Journal of Experimental & Clinical Cancer Research最新文献

筛选
英文 中文
Non-canonical activation of MAPK signaling by the lncRNA ASH1L-AS1-encoded microprotein APPLE through inhibition of PP1/PP2A-mediated ERK1/2 dephosphorylation in hepatocellular carcinoma. lncRNA ash1l - as1编码的微蛋白APPLE通过抑制PP1/ pp2a介导的ERK1/2去磷酸化而非典型激活MAPK信号
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-11 DOI: 10.1186/s13046-025-03465-w
Lei Zhao, Ke Si, Shenjian Luo, Lantian Zhang, Shuai Mao, Wenliang Zhang
{"title":"Non-canonical activation of MAPK signaling by the lncRNA ASH1L-AS1-encoded microprotein APPLE through inhibition of PP1/PP2A-mediated ERK1/2 dephosphorylation in hepatocellular carcinoma.","authors":"Lei Zhao, Ke Si, Shenjian Luo, Lantian Zhang, Shuai Mao, Wenliang Zhang","doi":"10.1186/s13046-025-03465-w","DOIUrl":"10.1186/s13046-025-03465-w","url":null,"abstract":"<p><strong>Background: </strong>MAPK/ERK1/2 signaling is often activated in hepatocellular carcinoma (HCC), yet classical RAS-RAF-MEK mutations are rare, indicating the involvement of non-canonical regulatory mechanisms. Long non-coding RNAs (lncRNAs) can encode microproteins that play key roles in cancer. LncRNA ASH1L-AS1 has coding potential, but its role in HCC remains unclear. Clarifying its role in MAPK signaling may uncover novel therapeutic targets for HCC.</p><p><strong>Methods: </strong>Translatable lncRNAs associated with HCC were identified by integrating data from the TCGA-LIHC cohort and the TransLnc database. The functional role of ASH1L-AS1 and its encoded microprotein APPLE was explored through in vitro and in vivo assays, such as CCK-8, EdU incorporation, wound healing, Transwell migration and invasion, and xenograft tumor models. Mechanistic investigations were conducted to elucidate molecular mechanisms and identify potential therapeutic strategies, including co-immunoprecipitation, mass spectrometry, ChIP-qPCR, luciferase reporter assays, truncation mutation analysis, immunofluorescence, Western blot, RNA sequencing, drug sensitivity analysis etc. RESULTS: A total of 696 translatable lncRNAs associated with HCC were identified, with their encoded products exhibiting specific subcellular localization. Among them, ASH1L-AS1 stood out due to strong translational evidence and its significant association with disease progression, poor prognosis, immunosuppressive tumor microenvironment, and estrogen signaling. We confirmed that ASH1L-AS1 encodes a microprotein, APPLE, which is stably expressed in HCC cells and consistently upregulated in tumor tissues regardless of RAS mutation status. Functionally, APPLE promotes ERK1/2 phosphorylation, activates MAPK signaling, and enhances HCC cell proliferation, migration, invasion, and tumor growth-effects reversed by APPLE knockdown or ERK1/2 inhibition. Mechanistically, APPLE binds to ERK1/2 and phosphatases PP1/PP2A, preventing ERK1/2 dephosphorylation and sustaining MAPK pathway activation. Additionally, the transcription factor E2F1 directly binds to the ASH1L-AS1 promoter (- 300 to - 290 bp), upregulating APPLE expression and further amplifying ERK1/2 signaling. Drug sensitivity analysis identified 220 treatment combinations potentially effective against HCC subtypes driven by hyperactivation of the E2F1-ASH1L-AS1/APPLE-ERK1/2 axis.</p><p><strong>Conclusions: </strong>This study characterized APPLE as a novel oncogenic microprotein encoded by lncRNA ASH1L-AS1, uncovering a non-canonical mechanism of MAPK activation in HCC. The identified E2F1-ASH1L-AS1/APPLE-ERK1/2 signaling axis provides new insights into HCC pathogenesis and represents a promising target for precision therapy, though further validation in clinical cohorts and preclinical studies is needed.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"200"},"PeriodicalIF":11.4,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12247205/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144621063","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
PHGDH drives 5-FU chemoresistance in colorectal cancer through the Hedgehog signaling. PHGDH通过Hedgehog信号传导驱动结直肠癌5-FU化疗耐药。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-10 DOI: 10.1186/s13046-025-03447-y
