1
|
Xu W, Li W, Kuai D, Li Y, Sun W, Liu X, Xu B. Identification of endoplasmic reticulum stress-related genes as prognostic markers in colon cancer. Cancer Biol Ther 2025; 26:2458820. [PMID: 40169935 PMCID: PMC11970746 DOI: 10.1080/15384047.2025.2458820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/20/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
Endoplasmic reticulum stress (ERS) has been implicated in the pathogenesis of various cancers, including colon cancer, by regulating tumor cell survival, growth, and immune response. However, the specific genes involved in ERS that could serve as prognostic markers in colon cancer remain underexplored. This study aims to identify and validate endoplasmic reticulum stress related genes (ERSRGs) in colon cancer that correlate with patient prognosis, thereby enhancing the understanding of ERS in oncological outcomes and potential therapeutic targeting. We utilized bioinformatics analyses to identify ERSRGs from publicly available colon cancer datasets. Differential expression analysis and survival analysis were performed to assess the prognostic significance of these genes. Validation was conducted through quantitative real-time PCR (RT-qPCR) on selected colon cancer cell lines. Our study identified nine ERS related genes (ASNS, ATF4, ATF6B, BOK, CLU, DDIT3, MANF, SLC39A14, TRAF2) involved in critical pathways including IL-12, PI3K-AKT, IL-7, and IL-23 signaling, and linked to 1-, 3-, and 5-year survival of patients with colon cancer. A multivariate Cox model based on these ERS related genes demonstrated significant prognostic power. Further, TRAF2 strong correlated with immune cells infiltration, suggesting its potential roles in modulating immune responses in the tumor microenvironment. The RT-qPCR validation confirmed the differential expression of these genes in human colon cancer cell lines versus human normal colonic epithelial cell line. The identified ERSRGs could serve as valuable prognostic markers and may offer new insights into the therapeutic targeting of ERS in colon cancer.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Dayu Kuai
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yaqiang Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xian Liu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Baohong Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Bassoy EY, Raja R, Rubino TE, Coscia F, Goergen K, Magtibay P, Butler K, Schmitt A, Oberg AL, Curtis M. Identification of TTLL8, POTEE, and PKMYT1 as immunogenic cancer-associated antigens and potential immunotherapy targets in ovarian cancer. Oncoimmunology 2025; 14:2460276. [PMID: 39891409 PMCID: PMC11792853 DOI: 10.1080/2162402x.2025.2460276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/27/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
Most high-grade serous ovarian cancers (OC) do not respond to current immunotherapies. To identify potential new actionable tumor antigens in OC, we performed immunopeptidomics on a human OC cell line expressing the HLA-A02:01 haplotype, which is commonly expressed across many racial and ethnic groups. From this dataset, we identified TTLL8, POTEE, and PKMYT1 peptides as candidate tumor antigens with low expression in normal tissues and upregulated expression in OC. Using tissue microarrays, we assessed the protein expression of TTLL8 and POTEE and their association with patient outcomes in a large cohort of OC patients. TTLL8 was found to be expressed in 56.7% of OC and was associated with a worse overall prognosis. POTEE was expressed in 97.2% of OC patients and had no significant association with survival. In patient TILs, increases in cytokine production and tetramer-positive populations identified antigen-specific CD8 T cell responses, which were dependent on antigen presentation by HLA class I. Antigen-specific T cells triggered cancer cell killing of antigen-pulsed OC cells. These findings suggest that TTLL8, POTEE, and PKMYT1 are potential targets for the development of antigen-targeted immunotherapy in OC.
