Pub Date : 2026-01-09DOI: 10.1186/s12935-025-04133-x
Haojie Dai, Xi Zhang, Lu Yin, Hongqi Chen, Kui Liu, Jian Li, Heng Li, Lian Sheng, Hongfei Wu, Jiawei Wang, Shaohua He, Qiang Li, Yang Lv
Background: Treg infiltration and programmed cell death are important factors influencing cancer progression, and they interact with each other. However, the significance of Treg-related programmed cell death (PCD) characteristics in clear cell renal cell carcinoma remains unclear.
Methods: Through Mendelian randomization, we identified PCD genes and Treg markers that are highly associated with ccRCC outcomes. Subsequently, based on Treg-related PCD genes, we constructed a diagnostic model utilizing a multi-layer perceptron (MLP) and integrated 10 machine learning algorithms to construct a prognostic model, which was then explained by the SHAP method. After exploring functional differences and chemotherapy sensitivity differences between high- and low-risk groups in the prognostic model, we validated the core gene of the model through in vitro cell experiments. Finally, we screened molecular drugs targeting the core genes using the DSigDB database and performed molecular docking and molecular dynamics validation.
Results: Utilizing Mendelian randomization (MR), we first established causal links between specific Treg subtypes and PCD gene CASP9 with renal cancer outcomes. Leveraging shared Treg-PCD molecular features, we developed a MLP-based diagnostic model achieving an AUC of 0.987 in external validation. Further, a robust prognostic index Treg-Programmed Cell Death Score (TPCDS) was constructed using 101 machine learning combinations, demonstrating superior stratification across multi-cohort data. High TPCDS correlated with immunosuppressive microenvironments including increased Tregs, T-cell exhaustion, HLA downregulation and poor immunotherapy response, while guiding chemotherapy sensitivity. Functional assays confirmed the core gene SLC11A1 as an oncogenic driver promoting proliferation, migration, and invasion. Molecular docking and dynamics simulations identified Atovaquone as a high-affinity inhibitor of SLC11A1.
Conclusion: We explored the significance of Treg and programmed cell death characteristics in the ccRCC tumor microenvironment and established clinically translatable tools for ccRCC diagnosis, prognosis, and personalized therapy selection, thus promoted the application of explainable machine learning models in precision oncology. Furthermore, We have identified SLC11A1 as a highly promising therapeutic target for ccRCC.
{"title":"Integrating machine learning and multi-omics analysis to explore Treg-associated programmed cell death features in clear cell renal cell carcinoma.","authors":"Haojie Dai, Xi Zhang, Lu Yin, Hongqi Chen, Kui Liu, Jian Li, Heng Li, Lian Sheng, Hongfei Wu, Jiawei Wang, Shaohua He, Qiang Li, Yang Lv","doi":"10.1186/s12935-025-04133-x","DOIUrl":"10.1186/s12935-025-04133-x","url":null,"abstract":"<p><strong>Background: </strong>Treg infiltration and programmed cell death are important factors influencing cancer progression, and they interact with each other. However, the significance of Treg-related programmed cell death (PCD) characteristics in clear cell renal cell carcinoma remains unclear.</p><p><strong>Methods: </strong>Through Mendelian randomization, we identified PCD genes and Treg markers that are highly associated with ccRCC outcomes. Subsequently, based on Treg-related PCD genes, we constructed a diagnostic model utilizing a multi-layer perceptron (MLP) and integrated 10 machine learning algorithms to construct a prognostic model, which was then explained by the SHAP method. After exploring functional differences and chemotherapy sensitivity differences between high- and low-risk groups in the prognostic model, we validated the core gene of the model through in vitro cell experiments. Finally, we screened molecular drugs targeting the core genes using the DSigDB database and performed molecular docking and molecular dynamics validation.