Caterina Mancini, Giulia Lori, Gianluca Mattei, Marta Iozzo, Dayana Desideri, Fabio Cianchi, Laura Fortuna, Federico Passagnoli, Daniela Massi, Filippo Ugolini, Luca Messerini, Salvatore Piscuoglio, Antonio Pezone, Francesca Magherini, Alessio Biagioni, Tiziano Lottini, Demetra Zambardino, Giuseppina Ivana Truglio, Elena Petricci, Alberto Magi, Annarosa Arcangeli, Luisa Maresca, Barbara Stecca, Erica Pranzini, Maria Letizia Taddei
{"title":"PHGDH drives 5-FU chemoresistance in colorectal cancer through the Hedgehog signaling.","authors":"Caterina Mancini, Giulia Lori, Gianluca Mattei, Marta Iozzo, Dayana Desideri, Fabio Cianchi, Laura Fortuna, Federico Passagnoli, Daniela Massi, Filippo Ugolini, Luca Messerini, Salvatore Piscuoglio, Antonio Pezone, Francesca Magherini, Alessio Biagioni, Tiziano Lottini, Demetra Zambardino, Giuseppina Ivana Truglio, Elena Petricci, Alberto Magi, Annarosa Arcangeli, Luisa Maresca, Barbara Stecca, Erica Pranzini, Maria Letizia Taddei","doi":"10.1186/s13046-025-03447-y","DOIUrl":"10.1186/s13046-025-03447-y","url":null,"abstract":"<p><strong>Background: </strong>Phosphoglycerate dehydrogenase (PHGDH) is the rate-limiting enzyme in the de novo Serine synthesis pathway (SSP), a highly regulated pathway overexpressed in several tumors. Specifically, PHGDH expression is dynamically regulated during different stages of tumor progression, promoting cancer aggressiveness. Previously, we demonstrated that high Serine (Ser) availability, obtained by increased exogenous uptake or increased PHGDH expression, supports 5-Fluorouracil (5-FU) resistance in colorectal cancer (CRC). Beyond its metabolic role in sustaining Ser biosynthesis, different \"non-enzymatic roles\" for PHGDH have recently been identified. The present study aims to investigate non-enzymatic mechanisms through which PHGDH regulates 5-FU response in CRC.</p><p><strong>Methods: </strong>Overexpression and gene silencing approaches have been used to modulate PHGDH expression in human CRC cell lines to investigate the role of this enzyme in 5-FU cellular response. Identified mechanisms have been validated in selected 5-FU resistant cell lines, CRC patients-derived tumor tissue samples, and patients-derived 3D organoids. Transcriptomic analysis was performed on wild-type and PHGDH-silenced cell lines, allowing the identification of pathways responsible for PHGDH-mediated 5-FU resistance. The relevance of identified genes was validated in vitro and in vivo in a CRC xenograft model.</p><p><strong>Results: </strong>PHGDH expression is highly variable among CRC tissues and patient-derived 3D organoids. A retrospective analysis of CRC patients highlighted a correlation between PHGDH expression and therapy response. Coherently, the modulation of PHGDH expression by gene silencing/overexpression affects 5-FU sensitivity in CRC cell lines. Transcriptomic analysis on CRC cell lines stably silenced for PHGDH evidenced down regulation in Hedgehog (HH) pathway. Accordingly, in vitro and in vivo studies demonstrated that the combined treatment of 5-FU and HH pathway inhibitors strongly hinders CRC cell survival and tumor growth in CRC xenograft models.</p><p><strong>Conclusions: </strong>PHGDH sustains 5-FU resistance in CRC by mediating the upregulation of the HH signaling; targeting the here identified PHGDH-HH axis increases 5-FU susceptibility in different CRC models suggesting the 5-FU/HH-inhibitors combinatorial therapeutic strategy as a valid approach to counteract drug resistance in CRC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"198"},"PeriodicalIF":11.4,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12243184/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144610236","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
Hypoxia-inducible APCDD1L-AS1 promotes osimertinib resistance by stabilising DLST to drive tricarboxylic acid cycle in lung adenocarcinoma. 缺氧诱导的APCDD1L-AS1通过稳定DLST驱动肺腺癌三羧酸循环促进奥希替尼耐药。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-09 DOI: 10.1186/s13046-025-03462-z
Quanli Zhang, Ye Shen, Yuru Che, Lili Jia, Xiang Xiao, Hao Xu, Chi Su, Kemin Sun, Limin Zheng, Jiawen Xu, Jingwen Hu, Chaofeng Zhang, Dihan Zhu, Ming Li