Collapse
Affiliation(s)
| | - Remya Raja
- Department of Immunology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Fabian Coscia
- Max-Delbruck-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Krista Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Paul Magtibay
- Department of Obstetrics and Gynecology, Mayo Clinic, Phoenix, AZ, USA
| | - Kristina Butler
- Department of Obstetrics and Gynecology, Mayo Clinic, Phoenix, AZ, USA
- College of Medicine and Science, Mayo Clinic, Phoenix, AZ, USA
| | - Alessandra Schmitt
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Ann L. Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Marion Curtis
- Department of Immunology, Mayo Clinic, Phoenix, AZ, USA
- College of Medicine and Science, Mayo Clinic, Phoenix, AZ, USA
- Department of Cancer Biology, Mayo Clinic, Phoenix, AZ, USA
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
3
|
Huang P, Chen Y, Shi Y, Zhong C, Lin H, Yu X, Chen K, Huang Z, Zhang L, Fang S, Lu J, Chen J. Phosphoribosyl transferase domain containing 1: A prognostic biomarker in testicular germ cell tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200958. [PMID: 40241724 PMCID: PMC12001118 DOI: 10.1016/j.omton.2025.200958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
Due to the heterogeneity and complex classification of testicular germ cell tumors (TGCTs), prognostic evaluation and therapeutic targets remain unclear. Therefore, identifying a novel biomarker to comprehensively assess TGCT prognosis and immunotherapy response is crucial. We collected data from 457 TGCT patient samples and 12 normal testicular samples across six cohorts. Differential expression analysis combined with univariate Cox regression identified prognostic markers for TGCT. Multivariate Cox regression and survival analysis further evaluated the prognostic value of phosphoribosyl transferase domain containing 1 (PRTFDC1). Immunohistochemistry on tissue microarrays validated PRTFDC1's predictive value in clinical samples. We then investigated the relationship between PRTFDC1 and somatic mutations, copy number variations, immune cell infiltration, and immunotherapy response. Through these analyses, we identified PRTFDC1 as an independent risk factor indicating poor prognosis in TGCT. Immunohistochemistry demonstrated high PRTFDC1 expression in TGCT tissues. Gene set enrichment analysis revealed that PRTFDC1 suppresses immune-related pathways. Immune infiltration showed that high PRTFDC1 expression is associated with low CD8+ T cell infiltration. Immunotherapy response analysis indicated that low PRTFDC1 expression predicts better immunotherapy response and favorable prognosis. In conclusion, this study elucidates the biological and clinical significance of PRTFDC1, suggesting it as an effective and reliable biomarker for predicting TGCT prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Peisheng Huang
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Yihao Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Yongcheng Shi
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Huawei Lin
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Xiaoxue Yu
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Kai Chen
- Department of Pathology, Guangzhou Medical University Affiliated Women and Children′s Medical Center, Guangzhou 510623, China
| | - Zhuoya Huang
- Department of Pathology, Huizhou Central People’s Hospital, No. 41, Eling North Road, Huizhou, Guangdong 516001, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California, Riverside, CA 92507, USA
| | - Shumin Fang
- Science Research Center, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Jianming Lu
- Department of Andrology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Jiahong Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| |
Collapse
|
4
|
Tadepalli S, Clements DR, Raquer-McKay HM, Lüdtke A, Saravanan S, Seong D, Vitek L, Richards CM, Carette JE, Mack M, Gottfried-Blackmore A, Graves EE, Idoyaga J. CD301b+ monocyte-derived dendritic cells mediate resistance to radiotherapy. J Exp Med 2025; 222:e20231717. [PMID: 40146036 PMCID: PMC11949126 DOI: 10.1084/jem.20231717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/14/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Monocytes infiltrating tumors acquire various states that distinctly impact cancer treatment. Here, we show that resistance of tumors to radiotherapy (RT) is controlled by the accumulation of monocyte-derived dendritic cells (moDCs). These moDCs are characterized by the expression of CD301b and have a superior capacity to generate regulatory T cells (Tregs). Accordingly, moDC depletion limits Treg generation and improves the therapeutic outcome of RT. Mechanistically, we demonstrate that granulocyte-macrophage colony-stimulating factor (GM-CSF) derived from radioresistant tumor cells following RT is necessary for the accumulation of moDCs. Our results unravel the immunosuppressive function of moDCs and identify GM-CSF as an immunotherapeutic target during RT.