</p><p><strong>Results: </strong>Utilizing Mendelian randomization (MR), we first established causal links between specific Treg subtypes and PCD gene CASP9 with renal cancer outcomes. Leveraging shared Treg-PCD molecular features, we developed a MLP-based diagnostic model achieving an AUC of 0.987 in external validation. Further, a robust prognostic index Treg-Programmed Cell Death Score (TPCDS) was constructed using 101 machine learning combinations, demonstrating superior stratification across multi-cohort data. High TPCDS correlated with immunosuppressive microenvironments including increased Tregs, T-cell exhaustion, HLA downregulation and poor immunotherapy response, while guiding chemotherapy sensitivity. Functional assays confirmed the core gene SLC11A1 as an oncogenic driver promoting proliferation, migration, and invasion. Molecular docking and dynamics simulations identified Atovaquone as a high-affinity inhibitor of SLC11A1.</p><p><strong>Conclusion: </strong>We explored the significance of Treg and programmed cell death characteristics in the ccRCC tumor microenvironment and established clinically translatable tools for ccRCC diagnosis, prognosis, and personalized therapy selection, thus promoted the application of explainable machine learning models in precision oncology. Furthermore, We have identified SLC11A1 as a highly promising therapeutic target for ccRCC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":"15"},"PeriodicalIF":6.0,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12801888/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1186/s12935-025-04160-8
Chunyan Zhang, Bing Han, Zhihua Jia, Min Liu, Yanxia Li, Jie Xu, Jie Zheng, Yajing Sun, Yuting Le, Shupeng Zhang, Zhijiang Shao, Jian Wang, Xiaozhi Liu
{"title":"Molecular subtype-specific breast cancer organoids: development of an FGF-free estradiol valerate culture system for precision drug screening.","authors":"Chunyan Zhang, Bing Han, Zhihua Jia, Min Liu, Yanxia Li, Jie Xu, Jie Zheng, Yajing Sun, Yuting Le, Shupeng Zhang, Zhijiang Shao, Jian Wang, Xiaozhi Liu","doi":"10.1186/s12935-025-04160-8","DOIUrl":"https://doi.org/10.1186/s12935-025-04160-8","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932402","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1186/s12935-025-04117-x
Dongbo Xu, Justine J Jacob, Kyle Wieczorek, Li Wang, Han Yu, Jianmin Wang, Bo Xu, Ahmed A Hussein, Khurshid Guru, David W Goodrich, Qiang Li
Approximately 90% of bladder cancer deaths are due to distant metastases rather than local tumor effects. The current first-line systemic treatment for metastatic bladder cancer (mBC) is chemoimmunotherapy or immunotherapy with pembrolizumab plus enfortumab vedotin (EV). However, most mBC patients treated with chemoimmunotherapy or pembrolizumab/EV do not respond or eventually relapse, highlighting the critical need for robust immunocompetent animal models to elucidate the mechanisms of primary and acquired resistance. We previously generated a syngeneic murine cell line CMV-TRP (triple knockout of Trp53, Rb1, Pten) via ex vivo transduction with adenovirus (Ad5CMVCre). To establish an mBC model, the TKO cells were effectively labeled with a lentiviral luciferase and GFP double-expressing reporter and injected into tail veins of C57 BL/6J mice. Tail vein injection of TKO cells reliably established distant metastases with lung and bone lesions. In immunotherapy experiments, mice injected with TKO-labeled cell lines were randomly treated with an anti-PD-1 or control IgG2a antibody. All mice developed lung and/or bone (hind limb or sacrum) metastases. There was no difference in tumor bioluminescence between the control group and anti-PD-1 group (median proton/second 6.94 × 108 vs. 4.32 × 108, p = 0.85). Kaplan-Meier analysis showed no difference in median survival between the control group and anti-PD-1 group (19 days vs. 20 days, p = 0.47). Histology and immunohistochemical profile of lung and bone metastases revealed high-grade basal-like urothelial carcinoma, closely resembling the profile observed in subcutaneous tumor. No significant changes in immune cell infiltrations (CD4+, CD8+, or F4/80+) between groups may explain anti-PD-1 immunotherapy resistance. Therefore, the novel TKO metastatic model represents a useful and reproducible tool for studying tumor-cell dissemination, bone/lung metastasis, and the underlying mechanisms of anti-PD-1 immunotherapy resistance.