{"title":"Hypoxia-inducible APCDD1L-AS1 promotes osimertinib resistance by stabilising DLST to drive tricarboxylic acid cycle in lung adenocarcinoma.","authors":"Quanli Zhang, Ye Shen, Yuru Che, Lili Jia, Xiang Xiao, Hao Xu, Chi Su, Kemin Sun, Limin Zheng, Jiawen Xu, Jingwen Hu, Chaofeng Zhang, Dihan Zhu, Ming Li","doi":"10.1186/s13046-025-03462-z","DOIUrl":"10.1186/s13046-025-03462-z","url":null,"abstract":"<p><p>Acquired resistance is unavoidable in lung adenocarcinoma (LUAD) treated with osimertinib, however, the underlying mechanisms remain largely unknown. Here, we report that the long non-coding RNA (lncRNA) APCDD1L-AS1 is upregulated in osimertinib-resistant LUAD tissues and cells and is associated with short survival of osimertinib-resistant LUAD patients. Our data showed that APCDD1L-AS1 upregulation is an independent risk factor for overall survival in patients with osimertinib-resistant LUAD. APCDD1L-AS1 knockdown enhanced osimertinib sensitivity both in vitro and in vivo, whereas APCDD1L-AS1 overexpression promoted osimertinib resistance. Mechanistically, APCDD1L-AS1 accelerates the tricarboxylic acid (TCA) cycle by forming complexes and maintaining the stability of dihydrolipoamide S-succinyltransferase (DLST), which inhibits the ubiquitination and degradation of DLST. Moreover, we demonstrate that hypoxia-inducible factor (HIF)-1α transcriptionally activates APCDD1L-AS1 by binding to the APCDD1L-AS1 promoter region under hypoxic conditions. Overall, our data confirm that APCDD1L-AS1 is upregulated by hypoxia-induced HIF-1α, which drives the TCA cycle by stabilising DLST to further promote osimertinib resistance in LUAD. Our findings provide new insights into the role of HIF-1α/APCDD1L-AS1/DLST axis-related reprogramming of hypoxia and the TCA balance in conferring osimertinib resistance in LUAD and confirm the therapeutic potential for targeting the APCDD1L-AS1.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"197"},"PeriodicalIF":11.4,"publicationDate":"2025-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12239447/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144602067","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
Single-cell transcriptome analysis reveals the malignant characteristics of tumour cells and the immunosuppressive landscape in HER2-positive inflammatory breast cancer. 单细胞转录组分析揭示了her2阳性炎性乳腺癌肿瘤细胞的恶性特征和免疫抑制景观。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-08 DOI: 10.1186/s13046-025-03454-z
Juan Huang, Yongwei Zhu, Wenjing Zeng, Yulong Zhang, Weizhi Xia, Fan Xia, Liyu Liu, Kuansong Wang, Yidi Guan, Taohong Shen, Bingjian Jiang, Lunquan Sun, Ayong Cao, Shouman Wang, Zhi Li
{"title":"Single-cell transcriptome analysis reveals the malignant characteristics of tumour cells and the immunosuppressive landscape in HER2-positive inflammatory breast cancer.","authors":"Juan Huang, Yongwei Zhu, Wenjing Zeng, Yulong Zhang, Weizhi Xia, Fan Xia, Liyu Liu, Kuansong Wang, Yidi Guan, Taohong Shen, Bingjian Jiang, Lunquan Sun, Ayong Cao, Shouman Wang, Zhi Li","doi":"10.1186/s13046-025-03454-z","DOIUrl":"10.1186/s13046-025-03454-z","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Inflammatory breast cancer (IBC), of which HER2 + is the predominant subtype, is extremely aggressive and difficult to treat. Previous studies have suggested that targeting the tumour microenvironment (TME) may provide new directions for IBC diagnosis and treatment.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;In this study, we used single-cell transcriptome technology (scRNA-seq) to investigate the molecular features of the TME of HER2 + IBC patients and performed a comprehensive and detailed comparison of the cellular components and molecular phenotypes of the TME between IBC patients and noninflammatory breast cancer (nIBC) patients to elucidate the cell types that are specifically enriched in the TME of IBC patients, as well as the molecular features that are responsible for the preferential remodelling of the cellular functional state in the TME.