immunotherapy resistance BC samples identified enolase 1 (ENO1) as a potent regulator of anti-PD-L1 treatment efficacy. Investigation of clinical BC samples demonstrated a correlation between ENO1 overexpression and immune evasion in BC, evidenced by reduced CD8<sup>+</sup> T cell infiltration and resistance to anti-PD-L1 therapy. Increased CD8<sup>+</sup> T cell infiltration and function were indicative of antitumor immunity, which was elicited by ENO1 knockdown, which also suppressed carcinogenesis. Single-cell RNA sequencing demonstrated that wild-type (WT) and ENO1 knockout (KO) tumors have different immune cell compositions with the latter preferring an immunostimulatory microenvironment. Mechanistically, ENO1 regulated CD8<sup>+</sup> T cell function and tumor-associated macrophage (TAM) polarization via the SPP1-ITGA4/ITGB1 pathway in the TME. Importantly, genetic and pharmacological inhibition of ENO1 sensitizes tumors to anti-tumor immunity and synergizes with anti-PD-L1 therapy. The results highlight tumor-intrinsic ENO1 as a critical regulator of tumor immune evasion in BC. Targeting ENO1 enhance the efficacy of immune checkpoint blockade therapy by promoting antitumor immunity.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"207"},"PeriodicalIF":11.4,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12261641/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144644036","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
CCNY-mediated phosphorylation and TET2-BACH1-driven DNA demethylation activate PRC1 to augment NSCLC progression. ccny介导的磷酸化和tet2 - bach1驱动的DNA去甲基化激活PRC1以增加NSCLC的进展。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-15 DOI: 10.1186/s13046-025-03472-x
Dayu Huang, Xianglin Chu, Chunxiao Wu, Xuan Wang, Mengkun Shi, Xiaofeng Chen, Yubao Lyu, Dapeng Li, Xuyu Gu
{"title":"CCNY-mediated phosphorylation and TET2-BACH1-driven DNA demethylation activate PRC1 to augment NSCLC progression.","authors":"Dayu Huang, Xianglin Chu, Chunxiao Wu, Xuan Wang, Mengkun Shi, Xiaofeng Chen, Yubao Lyu, Dapeng Li, Xuyu Gu","doi":"10.1186/s13046-025-03472-x","DOIUrl":"10.1186/s13046-025-03472-x","url":null,"abstract":"<p><strong>Background: </strong>The protein regulator of cytokinesis 1 (PRC1) is a prognostic marker characterized by low DNA methylation in lung cancer. This study aims to examine the function of PRC1 in non-small cell lung cancer (NSCLC) cells and investigates its regulatory mechanisms.</p><p><strong>Methods: </strong>PRC1 expression in NSCLC cells was assessed using qPCR and western blot analyses. Loss- and gain-of-function assays of PRC1 were performed in NSCLC cells to analyze its effect on cell cycle progression and growth. Genetic knockdown or pharmaceutical inhibition of cyclin Y (CCNY), tet methylcytosine dioxygenase 2 (TET2), and BTB domain and CNC homolog 1 (BACH1) was conducted to analyze their influence on PRC1 phosphorylation or transcription. Subcutaneous xenograft and orthotopic isograft tumor models were generated for in vivo verification. Tissue microarray (TMA) and bioinformatics analyses were employed to evaluate the clinical prognostic value of CCNY, TET2, and PRC1 in NSCLC.</p><p><strong>Results: </strong>PRC1 was highly expressed in NSCLC cells. Silencing either PRC1 or CCNY, which promotes PRC1 phosphorylation, substantially reduced cell growth in vitro, impaired spindle formation, promoted G2/M phase cell cycle arrest, increased multi-nucleated cells, and weakened tumorigenic activity of cells. Moreover, TET2 was found to induce DNA demethylation of PRC1 and activate its transcription by interacting with BACH1. Inhibition of TET2, BACH1, or the PLK1-PRC1 interaction weakened the tumorigenic potential of NSCLC cells in vivo. The TMA analysis revealed increased levels of CCNY, TET2, and phosphorylated PRC1 in tumor tissues. Bioinformatics analyses suggested that these molecules were correlated with unfavorable prognosis in NSCLC patients.</p><p><strong>Conclusion: </strong>This study demonstrates a critical oncogenic role of PRC1 in NSCLC. CCNY, which modulates PRC1 phosphorylation, and the TET2-BACH1 cascade, which modulates demethylation and transcription of PRC1, may serve as promising targets for NSCLC management.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"206"},"PeriodicalIF":11.4,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12261648/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144644033","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The LncRNA STEAP3-AS1 promotes liver metastasis in colorectal cancer by regulating histone lactylation through chromatin remodelling. LncRNA STEAP3-AS1通过染色质重塑调节组蛋白乳酸化,促进结直肠癌肝转移。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-15 DOI: 10.1186/s13046-025-03461-0