Collapse
Affiliation(s)
- Sirimuvva Tadepalli
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek R. Clements
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Hayley M. Raquer-McKay
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Anja Lüdtke
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjana Saravanan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - David Seong
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Lorraine Vitek
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher M. Richards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jan E. Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Andres Gottfried-Blackmore
- Department of Pharmacology, University of California San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, Division of Gastroenterology, University of California San Diego School of Medicine, San Diego, CA, USA
- Gastroenterology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Edward E. Graves
- Department of Radiation Oncology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pharmacology, University of California San Diego School of Medicine, San Diego, CA, USA
- Department of Molecular Biology, University of California San Diego School of Biological Sciences, San Diego, CA, USA
| |
Collapse
|
5
|
Ma J, Lang B, Wang L, Zhou Y, Fu C, Tian C, Xue L. Pan-Cancer Analysis and Experimental Validation of CEND1 as a Prognostic and Immune Infiltration-Associated Biomarker for Gliomas. Mol Biotechnol 2025; 67:2286-2304. [PMID: 38836983 DOI: 10.1007/s12033-024-01197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024]
Abstract
Cell cycle exit and neuronal differentiation 1 (CEND1), highly expressed in the brain, is a specific transmembrane protein which plays a tumor suppressor role. This study is performed to investigate the role of CEND1 in various cancers through pan-cancer analysis, and further investigate its functions in gliomas by cell experiments. The expression and subcellular localization of CEND1 in different cancer types were analyzed utilizing the data from the GEPIA, UCSC, UALCAN and HPA databases. Relationships of CEND1 expression with prognosis, immunomodulation-related genes, immune checkpoint genes, microsatellite instability (MSI), tumor mutation burden (TMB) and RNA modifications were analyzed based on the TCGA database. The ESTIMATE algorithm was utilized to evaluate tumors' StromalScore, Immune Score, and ESTIMATES Score. The cBioPortal database was employed to analyze the categories and frequencies of CEND1 gene alterations. Biological functions and co-expression patterns of CEND1 in gliomas were explored using the LinkedOmics database, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted. The interactions between CEND1 and drugs were explored employing the Comparative Toxicogenomics Database and molecular docking technology. Cell experiments were conducted to analyze triptonide's effects on glioma cells through CCK-8, flow cytometry and qRT-PCR. CEND1 was lowly expressed in gliomas, and high CEND1 expression was correlated to better overall survival of glioma patients (HR = 0.65, P = 0.02). Deep deletion was the main type of hereditary change of CEND1 mutation. CEND1 expression was markedly associated with immune infiltration, TMB, MSI, and RNA modification in various tumors (r > 0.3, P < 0.05). CEND1 co-expressed genes in gliomas were markedly correlated with immune responses and cell cycle (FDR < 0.05). Triptonide could bind well to CEND1 (-5.0 kcal/mol), and triptonide could facilitate CEND1 expression in glioma cells and cell apoptosis, and block the cell cycle progression (P < 0.05). CEND1 serves as a potential biomarker for pan-cancer. Particularly in gliomas, CEND1 is a key regulator of cell apoptosis and cell cycle, and a potential target for glioma treatment.
Collapse
Affiliation(s)
- Jinyang Ma
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| | - Bojuan Lang
- Department of Pathology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| | - Lei Wang
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China.