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;A total of 15,832 cells, including epithelial cells, endothelial cells, stromal cells, T cells, B cells, antibody secreting cells (ASCs) and myeloid cells, were obtained from tumour tissues from 3 HER2 + IBC patients for scRNA analysis. By comparing the TME with that of HER2 + nIBC patients in a public database, we found that the TME of HER2 + IBC patients had a greater level of lymphocyte infiltration than that of nIBC patients did, and an especially significant enrichment of ASCs (mainly plasmablasts or plasma cells). In the TME of HER2 + IBC patients, tumour-infiltrating T cells exhibited a dual molecular phenotype of high activation and high exhaustion, with tumour-infiltrating B cells preferring the extrafollicular developmental pathway, and tumour-infiltrating myeloid and mesenchymal cells exhibiting a greater immunosuppressive status. By performing a cellular interaction analysis, we revealed that PTN molecules were significantly overexpressed in HER2 + IBC tumour cells and that the cellular interactions mediated by these molecules were strongly correlated with the functional polarisation of the cellular components in the TME. We observed that HER2 + IBC tumour cells have an active interferon response and epithelial mesenchymal transition (EMT) signalling, and that their malignant process is strongly correlated with the inflammatory response. Moreover, we found that HER2 + IBC tumour-infiltrating B cells promoted necroptosis of endothelial cells through high expression of TNF, thus promoting inflammatory responses.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;We found a strong correlation between high expression of PTN molecules in HER2 + IBC tumour cells and their highly invasive characteristics and highly immunosuppressive TME. These results suggest that HER2 + IBC tumour cells can promote an inflammatory response by upregulating the expression of TNF molecules in B cells via PTN molecules and that the enhanced inflammatory response in turn promotes tumour progression, a malignant cycle that shapes a more immunosuppressive TME. Therefore, diagnostic ","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"196"},"PeriodicalIF":11.4,"publicationDate":"2025-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12235857/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144585523","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
Genomic profiling of a collection of patient-derived xenografts and cell lines identified ixabepilone as an active drug against chemo-resistant osteosarcoma. 一组患者来源的异种移植物和细胞系的基因组分析鉴定了伊沙皮酮是一种抗化疗耐药骨肉瘤的活性药物。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-08 DOI: 10.1186/s13046-025-03440-5
Maria Cristina Manara, Francesca Bruzzese, Laura Formentini, Lorena Landuzzi, Laura Pazzaglia, Maria Antonella Laginestra, Marianna Carrabotta, Maria Serena Roca, Federica Iannelli, Laura Grumetti, Laura Addi, Alessandro Parra, Camilla Cristalli, Michela Pasello, Alberto Bavelloni, Francesca Carreras, Francesca Ruzzi, Giuseppe Bianchi, Marco Gambarotti, Alberto Righi, Alfredo Budillon, Pier-Luigi Lollini, Katia Scotlandi
{"title":"Genomic profiling of a collection of patient-derived xenografts and cell lines identified ixabepilone as an active drug against chemo-resistant osteosarcoma.","authors":"Maria Cristina Manara, Francesca Bruzzese, Laura Formentini, Lorena Landuzzi, Laura Pazzaglia, Maria Antonella Laginestra, Marianna Carrabotta, Maria Serena Roca, Federica Iannelli, Laura Grumetti, Laura Addi, Alessandro Parra, Camilla Cristalli, Michela Pasello, Alberto Bavelloni, Francesca Carreras, Francesca Ruzzi, Giuseppe Bianchi, Marco Gambarotti, Alberto Righi, Alfredo Budillon, Pier-Luigi Lollini, Katia Scotlandi","doi":"10.1186/s13046-025-03440-5","DOIUrl":"10.1186/s13046-025-03440-5","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"195"},"PeriodicalIF":11.4,"publicationDate":"2025-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12235892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144585522","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
Deciphering the tumor immune microenvironment: single-cell and spatial transcriptomic insights into cervical cancer fibroblasts. 解读肿瘤免疫微环境:宫颈癌成纤维细胞的单细胞和空间转录组学见解。
IF 12.8 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-05 DOI: 10.1186/s13046-025-03432-5
Zhiheng Lin, Youwei Zhou, Zhenran Liu, Wenyang Nie, Hengjie Cao, Shengnan Li, Xuanling Li, Lijun Zhu, Guangyao Lin, Yanyu Ding, Yi Jiang, Zuxi Gu, Lianwei Xu, Zhijie Zhao, Huabao Cai