Jinjuan Lv, Xiaoqi Yu, Xiaoqian Liu, Qianshi Zhang, Mengyan Zhang, Jianfeng Gao, Zhiwei Sun, Feifan Zhang, Yunfei Zuo, Shuangyi Ren
{"title":"The LncRNA STEAP3-AS1 promotes liver metastasis in colorectal cancer by regulating histone lactylation through chromatin remodelling.","authors":"Jinjuan Lv, Xiaoqi Yu, Xiaoqian Liu, Qianshi Zhang, Mengyan Zhang, Jianfeng Gao, Zhiwei Sun, Feifan Zhang, Yunfei Zuo, Shuangyi Ren","doi":"10.1186/s13046-025-03461-0","DOIUrl":"10.1186/s13046-025-03461-0","url":null,"abstract":"<p><strong>Introduction: </strong>Liver metastasis is a common cause of death in patients with colorectal cancer (CRC); however, its molecular mechanism remains unclear. Here, we aimed to reveal the role of the lncRNA STEAP3-AS1 in regulating chromatin remodelling and histone lactylation and explore the mechanism by which the lncRNA STEAP3-AS1 promotes CRC liver metastasis. This study provide new research ideas and a theoretical basis for the clinical treatment of cancer.</p><p><strong>Methods: </strong>In this study, we used CRC organoid and nude mouse liver metastasis models to analyse the effect of the lncRNA STEAP3-AS1 on CRC liver metastasis. ATAC-seq, RNA-seq, DRIP-seq and Western blotting were used to screen for the lncRNA STEAP3-AS1 downstream prometastatic molecule MMP9 and the chromatin remodelling factor BRG1. The protein interactions between BRG1, p300, and HDAC3 were evaluated by Co-IP. The the binding of BRG1, ERG, P300, and H3K18la to the MMP9 gene promoter was detected using ChIP-qPCR.</p><p><strong>Results: </strong>The lncRNA STEAP3-AS1 interacts with its parental gene, STEAP3, to form an R-loop on the key chromatin remodelling factor BRG1, regulating the expression of BRG1. Further evidence suggests that BRG1 forms a protein complex with the histone lactylation eraser HDAC3 and the writer P300 to regulate the expression of H3K18la. Moreover, the ATAC-seq analysis revealed that the lncRNA STEAP3-AS1 promotes the chromatin accessibility of MMP9, and a motif and database analysis identified the tumour metastasis factor ERG as an MMP9 transcription factor. The lncRNA STEAP3-AS1 mediates regulation of H3K18la activation of MMP9 by the BRG1/ERG/P300 complex.</p><p><strong>Conclusions: </strong>In summary, these findings revealed that the lncRNA STEAP3-AS1 interacts with its parental gene STEAP3 to regulate H3K18la through BRG1, resulting in changes in chromatin accessibility, thereby driving ERG enrichment an the MMP9 promoter to activate MMP9 transcription and promote CRC liver metastasis. Our findings reveal a novel mechanism by which the lncRNA STEAP3-AS1 promotes CRC metastasis from an epigenetic perspective.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"205"},"PeriodicalIF":11.4,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12261760/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144644035","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
Liensinine reshapes the immune microenvironment and enhances immunotherapy by reprogramming metabolism through the AMPK-HIF-1α axis in hepatocellular carcinoma. Liensinine在肝细胞癌中通过AMPK-HIF-1α轴重编程代谢,重塑免疫微环境并增强免疫治疗。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-15 DOI: 10.1186/s13046-025-03477-6
Jiahao Liu, Xuan Zhang, Xiaofei Fan, Peng Liu, Ze Mi, Hongpei Tan, Pengfei Rong