{"title":"Modeling primary immunotherapy resistance in metastatic bladder cancer: a syngeneic, bioluminescent mouse model.","authors":"Dongbo Xu, Justine J Jacob, Kyle Wieczorek, Li Wang, Han Yu, Jianmin Wang, Bo Xu, Ahmed A Hussein, Khurshid Guru, David W Goodrich, Qiang Li","doi":"10.1186/s12935-025-04117-x","DOIUrl":"https://doi.org/10.1186/s12935-025-04117-x","url":null,"abstract":"<p><p>Approximately 90% of bladder cancer deaths are due to distant metastases rather than local tumor effects. The current first-line systemic treatment for metastatic bladder cancer (mBC) is chemoimmunotherapy or immunotherapy with pembrolizumab plus enfortumab vedotin (EV). However, most mBC patients treated with chemoimmunotherapy or pembrolizumab/EV do not respond or eventually relapse, highlighting the critical need for robust immunocompetent animal models to elucidate the mechanisms of primary and acquired resistance. We previously generated a syngeneic murine cell line CMV-TRP (triple knockout of Trp53, Rb1, Pten) via ex vivo transduction with adenovirus (Ad5CMVCre). To establish an mBC model, the TKO cells were effectively labeled with a lentiviral luciferase and GFP double-expressing reporter and injected into tail veins of C57 BL/6J mice. Tail vein injection of TKO cells reliably established distant metastases with lung and bone lesions. In immunotherapy experiments, mice injected with TKO-labeled cell lines were randomly treated with an anti-PD-1 or control IgG2a antibody. All mice developed lung and/or bone (hind limb or sacrum) metastases. There was no difference in tumor bioluminescence between the control group and anti-PD-1 group (median proton/second 6.94 × 10<sup>8</sup> vs. 4.32 × 10<sup>8</sup>, p = 0.85). Kaplan-Meier analysis showed no difference in median survival between the control group and anti-PD-1 group (19 days vs. 20 days, p = 0.47). Histology and immunohistochemical profile of lung and bone metastases revealed high-grade basal-like urothelial carcinoma, closely resembling the profile observed in subcutaneous tumor. No significant changes in immune cell infiltrations (CD4<sup>+</sup>, CD8<sup>+</sup>, or F4/80<sup>+</sup>) between groups may explain anti-PD-1 immunotherapy resistance. Therefore, the novel TKO metastatic model represents a useful and reproducible tool for studying tumor-cell dissemination, bone/lung metastasis, and the underlying mechanisms of anti-PD-1 immunotherapy resistance.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1186/s12935-025-04152-8
Giada Juli, Domenica Ronchetti, Stefania Signorelli, Sara Squillacioti, Nicoletta Polerà, Anna Maienza, Michelangelo Vocaturo, Antonio Galvano, Elisa Taiana, Pierosandro Tagliaferri, Pierfrancesco Tassone, Daniele Caracciolo
Background: Alternative Non-Homologous End Joining (Alt-NHEJ) DNA repair is considered a major player in cancer genomic instability. Here, we investigated cGAS-STING pathway as crucial node in the interplay between Alt-NHEJ repair and immune response, in the aim to discover novel therapeutic vulnerability in Multiple Myeloma (MM). METHODS: In silico analyses were performed by querying publicly available MM datasets (GSE66293 and CoMMpass). Anti-proliferative activity was evaluated by CellTiter-Glo, while flow cytometry analysis was used to determine the apoptotic process, cell cycle, phagocytosis, micronuclei detection, Calreticulin and T-cell activation markers. Protein expression was detected by western blot of whole or fractioned protein extracts.
Results: By interrogating public MM datasets, a significant correlation between hyperactivation of cGAS-STING mRNA signature and poor PFS and OS in MM was observed. Indeed, Gene Set Enrichment Analysis (GSEA) showed enrichment of DNA repair, TNFA signaling and oxidative phosphorylation in patients with cGAS-STING activation patients, associated to higher mRNA expression of DNA Ligase 3 (LIG3) and PARP1. On this basis, we evaluated the activity of Alt-NHEJ inhibitor Talazoparib (PARP1-inhibitor) on MM cell lines, focusing on their capability to modulate cGAS-STING pathway. We first detected a significant reduction of cell proliferation and the induction of apoptosis following Talazoparib treatment, which in turn induced DNA damage response and cell cycle blockade, and finally cGAS-STING pathway activation as result of PARP1-trapping into chromatin. Next, by performing co-culture experiments with healthy donor's peripheral blood mononuclear cells (PBMCs), we finally demonstrated the induction of immunogenic cell death, which was abrogated in cGAS-knockout cells, underscoring the pathway's functional relevance.
Conclusion: Taken together, our findings indicate that Alt-NHEJ inhibitors are potential immune-stimulating agents for MM with hyperactivation of cGAS-STING pathway, coherently with our working hypothesis.