{"title":"Deciphering the tumor immune microenvironment: single-cell and spatial transcriptomic insights into cervical cancer fibroblasts.","authors":"Zhiheng Lin, Youwei Zhou, Zhenran Liu, Wenyang Nie, Hengjie Cao, Shengnan Li, Xuanling Li, Lijun Zhu, Guangyao Lin, Yanyu Ding, Yi Jiang, Zuxi Gu, Lianwei Xu, Zhijie Zhao, Huabao Cai","doi":"10.1186/s13046-025-03432-5","DOIUrl":"10.1186/s13046-025-03432-5","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Cervical cancer (CC) remains a significant global health challenge despite advancements in screening, HPV vaccination, and therapeutic strategies. Tumor heterogeneity, driven by epigenetic modifications, affects immune evasion, metastasis, and treatment response. Cancer-associated fibroblasts (CAFs) play a crucial role in CC progression and therapy resistance. Single-cell sequencing offers new insights but remains underutilized in CC research. This study integrates single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, and deconvolution analysis to identify key genes and immunotherapy targets. By constructing a prognostic model and exploring the immune microenvironment, we aim to provide novel insights into CC pathogenesis and potential therapeutic strategies.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;We utilized scRNA-seq, spatial transcriptomics, deconvolution analysis, and pseudotime trajectory mapping to delineate fibroblast subtypes within the tumor immune microenvironment (TIME) of CC. Functional annotations, differential gene expression profiling, cell-cell communication pathways, and transcription factor networks were systematically analyzed. A prognostic model based on bulk RNA-seq data was constructed and validated through survival analysis, with correlations to immune microenvironment characteristics. Functional experiments investigated the role of SDC1, a critical mediator of fibroblast-tumor crosstalk. Additionally, Fibroblast-tumor cell co-culture systems and functional assays were employed to investigate the paracrine role of SDC1. The CAF MYH11⁺ subpopulation was isolated via fluorescence-activated cell sorting (FACS). Multiplex immunofluorescence and immunohistochemical analyses were performed on both cultured cells and human cervical cancer tissue samples to characterize the spatial distribution and dynamic remodeling of MYH11 during stromal reorganization.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Six distinct fibroblast subtypes were identified, including the C0 MYH11 + fibroblasts, which exhibited unique roles in stemness maintenance, metabolic activity, and immune regulation. Spatial and functional analyses revealed that the C0 subtype is central to tumor-fibroblast interactions, particularly through the MDK-SDC1 signaling axis. The prognostic model incorporating fibroblast-specific markers demonstrated robust predictive power for patient survival outcomes. Additionally, in vitro SDC1 knockdown significantly inhibited CC cell proliferation, migration, and invasion. Fibroblasts show spatially regulated heterogeneity, with activation markers enriched in the tumor zone and MYH11 highest in normal zones, indicating dynamic stromal remodeling. C0 MYH11 + CAF Promotes Tumor Cell Proliferation, Migration, and Inhibits Apoptosis via Soluble SDC1.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;Our results illustrate, in some ways, the possible immunomodulatory and tumor supporting roles of CAFs in CC TIME and highlight the ","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"194"},"PeriodicalIF":12.8,"publicationDate":"2025-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12228347/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565385","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
Adipocytes-induced ANGPTL4/KLF4 axis drives glycolysis and metastasis in triple-negative breast cancer. 脂肪细胞诱导的ANGPTL4/KLF4轴驱动三阴性乳腺癌的糖酵解和转移。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-04 DOI: 10.1186/s13046-025-03458-9
Dou Yin, Nana Fang, Yaling Zhu, Xiaoqing Bao, Juan Yang, Qingyu Zhang, Ruimeng Wang, Jiahui Huang, Qibing Wu, Fang Ma, Xiaohui Wei