{"title":"Liensinine reshapes the immune microenvironment and enhances immunotherapy by reprogramming metabolism through the AMPK-HIF-1α axis in hepatocellular carcinoma.","authors":"Jiahao Liu, Xuan Zhang, Xiaofei Fan, Peng Liu, Ze Mi, Hongpei Tan, Pengfei Rong","doi":"10.1186/s13046-025-03477-6","DOIUrl":"10.1186/s13046-025-03477-6","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality, with limited treatment options in advanced stages. Liensinine, a natural alkaloid derived from Nelumbo nucifera, has shown promise as an anticancer agent. However, its underlying mechanisms, particularly in modulating tumor metabolism and immune responses, remain poorly understood. This study aimed to investigate the antitumor effects of Liensinine in HCC, focusing on its ability to modulate metabolic pathways, immune responses, and the tumor microenvironment.</p><p><strong>Methods: </strong>HCC cell lines (HUH7 and Hep1-6) were treated with Liensinine in vitro to assess cell viability, migration, proliferation, and apoptosis. Metabolic reprogramming was analyzed through RNA sequencing, Seahorse metabolic assays, and glucose/lactate measurements. The effects on immune cells were studied by treating THP-1 macrophages and peripheral blood mononuclear cells (PBMCs) with conditioned media from Liensinine-treated cells. In vivo, subcutaneous xenograft and orthotopic liver cancer models were used to evaluate the therapeutic efficacy of Liensinine combination with radiotherapy and immunotherapy.</p><p><strong>Results: </strong>Liensinine inhibited HCC cell viability, migration, and proliferation, promoting apoptosis and shifting metabolism from glycolysis to oxidative phosphorylation. This metabolic reprogramming was linked to the activation of the AMPK-HIF-1α axis and increased ROS production. Furthermore, Liensinine induced Endoplasmic reticulum (ER) stress, as evidenced by elevated levels of CHOP and ATF4, which contributed to AMPK activation and suppression of HIF-1α. Liensinine reduced PD-L1 expression, enhanced M1 macrophage polarization, and promoted CD8 + T cell infiltration into tumors. In vivo, Liensinine significantly suppressed tumor growth, reduced vascular density, and reshaped the immune microenvironment by promoting M1 macrophage polarization. Combination therapy with Liensinine, radiotherapy, and immunotherapy resulted in synergistic effects, including enhanced tumor cell apoptosis, increased immune cell infiltration, and improved therapeutic efficacy.</p><p><strong>Conclusion: </strong>Liensinine exerts potent antitumor effects in HCC by reprogramming tumor metabolism, inducing ER stress, enhancing immune responses, and modulating the TME. The combination of Liensinine with immunotherapy and radiotherapy significantly improves therapeutic efficacy, suggesting its potential as a novel treatment strategy for HCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"208"},"PeriodicalIF":11.4,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12261578/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144644034","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
m5C-modified circRREB1 promotes lung cancer progression by inducing mitophagy. m5c修饰的circRREB1通过诱导线粒体自噬促进肺癌进展。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-14 DOI: 10.1186/s13046-025-03460-1
Dunyu Cai, Xingcai Chen, Haotian Xu, Qingyun Zhao, Xiaodong Zhou, Jiaxi Wu, Shengyi Yuan, Yihong Gao, Deqing Li, Ruirui Zhang, Wenyi Peng, Gang Li, Aruo Nan
{"title":"m5C-modified circRREB1 promotes lung cancer progression by inducing mitophagy.","authors":"Dunyu Cai, Xingcai Chen, Haotian Xu, Qingyun Zhao, Xiaodong Zhou, Jiaxi Wu, Shengyi Yuan, Yihong Gao, Deqing Li, Ruirui Zhang, Wenyi Peng, Gang Li, Aruo Nan","doi":"10.1186/s13046-025-03460-1","DOIUrl":"10.1186/s13046-025-03460-1","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is the most common malignant tumour and the leading cause of cancer-related death. circular RNAs (circRNAs) have important biological functions and are closely related to tumour development. The 5-methylcytosine (m5C) modification can regulate the molecular fate of RNA molecules and thus influence disease development.