{"title":"PARP1 trapping activates cGAS-STING pathway to induce immunogenic cell death in multiple myeloma.","authors":"Giada Juli, Domenica Ronchetti, Stefania Signorelli, Sara Squillacioti, Nicoletta Polerà, Anna Maienza, Michelangelo Vocaturo, Antonio Galvano, Elisa Taiana, Pierosandro Tagliaferri, Pierfrancesco Tassone, Daniele Caracciolo","doi":"10.1186/s12935-025-04152-8","DOIUrl":"https://doi.org/10.1186/s12935-025-04152-8","url":null,"abstract":"<p><strong>Background: </strong>Alternative Non-Homologous End Joining (Alt-NHEJ) DNA repair is considered a major player in cancer genomic instability. Here, we investigated cGAS-STING pathway as crucial node in the interplay between Alt-NHEJ repair and immune response, in the aim to discover novel therapeutic vulnerability in Multiple Myeloma (MM). METHODS: In silico analyses were performed by querying publicly available MM datasets (GSE66293 and CoMMpass). Anti-proliferative activity was evaluated by CellTiter-Glo, while flow cytometry analysis was used to determine the apoptotic process, cell cycle, phagocytosis, micronuclei detection, Calreticulin and T-cell activation markers. Protein expression was detected by western blot of whole or fractioned protein extracts.</p><p><strong>Results: </strong>By interrogating public MM datasets, a significant correlation between hyperactivation of cGAS-STING mRNA signature and poor PFS and OS in MM was observed. Indeed, Gene Set Enrichment Analysis (GSEA) showed enrichment of DNA repair, TNFA signaling and oxidative phosphorylation in patients with cGAS-STING activation patients, associated to higher mRNA expression of DNA Ligase 3 (LIG3) and PARP1. On this basis, we evaluated the activity of Alt-NHEJ inhibitor Talazoparib (PARP1-inhibitor) on MM cell lines, focusing on their capability to modulate cGAS-STING pathway. We first detected a significant reduction of cell proliferation and the induction of apoptosis following Talazoparib treatment, which in turn induced DNA damage response and cell cycle blockade, and finally cGAS-STING pathway activation as result of PARP1-trapping into chromatin. Next, by performing co-culture experiments with healthy donor's peripheral blood mononuclear cells (PBMCs), we finally demonstrated the induction of immunogenic cell death, which was abrogated in cGAS-knockout cells, underscoring the pathway's functional relevance.</p><p><strong>Conclusion: </strong>Taken together, our findings indicate that Alt-NHEJ inhibitors are potential immune-stimulating agents for MM with hyperactivation of cGAS-STING pathway, coherently with our working hypothesis.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932439","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1186/s12935-025-04154-6
Alessandra Lo Cicero, Francesco La Monica, Gabriele Lo Buglio, Simona Campora, Federico Gangemi, Paolo Cinà, Monica Salamone, Margot Lo Pinto, Simone Dario Scilabra, Giulio Ghersi
Background: Tumorigenesis is a complex and dynamic process in which the tumor microenvironment (TME) plays a central role. In solid tumors, the TME contributes to key mechanisms of tumor progression, including metastasis, immune evasion, and resistance to therapies. One major challenge in preclinical cancer research is the development of reliable three-dimensional (3D) in vitro models, which more accurately replicate the in vivo tumor architecture and microenvironmental conditions, such as hypoxia and extracellular matrix (ECM) organization. However, reproducing functional vascular networks and neo-angiogenesis within these models remains a key challenge.
Methods: In this study, an advanced 3D tumor model, referred to as angiotumoroids, was developed by co-culturing primary murine breast tumor cells (PTCs) with species-specific adipose-derived microvascular fragments (MVFs). Angiotumoroids were characterized using scanning electron microscopy and immunostaining, and angiogenesis was evaluated through collagen gel sprouting assays. High-resolution proteomic profiling was conducted, focusing on signatures associated with angiogenesis, extracellular matrix (ECM) composition, and tissue remodeling. Additionally, the response and internalization to anticancer drug treatments were evaluated.