{"title":"Adipocytes-induced ANGPTL4/KLF4 axis drives glycolysis and metastasis in triple-negative breast cancer.","authors":"Dou Yin, Nana Fang, Yaling Zhu, Xiaoqing Bao, Juan Yang, Qingyu Zhang, Ruimeng Wang, Jiahui Huang, Qibing Wu, Fang Ma, Xiaohui Wei","doi":"10.1186/s13046-025-03458-9","DOIUrl":"10.1186/s13046-025-03458-9","url":null,"abstract":"<p><strong>Background: </strong>The adipocyte-rich tumor microenvironment (TME) is recognized as a key factor in promoting cancer progression. A distinct characteristic of peritumoral adipocytes is their reduced lipid content and the acquisition of a proinflammatory phenotype. However, the underlying mechanisms by which adipocytes rewire metabolism and boost tumor progression in triple-negative breast cancer (TNBC) remain poorly understood.</p><p><strong>Methods: </strong>We utilized transcriptomic analysis, bioinformatic analysis, metabolic flux analysis, protein-protein docking, gene and protein expression profiling, in vivo metastasis analysis and breast cancer specimens to explore how adipocytes reprogram tumor metabolism and progression in TNBC.</p><p><strong>Results: </strong>Our findings reveal that Angiopoietin-like 4 (ANGPTL4) exhibits significantly higher expression levels in adipocyte-rich tumor circumstance compared to the symbiotic environment lacking of adipocyte. Furthermore, ANGPTL4 expression in tumor cells is essential for adipocyte-driven glycolysis and metastasis. Interleukin 6 (IL-6), enriched in cancer-associated adipocytes, and lipolysis-derived free fatty acids (FFAs) released from adipocytes, amplify ANGPTL4-mediated glycolysis and metastasis through activation of STAT3 and PPARα pathways in TNBC cells. Additionally, ANGPTL4 interacts with transcription factor KLF4 and enhances KLF4 activity, which further drives glycolysis and metastasis, whereas KLF4 knockdown attenuates migration and glycolysis in TNBC cells. Importantly, Elevated ANGPTL4 and KLF4 expression was observed in metastatic breast cancer specimens compared to non-metastatic cases and was positively correlated with poor prognosis.</p><p><strong>Conclusion: </strong>Collectively, our results uncover a complex metabolic interaction between adipocytes and TNBC cells that promotes tumor aggressiveness. ANGPTL4 emerges as a key mediator in this process, making it a promising therapeutic target to inhibit TNBC progression.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"192"},"PeriodicalIF":11.4,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12231887/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565384","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
M2 macrophage-secreted KYNU promotes stemness remodeling and malignant behavior in endometrial cancer via the SOD2-mtROS-ERO1α-UPRER axis. M2巨噬细胞分泌的KYNU通过SOD2-mtROS-ERO1α-UPRER轴促进子宫内膜癌的干性重塑和恶性行为。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-04 DOI: 10.1186/s13046-025-03285-y
Xin Pan, Wantong Wang, Yuting Wang, JiaHui Gu, Xiaoxin Ma
{"title":"M2 macrophage-secreted KYNU promotes stemness remodeling and malignant behavior in endometrial cancer via the SOD2-mtROS-ERO1α-UPR<sup>ER</sup> axis.","authors":"Xin Pan, Wantong Wang, Yuting Wang, JiaHui Gu, Xiaoxin Ma","doi":"10.1186/s13046-025-03285-y","DOIUrl":"10.1186/s13046-025-03285-y","url":null,"abstract":"<p><strong>Background: </strong>M2 macrophages are known to be involved in tumorigenesis. However, the mechanism by which they promote tumor progression in endometrial cancer (EC) remains largely unknown. Kynureninase (KYNU) has been found to be associated with the progression of various tumors, but research on endometrium is limited to embryo transfer. Therefore, a better understanding of KYNU as a potential therapeutic target in EC treatment is needed. This study aimed to elucidate the mechanism by which M2 macrophage-secreted KYNU influences the malignant biological and stemness remodeling of EC via the SOD2-mtROS-ERO1α and endoplasmic reticulum unfolded protein response (UPR<sup>ER</sup>) pathway.</p><p><strong>Methods: </strong>We used flow cytometry for cell sorting. Fluorescence experiments were conducted to reveal spatial position of protein, and. Western blot and qRT‒PCR were used to detect the protein and mRNA levels, respectively. The interaction between KYNU and superoxide dismutase 2 (SOD2) was demonstrated using coimmunoprecipitation experiments. Furthermore, the mechanism between activating transcription factor 4 (ATF4) and the KYNU was assessed using chromatin immunoprecipitation and dual luciferase assays. Cell Counting Kit-8, flow cytometry, and transwell assays were used to detect tumor cell proliferation, apoptosis, and invasion capacities. Student's t test and one-way analysis of variance (ANOVA) were used to compare groups.