</p><p><strong>Methods: </strong>High-throughput RNA sequencing was used to construct the differential expression profiles of circRNAs. The m5C modification of circRREB1 was explored through methylated RNA immunoprecipitation (MeRIP) and crosslinking-immunoprecipitation (CLIP). RNA stability experiments, fluorescence in situ hybridization (FISH), and nuclear-cytoplasmic fractionation experiments were performed to explore the effects of the m5C modification on circRREB1. A system for the silencing and overexpression of circRREB1 was established, and in vitro and in vivo experiments were conducted to study the biological functions of circRREB1. Tagged RNA affinity purification (TRAP), RNA immunoprecipitation (RIP), and coimmunoprecipitation (Co-IP) experiments were conducted to reveal the molecular mechanisms of circRREB1.</p><p><strong>Results: </strong>In this study, we found that circRREB1 is highly expressed in lung cancer tissues and cells and that patients with high circRREB1 expression have a poor prognosis. We discovered that circRREB1 undergoes the m5C modification mediated by the methyltransferase NSUN2. This modification facilitates its nuclear export via the m5C reader ALYREF. Functional studies demonstrated that circRREB1 promotes lung cancer progression both in vitro and in vivo. Mechanistically, circRREB1 directly binds to HSPA8 and stabilizes it by inhibiting ubiquitin-dependent degradation, thereby inducing mitophagy through the HSPA8/PINK1/Parkin signalling axis and ultimately promoting the development of lung cancer.</p><p><strong>Conclusions: </strong>This study revealed the presence of m5C modifications on circRREB1 and showed that m5C-modified circRREB1 can induce mitophagy, ultimately promoting lung cancer. These findings provide not only a theoretical basis for further exploration of the mechanisms underlying lung cancer development but also potential targets for lung cancer therapy.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"203"},"PeriodicalIF":11.4,"publicationDate":"2025-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12257754/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144627634","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
Acute degradation of nucleolin reveals its novel functions in cell cycle progression and cell division in triple negative breast cancer. 核蛋白的急性降解揭示了其在三阴性乳腺癌细胞周期进程和细胞分裂中的新功能。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-14 DOI: 10.1186/s13046-025-03401-y
Joseph Mills, Anna Tessari, Vollter Anastas, Damu Sunilkumar, Nastaran Samadi Rad, Saranya Lamba, Ilaria Cosentini, Ashley Reers, Zirui Zhu, Wayne O Miles, Vincenzo Coppola, Emanuele Cocucci, Thomas J Magliery, Heather Shive, Alexander E Davies, Lara Rizzotto, Carlo M Croce, Dario Palmieri
{"title":"Acute degradation of nucleolin reveals its novel functions in cell cycle progression and cell division in triple negative breast cancer.","authors":"Joseph Mills, Anna Tessari, Vollter Anastas, Damu Sunilkumar, Nastaran Samadi Rad, Saranya Lamba, Ilaria Cosentini, Ashley Reers, Zirui Zhu, Wayne O Miles, Vincenzo Coppola, Emanuele Cocucci, Thomas J Magliery, Heather Shive, Alexander E Davies, Lara Rizzotto, Carlo M Croce, Dario Palmieri","doi":"10.1186/s13046-025-03401-y","DOIUrl":"10.1186/s13046-025-03401-y","url":null,"abstract":"<p><strong>Introduction: </strong>Nucleoli are large nuclear sub-compartments where vital processes, such as ribosome assembly, take place. Most nucleolar proteins are essential; thus, their abrogation cannot be achieved through conventional approaches. This technical obstacle has limited our understanding of the biological functions of nucleolar proteins in cell homeostasis and cancer pathogenesis.</p><p><strong>Methods: </strong>We applied the Auxin Inducible Degron (AID) proteolytic system, paired with CRISPR/Cas9 knock-in gene-editing, to obtain an unprecedented characterization of the biological activities of Nucleolin (NCL), one of the most abundant nucleolar proteins, in Triple Negative Breast Cancer (TNBC) cells. Then, we combined live-cell imaging, RNA-sequencing, and quantitative proteomics, to characterize the impact of NCL acute abrogation on the behavior of TNBC cells. Finally, we used in silico analyses to validate NCL molecular role in TNBC patients.