Results: MVFs are successfully integrated in angiotumoroids, resulting in the formation of vasculature-like structures and demonstrating robust structural organization with dynamic modulation of matrix metalloproteinase 9. Formation of neovasculature was visualized through sprouting and branching, driven by both direct PTC-MVF interactions and PTC-conditioned media, highlighting the roles of juxtacrine and paracrine signaling. Proteomic profiling revealed distinct expression patterns associated with angiogenesis, ECM components (including collagen types I and IV), and active ECM remodeling with elevated MMP expression. Additionally, angiotumoroids showed increased expression of ATP-binding cassette (ABC) transporters, particularly ABCB1 (P-glycoprotein), suggesting potential mechanisms of drug efflux. Functionally, angiotumoroids demonstrated reduced sensitivity to doxorubicin compared to PTC spheroids, maintaining structural integrity and higher cell viability post-treatment. Time-course analysis revealed preferential doxorubicin accumulation in MVF-enriched regions, as confirmed by colocalization with CD31, indicating a spatially regulated distribution of the drug mediated by the vascular compartment.
Conclusions: Collectively, these findings establish angiotumoroids as a robust and physiologically relevant in vitro model for studying tumor vascularization, ECM dynamics, and therapeutic response. This platform holds significant promise for predictive cancer research and preclinical drug screening, bridging the gap between traditional in vitro systems and in vivo models.
{"title":"A vascularized three-dimensional model integrating primary breast tumor cells and microvascular fragments: mimicking the tumor microenvironment involved in chemoresistance.","authors":"Alessandra Lo Cicero, Francesco La Monica, Gabriele Lo Buglio, Simona Campora, Federico Gangemi, Paolo Cinà, Monica Salamone, Margot Lo Pinto, Simone Dario Scilabra, Giulio Ghersi","doi":"10.1186/s12935-025-04154-6","DOIUrl":"https://doi.org/10.1186/s12935-025-04154-6","url":null,"abstract":"<p><strong>Background: </strong>Tumorigenesis is a complex and dynamic process in which the tumor microenvironment (TME) plays a central role. In solid tumors, the TME contributes to key mechanisms of tumor progression, including metastasis, immune evasion, and resistance to therapies. One major challenge in preclinical cancer research is the development of reliable three-dimensional (3D) in vitro models, which more accurately replicate the in vivo tumor architecture and microenvironmental conditions, such as hypoxia and extracellular matrix (ECM) organization. However, reproducing functional vascular networks and neo-angiogenesis within these models remains a key challenge.</p><p><strong>Methods: </strong>In this study, an advanced 3D tumor model, referred to as angiotumoroids, was developed by co-culturing primary murine breast tumor cells (PTCs) with species-specific adipose-derived microvascular fragments (MVFs). Angiotumoroids were characterized using scanning electron microscopy and immunostaining, and angiogenesis was evaluated through collagen gel sprouting assays. High-resolution proteomic profiling was conducted, focusing on signatures associated with angiogenesis, extracellular matrix (ECM) composition, and tissue remodeling. Additionally, the response and internalization to anticancer drug treatments were evaluated.</p><p><strong>Results: </strong>MVFs are successfully integrated in angiotumoroids, resulting in the formation of vasculature-like structures and demonstrating robust structural organization with dynamic modulation of matrix metalloproteinase 9. Formation of neovasculature was visualized through sprouting and branching, driven by both direct PTC-MVF interactions and PTC-conditioned media, highlighting the roles of juxtacrine and paracrine signaling. Proteomic profiling revealed distinct expression patterns associated with angiogenesis, ECM components (including collagen types I and IV), and active ECM remodeling with elevated MMP expression. Additionally, angiotumoroids showed increased expression of ATP-binding cassette (ABC) transporters, particularly ABCB1 (P-glycoprotein), suggesting potential mechanisms of drug efflux. Functionally, angiotumoroids demonstrated reduced sensitivity to doxorubicin compared to PTC spheroids, maintaining structural integrity and higher cell viability post-treatment. Time-course analysis revealed preferential doxorubicin accumulation in MVF-enriched regions, as confirmed by colocalization with CD31, indicating a spatially regulated distribution of the drug mediated by the vascular compartment.</p><p><strong>Conclusions: </strong>Collectively, these findings establish angiotumoroids as a robust and physiologically relevant in vitro model for studying tumor vascularization, ECM dynamics, and therapeutic response. This platform holds significant promise for predictive cancer research and preclinical drug screening, bridging the gap between traditional in vitro systems and in vivo models.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932307","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-07DOI: 10.1186/s12935-025-04158-2