| | - Youdong Zhou
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| | - Changtao Fu
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| | - Chunlei Tian
- Department of Neurology, The First College of Clinical Medical Sciences, China Three Gorges University & Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| | - Lixin Xue
- Department of Neurosurgery, Zhijiang Branch of Yichang Central People's Hospital, Yichang, 443003, Hubei, People's Republic of China
| |
Collapse
|
6
|
Khalife M, Jia T, Caron P, Shreim A, Genoux A, Cristini A, Pucciarelli A, Leverve M, Lepeltier N, García-Rodríguez N, Dalonneau F, Ramachandran S, Fernandez Martinez L, Marcion G, Lemaitre N, Brambilla E, Garrido C, Hammond E, Huertas P, Gazzeri S, Sordet O, Eymin B. SRSF2 overexpression induces transcription-/replication-dependent DNA double-strand breaks and interferes with DNA repair pathways to promote lung tumor progression. NAR Cancer 2025; 7:zcaf011. [PMID: 40181846 PMCID: PMC11963763 DOI: 10.1093/narcan/zcaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 02/04/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
SRSF2 (serine/arginine-rich splicing factor 2) is a critical regulator of pre-messenger RNA splicing, which also plays noncanonical functions in transcription initiation and elongation. Although elevated levels of SRSF2 are associated with advanced stages of lung adenocarcinoma (LUAD), the mechanisms connecting SRSF2 to lung tumor progression remain unknown. We show that SRSF2 overexpression increases global transcription and replicative stress in LUAD cells, which correlates with the production of DNA damage, notably double-strand breaks (DSBs), likely resulting from conflicts between transcription and replication. Moreover, SRSF2 regulates DNA repair pathways by promoting homologous recombination and inhibiting nonhomologous end joining. Mechanistically, SRSF2 interacts with and enhances MRE11 (meiotic recombination 11) recruitment to chromatin, while downregulating 53BP1 messenger RNA and protein levels. Both events are likely contributing to SRSF2-mediated DNA repair process rerouting. Lastly, we show that SRSF2 and MRE11 expression is commonly elevated in LUAD and predicts poor outcome of patients. Altogether, our results identify a mechanism by which SRSF2 overexpression promotes lung cancer progression through a fine control of both DSB production and repair. Finally, we show that SRSF2 knockdown impairs late repair of ionizing radiation-induced DSBs, suggesting a more global function of SRSF2 in DSB repair by homologous recombination.
Collapse
Affiliation(s)
- Manal Khalife
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Tao Jia
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Pierre Caron
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Amani Shreim
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Aurelie Genoux
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Agnese Cristini
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Amelie Pucciarelli
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Marie Leverve
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Nina Lepeltier
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Néstor García-Rodríguez
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Fabien Dalonneau
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Shaliny Ramachandran
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Lara Fernandez Martinez
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Guillaume Marcion
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Nicolas Lemaitre
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Elisabeth Brambilla
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Carmen Garrido
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Ester M Hammond
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Pablo Huertas
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Sylvie Gazzeri
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Olivier Sordet
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Beatrice Eymin
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| |
Collapse
|
7
|
Castro-Oropeza R, Velazquez-Velazquez C, Vazquez-Santillan K, Mantilla-Morales A, Ruiz Tachiquin ME, Torres J, Rios-Sarabia N, Mayani H, Piña-Sanchez P. Landscape of lncRNAs expressed in Mexican patients with triple‑negative breast cancer. Mol Med Rep 2025; 31:163. [PMID: 40211710 PMCID: PMC12015155 DOI: 10.3892/mmr.2025.13528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/24/2025] [Indexed: 04/25/2025] Open
Abstract