</p><p><strong>Results: </strong>Acute abrogation of endogenous NCL impacted both the transcriptome and the proteome of TNBC cells, particularly affecting critical players involved in ribosome biogenesis and in cell cycle progression. Unexpectedly, NCL depletion limited cancer cell ability to effectively complete cytokinesis, ultimately leading to the accumulation of bi-nucleated cells. In silico analyses confirmed that the levels of regulators of cell cycle progression and chromosome segregation correlated with NCL abundance in TNBC patients. Finally, NCL degradation enhanced the activity of pharmaceutical inhibitors of cellular mitosis, such as the Anaphase Promoting Complex inhibitor APCin.</p><p><strong>Conclusions: </strong>Our findings indicate a novel role for NCL in supporting the completion of the cell division in TNBC models, and that its abrogation could enhance the therapeutic activity of mitotic-progression inhibitors.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"204"},"PeriodicalIF":11.4,"publicationDate":"2025-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12257848/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144627633","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
In-depth assessment of BRAF, NRAS, KRAS, EGFR, and PIK3CA mutations on cell-free DNA in the blood of melanoma patients receiving immune checkpoint inhibition. 深入评估接受免疫检查点抑制的黑色素瘤患者血液中游离DNA的BRAF、NRAS、KRAS、EGFR和PIK3CA突变
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-12 DOI: 10.1186/s13046-025-03457-w
Isabel Heidrich, Charlotte Rautmann, Cedric Ly, Robin Khatri, Julian Kött, Glenn Geidel, Alessandra Rünger, Antje Andreas, Inga Hansen-Abeck, Finn Abeck, Anne Menz, Stefan Bonn, Stefan W Schneider, Daniel J Smit, Christoffer Gebhardt, Klaus Pantel
{"title":"In-depth assessment of BRAF, NRAS, KRAS, EGFR, and PIK3CA mutations on cell-free DNA in the blood of melanoma patients receiving immune checkpoint inhibition.","authors":"Isabel Heidrich, Charlotte Rautmann, Cedric Ly, Robin Khatri, Julian Kött, Glenn Geidel, Alessandra Rünger, Antje Andreas, Inga Hansen-Abeck, Finn Abeck, Anne Menz, Stefan Bonn, Stefan W Schneider, Daniel J Smit, Christoffer Gebhardt, Klaus Pantel","doi":"10.1186/s13046-025-03457-w","DOIUrl":"10.1186/s13046-025-03457-w","url":null,"abstract":"<p><strong>Introduction: </strong>Circulating tumor DNA (ctDNA) holds promise for guiding immune checkpoint inhibitor (ICI) therapy and stratifying responders from non-responders. While tumor-informed ctDNA detection approaches are sensitive and mutation-inclusive, they require tumor tissue, which limits applicability in real-world settings. Conversely, tumor-agnostic methods often have limited genomic coverage. In this study, we evaluated a tumor-agnostic, broad-panel ctDNA assay in patients with advanced melanoma treated with ICI.</p><p><strong>Methods: </strong>We conducted a prospective analysis of 241 longitudinal samples from 39 patients with unresectable stage III/IV melanoma using a SYSMEX targeted NGS panel covering 1,114 COSMIC mutations. Plasma samples were collected at baseline and during ICI therapy. The assay's sensitivity reached seven mutant molecules, corresponding to a 0.07% mutation allele frequency (MAF). ctDNA profiles were compared with matched tumor tissue and correlated with clinical features and survival.</p><p><strong>Results: </strong>At baseline, ctDNA was detected in 64.5% of patients. Common mutations included BRAF<sup>V600E</sup> (43.8%) and NRAS<sup>G12D</sup> (36.4%), followed by KRAS, EGFR, and PIK3CA variants. Overall tissue-plasma concordance was 51.6%, with more extended biopsy-plasma intervals associated with discordance (p = 0.0105). Notably, 12.2% of cases exhibited partial concordance, characterized by shared mutations and additional plasma-only alterations, underscoring the complementary value of blood-based profiling. Persistent or re-emerging ctDNA positivity post-therapy correlated with shorter progression-free survival (PFS, p = 0.003), while ctDNA-negative patients showed significantly improved outcomes. Patients that remained ctDNA-negative had significantly longer progression-free survival (median not