Aimin Guo, Shanshan Wang, Linan Huang, Yanping He, Qing Xi, David J Craik, Jun Zhang
Long non-coding RNAs (lncRNAs) are broad-spectrum cellular transcripts that can directly act as RNA regulators and/or partly encode functional peptides (lncRNA-encoded peptides, LRPs) in cancer cells. Recently, cancer LRPs have been found to be involved in cancer cell variability and proliferation, thus gaining widespread attention for their potential in cancer diagnosis, prognosis and therapy. As structures determine functions, the structural diversities of LRPs are the sources of functional variations of LRPs in cancers. Since 6135 cancer LRPs are listed in SPENCER database and 24 SPENCER-unlisted cancer LRPs are reported in several previous studies, this article reviews recent advances of cancer LRPs, analyzes amino acid compositions of them, and undertakes in silico evaluations to assess their structural and functional attributes. These LRPs are dominated by the amino acids Glu, Leu, and Ser and are rarer in the amino acids Cys, His, and Trp, and that many of the LRPs are rich in secondary or tertiary structures. Like mRNA-encoded peptides, these structure-rich cancer LRPs have a wide range of functions, including anti-cancer, cell-penetrating, anti-inflammatory, and antibacterial activities. Relatively, two groups of anticancer values (predicted by AntiCP 2.0 and PreTP-Stack) of these LRPs commonly showed positive and negative correlations with their total charge content and metal-bind aa content, respectively. The increasing amount of data and analysis on cancer LRPs, as reported here, offers opportunities to enhance practical cancer diagnosis and treatment, and to overcome remaining research challenges for cancer LRPs.
{"title":"Recent advances in structural and functional diversities of cancer lncRNA-encoded peptides: current opportunities and challenges for enhancing cancer diagnosis and treatment.","authors":"Aimin Guo, Shanshan Wang, Linan Huang, Yanping He, Qing Xi, David J Craik, Jun Zhang","doi":"10.1186/s12935-025-04158-2","DOIUrl":"https://doi.org/10.1186/s12935-025-04158-2","url":null,"abstract":"<p><p>Long non-coding RNAs (lncRNAs) are broad-spectrum cellular transcripts that can directly act as RNA regulators and/or partly encode functional peptides (lncRNA-encoded peptides, LRPs) in cancer cells. Recently, cancer LRPs have been found to be involved in cancer cell variability and proliferation, thus gaining widespread attention for their potential in cancer diagnosis, prognosis and therapy. As structures determine functions, the structural diversities of LRPs are the sources of functional variations of LRPs in cancers. Since 6135 cancer LRPs are listed in SPENCER database and 24 SPENCER-unlisted cancer LRPs are reported in several previous studies, this article reviews recent advances of cancer LRPs, analyzes amino acid compositions of them, and undertakes in silico evaluations to assess their structural and functional attributes. These LRPs are dominated by the amino acids Glu, Leu, and Ser and are rarer in the amino acids Cys, His, and Trp, and that many of the LRPs are rich in secondary or tertiary structures. Like mRNA-encoded peptides, these structure-rich cancer LRPs have a wide range of functions, including anti-cancer, cell-penetrating, anti-inflammatory, and antibacterial activities. Relatively, two groups of anticancer values (predicted by AntiCP 2.0 and PreTP-Stack) of these LRPs commonly showed positive and negative correlations with their total charge content and metal-bind aa content, respectively. The increasing amount of data and analysis on cancer LRPs, as reported here, offers opportunities to enhance practical cancer diagnosis and treatment, and to overcome remaining research challenges for cancer LRPs.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gliomas represent one of the most common types of primary brain tumor. Due to their poor prognosis and propensity for recurrence, new therapeutic targets are urgently required. A consensus is emerging that there is a significant relationship between tumor formation and embryonic development. However, the precise mechanisms and regulatory targets remain unclear. A variety of bioinformatics techniques, including GSVA, differential expression analysis, machine learning algorithms and others, were employed to elucidate the significance of germ layer development (GLD) in glioma and identify MEST as the key gene. To validate the results, in vivo and in vitro experiments were conducted, including tumor xenografts, RT-qPCR, immunocytofluorescence, transwell assays and others, which confirmed the central role of the selected oncogenic gene. Here, we performed a comprehensive bioinformatics analysis of GLD genes, providing a novel insight into the landscape of the GLD in gliomas, and confirmed the GLD-related gene MEST as a key oncogenic therapeutic target via machine learning feature selection framework. Furthermore, we have identified the core gene MEST and have conducted extensive research to elucidate its pivotal role in glioma progression through in vivo and in vitro experiments. We leveraged the GLD patterns in glioma and found that the MEST might promote the glioma development through activating RAS signaling and Wnt signaling.