</p><p><strong>Results: </strong>M2 macrophage-secreted KYNU induced malignant behavior and stemness via the SOD2-mtROS-ERO1α-UPR<sup>ER</sup> pathway, contributing to a positive feedback loop for tumor cell self-protection. Mechanistically, KYNU and its metabolite 3-hydroxyanthranillic acid (3-HAA) upregulated the expression of SOD2, thereby decreasing mitochondrial reactive oxygen species (mtROS). KYNU inhibitors affected the spatial overflow of mtROS from mitochondria to the endoplasmic reticulum (ER). Endoplasmic reticulum oxidoreductin 1α (ERO1α) was sensitively affected by KYNU-induced changes in the redox environment, stimulating the PERK-eIF2α-ATF4 pathway of the UPR<sup>ER</sup>. This in turn promoted oxidative folding, reduced the level of misfolded protein (MFP), and maintained tumor survival and progression. Additionally, ATF4 acted as a transcription factor in the KYNU promoter region, amplifying KYNU tumorigenesis in a positive feedback manner.</p><p><strong>Conclusion: </strong>M2-secreted KYNU promotes the malignant behavior and stemness remodeling of EC via the SOD2-mtROS-ERO1α-UPR<sup>ER</sup> axis and establishes a positive feedback loop. Thus, KYNU is a potential therapeutic target for EC treatment.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"193"},"PeriodicalIF":11.4,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12231660/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565400","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 reprogramming impact of SMAC-mimetic on glioblastoma stem cells and the immune tumor microenvironment evolution. smac -拟态基因重编程对胶质母细胞瘤干细胞的影响及免疫肿瘤微环境演化。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-04 DOI: 10.1186/s13046-025-03452-1
Qiong Wu, Jianan Chen, Anders E Berglund, Dongliang Du, Robert J Macaulay, Arnold B Etame
{"title":"The reprogramming impact of SMAC-mimetic on glioblastoma stem cells and the immune tumor microenvironment evolution.","authors":"Qiong Wu, Jianan Chen, Anders E Berglund, Dongliang Du, Robert J Macaulay, Arnold B Etame","doi":"10.1186/s13046-025-03452-1","DOIUrl":"10.1186/s13046-025-03452-1","url":null,"abstract":"<p><strong>Background: </strong>Intrinsically resistant glioma stem cells (GSCs) in the setting of a highly immunosuppressive tumor microenvironment (TME) remain the most predominant phenomenon leading to unfavorable therapeutic outcomes in glioblastoma (GBM). Hence there is an unmet need for novel anti-GBM therapeutic paradigms that can effectively target GSCs while simultaneously reprogramming the TME.</p><p><strong>Methods: </strong>In this study, we leverage evidence from SMAC mimetic screening to evaluate and characterize the anti-tumor and immune TME modulating impacts of the lead SMAC mimetic Xevinapant at the single cell level in GBM. We utilized viability assays and orthotopic human and murine GBM models to assess the survival impacts of Xevinapant on GSCs in vitro and in vivo. Moreover, we employed single-cell RNA sequencing (scRNA-seq) to investigate the modulation impact of Xevinapant on GBM TME. Lastly, we investigated drug combination synergies to address potential mechanisms of tolerance or resistance to Xevinapant.</p><p><strong>Results: </strong>According to our observations, in vitro exposure to Xevinapant induced apoptosis along with significant viability reduction in a dose-dependent manner, in both human and mouse GSCs. Moreover, Xevinapant treatment produced robust anti-tumor effects in vivo and significantly prolonged animal overall survival. Based on single-cell RNA seq analysis, Xevinapant did not only enhance GSCs apoptosis but also activated antitumor effector immune response leading to favorable reprogramming of immunosuppressive TME. Furthermore, we established and queried Xevinapant therapeutic signatures to the LINCS database in an effort to identify small molecules that could reverse treatment-induced tolerance to Xevinapant. We have identified a novel set of candidate small molecules with robust synergy when combined with Xevinapant.</p><p><strong>Conclusions: </strong>In summary, Xevinapant exhibits robust anti-tumor activity on GSCs and favorable immune modulation of the TME in GBM, hence providing a rationale for further clinical investigation in GBM.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"191"},"PeriodicalIF":11.4,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12231904/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565401","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