reached) compared to those with persistent ctDNA positivity (median 3 months) or those converting to positive (median 7.5 months; p = 0.0073). Early NRAS and KRAS ctDNA levels strongly predicted poor response (p = 0.0069 and p = 0.028). The prognostic impact extended beyond canonical drivers, as non-hotspot variants also correlated with the outcome. Notably, even low-level ctDNA persistence (5-10 MM/mL) carried adverse prognostic implications (p = 0.0054). Concerning a shorter PFS, ctDNA positivity was also associated with elevated S100 levels (p = 0.047). Organ-specific mutation enrichment (e.g., KRAS<sup>G12D</sup> in brain, EGFR<sup>G719A</sup> in lymph nodes) suggested possible metastatic tropism.</p><p><strong>Conclusion: </strong>Broad tumor-agnostic ctDNA analysis effectively identified clinically relevant mutations and predicted outcomes in ICI-treated melanoma patients. This approach enables tissue-independent and real-time ctDNA monitoring and may inform patient selection and therapeutic strategies in future interventional trials.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"202"},"PeriodicalIF":11.4,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12255093/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144621062","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
PARP9-PARP13-PARP14 axis tunes colorectal cancer response to radiotherapy. PARP9-PARP13-PARP14轴调节结直肠癌对放疗的反应。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-11 DOI: 10.1186/s13046-025-03439-y
Rimvile Prokarenkaite, Karolina Kuodyte, Greta Gudoityte, Elzbieta Budginaite, Daniel Naumovas, Egle Strainiene, Kristijonas Velickevicius, Audrius Dulskas, Ernestas Sileika, Jonas Venius, Virginijus Tunaitis, Augustas Pivoriunas, Vytaute Starkuviene, Vaidotas Stankevicius, Kestutis Suziedelis
{"title":"PARP9-PARP13-PARP14 axis tunes colorectal cancer response to radiotherapy.","authors":"Rimvile Prokarenkaite, Karolina Kuodyte, Greta Gudoityte, Elzbieta Budginaite, Daniel Naumovas, Egle Strainiene, Kristijonas Velickevicius, Audrius Dulskas, Ernestas Sileika, Jonas Venius, Virginijus Tunaitis, Augustas Pivoriunas, Vytaute Starkuviene, Vaidotas Stankevicius, Kestutis Suziedelis","doi":"10.1186/s13046-025-03439-y","DOIUrl":"10.1186/s13046-025-03439-y","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) is the third most prevalent cancer worldwide. Despite substantial advancements in CRC therapy in recent years, ionizing radiation (IR) continues to be the predominant treatment for colon malignances. However, it still lacks the precision required for excellent therapeutic outcomes, ultimately resulting in tumor radioresistance. This study seeks to explore the potential of atypical PARPs including PARP9, PARP12, PARP13 and PARP14 as innovative radiosensitizing targets for CRC.</p><p><strong>Methods: </strong>We utilized CRISPR/Cas9-mediated gene editing to knockout the PARP9, PARP12, PARP13 and PARP14 in HT29 and DLD1 cells. The cells were exposed to either a single dose of 6-10 Gy or to fractionated dose of 5 × 2 Gy X-ray radiation cultivating cells in 2D, laminin-rich ECM 3D and multicellular spheroid models. The transcriptomes of nonirradiated and irradiated cells were analyzed using microarrays. Gene set enrichment analysis was conducted to determine the pathways in which PARP13 is engaged. Cell viability was assessed using a clonogenic assay. Gene expression levels in cells and patient samples were quantified using RT-qPCR.</p><p><strong>Results: </strong>The expression of PARP9, PARP12, PARP13 and PARP14 was particularly elevated in irradiated colorectal cancer HT29 cells in a microenvironment-dependent manner. PARP13 deficiency significantly enhanced the sensitivity of HT29 cells to both single-dose and multifractionated irradiation regimens, resulting in reduced colony formation and spheroidal integrity. Microarray analysis indicated that PARP13 may modulate the expression genes associated with immune response signaling pathways, including members of PARP family. Furthermore, PARP13 loss in HT29 cells markedly impaired the expression of immune response related genes following multifractionated ionizing irradiation. Finally, chemoradiotherapy significantly elevated the expression of PARP9, PARP12, PARP13 and PARP14 in rectal tumors, while having no effect on adjacent normal colon tissues. Elevated pre-treatment PARP9 expression levels and a blunted post-treatment increase in PARP9 and PARP14 expression predicted poor overall survival in rectal cancer patients, while PARP13 emerged as the most significant discriminator between tumor and healthy tissue.