{"title":"Leveraging the germ layer development patterns to predict prognosis and identify MEST as a novel therapeutic target in glioma.","authors":"Wei Zhang, Shunjin Xia, Yanming Xiao, Hongwei Liu, Chaoqian Wang, Luohuan Dai, Yinhua Chen, Xuelei Lin, Hongyi Liu, Nian Jiang","doi":"10.1186/s12935-025-04163-5","DOIUrl":"https://doi.org/10.1186/s12935-025-04163-5","url":null,"abstract":"<p><p>Gliomas represent one of the most common types of primary brain tumor. Due to their poor prognosis and propensity for recurrence, new therapeutic targets are urgently required. A consensus is emerging that there is a significant relationship between tumor formation and embryonic development. However, the precise mechanisms and regulatory targets remain unclear. A variety of bioinformatics techniques, including GSVA, differential expression analysis, machine learning algorithms and others, were employed to elucidate the significance of germ layer development (GLD) in glioma and identify MEST as the key gene. To validate the results, in vivo and in vitro experiments were conducted, including tumor xenografts, RT-qPCR, immunocytofluorescence, transwell assays and others, which confirmed the central role of the selected oncogenic gene. Here, we performed a comprehensive bioinformatics analysis of GLD genes, providing a novel insight into the landscape of the GLD in gliomas, and confirmed the GLD-related gene MEST as a key oncogenic therapeutic target via machine learning feature selection framework. Furthermore, we have identified the core gene MEST and have conducted extensive research to elucidate its pivotal role in glioma progression through in vivo and in vitro experiments. We leveraged the GLD patterns in glioma and found that the MEST might promote the glioma development through activating RAS signaling and Wnt signaling.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Oral cancer (OC) is the most common type of head and neck cancer, with a high mortality rate, and is a leading cause of cancer-related deaths worldwide. Drug-induced ferroptosis is a novel form of non-apoptotic cell death that offers a promising strategy for cancer therapy. Accumulating evidence has emphasized the significant role of methotrexate (MTX) in the treatment of many malignancies; however, its role in the ferroptosis pathway in OCs and its underlying mechanisms remain poorly understood.
Methods: After treating the OC cells with MTX, several cellular function assays were performed, including cell proliferation, apoptosis, colony formation, and wound healing assays. Distinctive features of ferroptosis were detected, and qPCR and western blot (WB) assays were performed to validate the expression of genes and proteins related to ferroptosis pathways in MTX-treated cells. In vitro experiments were conducted to further explore the mechanisms by which MTX regulates the stability of nuclear factor erythroid 2-related factor 2 (NRF2) in OC cells. Finally, in a mouse model using MOC1 cells, some experiments were performed to demonstrate MTX-induced ferroptosis and tumor suppression.
Results: In this study, based on in vitro and in vivo experiments, we found that MTX significantly reduced OC cell viability by inducing ferroptosis. Mechanistically, MTX administration increased the phosphorylation of Kelch-like ECH-associated protein 1 (KEAP1) at threonine 43 via activation of the ERK/MAPK signaling pathway, thereby maintaining the protein complex formed by KEAP1 and NRF2. As result of the decreased NRF2 expression, the levels of SLC7A11 and GPX4 proteins were markedly suppressed in MTX-treated OC cells, ultimately leading to the induction of ferroptosis in OC.
Conclusions: Our data demonstrated that MTX-mediated activation of the ERK/KEAP1 signaling pathway significantly induced ferroptosis by inhibiting the NRF2/HO-1/SLC7A11/GPX4 axis, thereby suppressing OC progression. These findings suggest that MTX is a promising candidate for OC treatment, offering a meaningful and effective therapeutic-strategy.