Amino acid transporter LAT1 (SLC7A5) promotes metabolic rewiring in TNBC progression through the L-Trp/QPRT/NAD+ pathway. 氨基酸转运体LAT1 (SLC7A5)通过L-Trp/QPRT/NAD+途径促进TNBC进展中的代谢重新布线。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2025-07-03 DOI: 10.1186/s13046-025-03446-z
Margot Y Fedoroff, Lei Zhao, Shaomin Wang, Alok Bhushan, Haifeng Yang, Karen M Bussard, Stephen C Peiper, Jun He
{"title":"Amino acid transporter LAT1 (SLC7A5) promotes metabolic rewiring in TNBC progression through the L-Trp/QPRT/NAD<sup>+</sup> pathway.","authors":"Margot Y Fedoroff, Lei Zhao, Shaomin Wang, Alok Bhushan, Haifeng Yang, Karen M Bussard, Stephen C Peiper, Jun He","doi":"10.1186/s13046-025-03446-z","DOIUrl":"10.1186/s13046-025-03446-z","url":null,"abstract":"<p><strong>Background: </strong>Cancer cells uptake excessive nutrients by expressing higher levels of glucose and/or amino acid transporters to meet their increased energy demands. L-type amino acid transporter 1 (LAT1), is regarded as a cancer-specific transporter for the uptake of large neutral amino acids such as L-tryptophan. However, the mechanism by which LAT1 rewires cellular metabolism to promote cancer progression and chemoresistance have not yet been investigated.</p><p><strong>Methods: </strong>The protein levels of LAT1, p-PKM2, and p-LDHA were determined in breast cancer tissue arrays by immunohistochemistry staining followed by survival analysis. The orthotopic breast cancer models in mice, syngeneic breast cancer models, and patient-derived xenograft (PDX) mouse models were used to study the effects of LAT1 inhibition in tumor growth and chemoresistance. Steady-state polar metabolite analysis was performed to profile changes in cellular metabolism by LC-MS. The pyruvate and lactate assays as well as the seahorse assay using LAT1 knockdown cells and control cells were conducted to evaluate cellular glycolytic activities.</p><p><strong>Results: </strong>The LAT1 protein levels were positively correlated with poor survival in triple-negative breast cancer (TNBC) patients. LAT1 silencing resulted in reduced TNBC cell viability, proliferation, migration, invasion in vitro, as well as tumor growth in vivo. The knockdown of LAT1 reduced glycolytic activities via activating PKM2 and LDHA, two key glycolytic enzymes essential for cancer cell growth. Mechanistically, we demonstrated that LAT1 promoted de novo NAD + synthesis by facilitating L-tryptophan uptake and upregulating quinolinate phosphoribosyltransferase (QPRT), the rate-limiting enzyme in this pathway. This resulted in an increased cytosolic NAD<sup>+</sup>/NADH ratio, which enhanced the phosphorylation of pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA), thus promoting TNBC tumor progression. Notably, upregulation of this pathway was observed in primary cells from doxorubicin (Dox)-resistant TNBC patient-derived xenograft (PDX) tumors and in Dox-resistant MDA-MB-231 cells. LAT1 inhibition sensitized resistant cells to Dox-induced cytotoxicity while supplementation of L-Trp/NAD + partially reversed the enhanced sensitivity to Doxorubicin induced by LAT1 knockdown. Furthermore, treatment with a LAT1-specific inhibitor JPH203 synergistically enhanced the efficacy of doxorubicin in TNBC cells.</p><p><strong>Conclusion: </strong>These findings identify a novel role of LAT1 in promoting TNBC progression and chemo-resistance by amplifying the Warburg effect, positioning LAT1 as a promising therapeutic target for TNBC treatment.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"190"},"PeriodicalIF":11.4,"publicationDate":"2025-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12224598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144561825","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