</p><p><strong>Conclusions: </strong>Collectively, the PARP9/13/14 axis is implicated in the response of CRC to radiation treatment in both preclinical and clinical settings, demonstrating the atypical members of the PARP family as attractive targets for neoadjuvant radiotherapy.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"199"},"PeriodicalIF":11.4,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12247367/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144621064","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
WDR3 undergoes phase separation to mediate the therapeutic mechanism of Nilotinib against osteosarcoma. WDR3通过相分离介导尼洛替尼对骨肉瘤的治疗机制。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-11 DOI: 10.1186/s13046-025-03456-x
Minglei Li, Nan Li, Yuying Fan, Zhan Zhang, Long Zhou, Yifan Yu, Man Ni, Mingzi Tan, WanJie Huang, Tong Zhu
{"title":"WDR3 undergoes phase separation to mediate the therapeutic mechanism of Nilotinib against osteosarcoma.","authors":"Minglei Li, Nan Li, Yuying Fan, Zhan Zhang, Long Zhou, Yifan Yu, Man Ni, Mingzi Tan, WanJie Huang, Tong Zhu","doi":"10.1186/s13046-025-03456-x","DOIUrl":"10.1186/s13046-025-03456-x","url":null,"abstract":"<p><strong>Background: </strong>Osteosarcoma is highly invasive with a poor prognosis. The phenomenon of liquid-liquid phase separation (LLPS) can promote the formation of biomolecules and participate in the tumor regulation mechanism. Therefore, mining prognostic markers related to LLPS could allow patients to benefit from targeted therapies.</p><p><strong>Method: </strong>Microarray analysis was performed to identify LLPS-related biomarkers, followed by the validation of binding interactions between genes and drugs via molecular docking analysis. Functions of key genes were investigated in U2-OS cells and xenograft mice. LLPS of WDR3 were observed by the droplet formation assay and fluorescence recovery after photobleaching. The intrinsically disordered region (IDR) of WDR3 was mutated to disrupt LLPS, which was then rescued by the fusion of hnRNAP1 IDR. Therapeutic mechanism of Nilotinib mediated by LLPS was explored in vitro and in vivo.</p><p><strong>Results: </strong>Five LLPS-related biomarkers were screened by bioinformatics analyses to predict the osteosarcoma prognosis. These prognostic genes were significantly associated with the immune cell infiltration, tumor immune escape and drug sensitivity. Among them, WDR3 was a prognostic risk factor for osteosarcoma and stably bound to Nilotinib in the molecular docking model. In transfected U2-OS cells and xenograft mice, the downregulation of WDR3 significantly inhibited the malignant progression of osteosarcoma. More importantly, WDR3 could form droplets in U2-OS cells and restore the fluorescence intensity of WDR3 condensates with liquid-like behavior after photobleaching. The mutation in IDR impaired the phase separation ability of WDR3, whereas the fusion with hnRNAP1 IDR rescued the phase separation abnormality caused by WDR3 mutation. Moreover, the treatment with Nilotinib improved the progression of osteosarcoma in vivo and in vitro, while inhibiting the production of WDR3 phase-separated condensates.</p><p><strong>Conclusion: </strong>WDR3 phase separation involves in the therapeutic mechanism of Nilotinib against osteosarcoma, and thus may serve as a potent biomarker to ameliorate adverse events following osteosarcoma treatment.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"201"},"PeriodicalIF":11.4,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12247437/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144621065","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信