背景:口腔癌(OC)是头颈部最常见的癌症类型,死亡率高,是全球癌症相关死亡的主要原因。药物诱导的铁下垂是一种新的非凋亡细胞死亡形式,为癌症治疗提供了一种有前途的策略。越来越多的证据强调了甲氨蝶呤(MTX)在治疗许多恶性肿瘤中的重要作用;然而,其在OCs中铁下垂途径中的作用及其潜在机制仍然知之甚少。方法:用MTX处理OC细胞后,进行细胞功能测定,包括细胞增殖、凋亡、菌落形成和伤口愈合测定。检测到铁下垂的独特特征,并采用qPCR和western blot (WB)方法验证mtx处理细胞中铁下垂通路相关基因和蛋白的表达。体外实验进一步探讨MTX调控OC细胞核因子红细胞2相关因子2 (NRF2)稳定性的机制。最后,在使用MOC1细胞的小鼠模型中,进行了一些实验来证明mtx诱导的铁下垂和肿瘤抑制。结果:本研究通过体外和体内实验,我们发现MTX通过诱导铁下垂显著降低OC细胞活力。机制上,MTX通过激活ERK/MAPK信号通路,增加kelch样ECH-associated protein 1 (KEAP1)苏氨酸43位点的磷酸化,从而维持KEAP1和NRF2形成的蛋白复合物。由于NRF2表达降低,mtx处理的OC细胞中SLC7A11和GPX4蛋白水平被显著抑制,最终导致OC中铁下垂。结论:我们的数据表明,mtx介导的ERK/KEAP1信号通路的激活通过抑制NRF2/HO-1/SLC7A11/GPX4轴显著诱导铁凋亡,从而抑制OC的进展。这些发现表明甲氨蝶呤是一种有希望的卵巢癌治疗候选药物,提供了一种有意义和有效的治疗策略。
{"title":"Methotrexate-triggered ferroptosis suppresses oral cancer progression by phosphorylated KEAP1-mediated NRF2 degradation to inhibit SLC7A11/GPX4 signaling pathway.","authors":"Chenchen Yu, Tingting Zhang, Jialu Yuan, Yijing Su, Hongli Zhang, Liqin Xu, Xiaomin Li, Jianan Cui, Rui Xu, Yan Zhou, Hongming Huang, Xiaorong Zhou, Yongqiang Zhou, Erhao Zhang","doi":"10.1186/s12935-025-04019-y","DOIUrl":"https://doi.org/10.1186/s12935-025-04019-y","url":null,"abstract":"<p><strong>Background: </strong>Oral cancer (OC) is the most common type of head and neck cancer, with a high mortality rate, and is a leading cause of cancer-related deaths worldwide. Drug-induced ferroptosis is a novel form of non-apoptotic cell death that offers a promising strategy for cancer therapy. Accumulating evidence has emphasized the significant role of methotrexate (MTX) in the treatment of many malignancies; however, its role in the ferroptosis pathway in OCs and its underlying mechanisms remain poorly understood.</p><p><strong>Methods: </strong>After treating the OC cells with MTX, several cellular function assays were performed, including cell proliferation, apoptosis, colony formation, and wound healing assays. Distinctive features of ferroptosis were detected, and qPCR and western blot (WB) assays were performed to validate the expression of genes and proteins related to ferroptosis pathways in MTX-treated cells. In vitro experiments were conducted to further explore the mechanisms by which MTX regulates the stability of nuclear factor erythroid 2-related factor 2 (NRF2) in OC cells. Finally, in a mouse model using MOC1 cells, some experiments were performed to demonstrate MTX-induced ferroptosis and tumor suppression.</p><p><strong>Results: </strong>In this study, based on in vitro and in vivo experiments, we found that MTX significantly reduced OC cell viability by inducing ferroptosis. Mechanistically, MTX administration increased the phosphorylation of Kelch-like ECH-associated protein 1 (KEAP1) at threonine 43 via activation of the ERK/MAPK signaling pathway, thereby maintaining the protein complex formed by KEAP1 and NRF2. As result of the decreased NRF2 expression, the levels of SLC7A11 and GPX4 proteins were markedly suppressed in MTX-treated OC cells, ultimately leading to the induction of ferroptosis in OC.</p><p><strong>Conclusions: </strong>Our data demonstrated that MTX-mediated activation of the ERK/KEAP1 signaling pathway significantly induced ferroptosis by inhibiting the NRF2/HO-1/SLC7A11/GPX4 axis, thereby suppressing OC progression. These findings suggest that MTX is a promising candidate for OC treatment, offering a meaningful and effective therapeutic-strategy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145910579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}