Long non‑coding RNAs (lncRNAs) are key regulators of gene expression, that can regulate a range of carcinogenic processes. Moreover, they exhibit stability in biological fluids, with some displaying tissue specificity. As their expression depends on specific conditions or is linked to the regulation of particular signaling pathways, lncRNAs are promising candidates for providing insights into the likely progression of the disease. This allows for the stratification of patients based on their risk of progression, making them potential prognostic biomarkers in various types of cancer. In addition, the tissue‑specific expression profile of lncRNAs renders them ideal candidates for detection, prognosis and monitoring of cancer progression. The present study aims to provide an overview of differentially expressed lncRNAs in Mexican patients with triple‑negative breast cancer (TNBC), a subtype of breast cancer. The aim was to identify potential prognostic biomarkers that can be applied to improve the clinical management of Mexican patients with TNBC. Human Transcriptome Array 2.0 microarrays were used to analyze the transcriptome of TNBC and luminal tumors, which are reported to have a good prognosis amongst aggressive tumor types. Subsequently, results from these microarrays were validated in a cohort from The Cancer Genome Atlas, an independent cohort of Mexican patients and in breast cancer cell lines (MCF7, ZR75, T47D, MDA‑MB‑231, MDA‑MB‑468 and BT20). A total of 746 differentially expressed transcripts were identified, including 102 lncRNAs in TNBC compared with luminal tumors. Among the lncRNAs with the most significant changes in expression levels, SOX9‑AS was highly expressed in TNBC, whereas the expression of Lnc‑peroxidasin‑3:1 (Lnc‑PXDN‑3:1), Lnc‑RNA Synapse Defective Rho GTPase Homolog (Lnc‑SYDE) and long intergenic non‑coding RNA (LINC)01087 were decreased. In addition, the low expression of lncRNA LINC01087, LINC02568, ACO22196, and lncRNA eosinophil granule ontogeny transcript (Lnc‑EGOT) was associated with poor overall survival (OS). Further analysis revealed that the high expression levels of Lnc‑PXDN‑3:1, Lnc RNA fibrous sheath interacting protein 1‑6:3 and (LINC)00182 were associated with reduced survival in patients with the luminal subtype of breast cancer. Similarly, low expression levels of lncRNAs such as GATA binding protein 3‑1 (Lnc‑GATA‑3‑1), LINC01087, and BX679671.1 in luminal subtypes of breast cancer, as well as LINC00504 and LncRNA rho guanine nucleotide exchange factor 38 intronic transcript 1 (Lnc‑ARHGEF38‑IT1) in basal subtypes have been linked to poorer survival. The interactions and functions of LINC01087 were then investigated, revealing the interaction of LINC01087 with RNAs and transcription factors, highlighting their potential involvement in the estrogen receptor pathway. The present study provided a detailed analysis of the expression of lncRNAs in TNBC, which highlights the role of lncRNAs as a biomarker in the survival outcomes of patients with breast cancer to improve the understanding of transcriptional regulation in TNBC.
Collapse
Affiliation(s)
- Rosario Castro-Oropeza
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Cindy Velazquez-Velazquez
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Karla Vazquez-Santillan
- Laboratory of Innovation in Precision Medicine, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Alejandra Mantilla-Morales
- Department of Pathology, High Specialty Medical Unit Oncology Hospital, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Martha-Eugenia Ruiz Tachiquin
- Molecular Biology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Javier Torres
- Infectious and Parasitic Diseases Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Nora Rios-Sarabia
- Infectious and Parasitic Diseases Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Hector Mayani
- Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Patricia Piña-Sanchez
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| |
Collapse
|
8
|
Shi W, Dong J, Zhong B, Hu X, Zhao C. Predicting the Prognosis of Bladder Cancer Patients Through Integrated Multi-omics Exploration of Chemotherapy-Related Hypoxia Genes. Mol Biotechnol 2025; 67:2367-2381. [PMID: 38806990 DOI: 10.1007/s12033-024-01203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Bladder cancer is a prevalent malignancy with high mortality rates worldwide. Hypoxia is a critical factor in the development and progression of cancers. However, whether and how hypoxia-related genes (HRGs) could affect the development and the chemotherapy response of bladder cancer is still largely unexplored. This study comprehensively explored the complex molecular landscape associated with hypoxia in bladder cancer by analyzing 260 hypoxia genes based on transcriptomic and genomic data in 411 samples. Employing the 109 dysregulated hypoxia genes for consensus clustering, we delineated two distinct bladder cancer clusters characterized by disparate survival outcomes and distinct oncogenic roles. We defined a HPscore that was correlated with a variety of clinical features, including TNM stages and pathologic grades. Tumor immune landscape analysis identified three immune clusters and close interactions between hypoxia genes and the various immune cells. Utilizing a network-based method, we defined 129 HRGs exerting influence on apoptotic processes and critical signaling pathways in cancer. Further analysis of chemotherapy drug sensitivity identified potential drug-target HRGs. We developed a Risk Score model that was related to the overall survival of bladder cancer patients based on doxorubicin-target HRGs: ACTG2, MYC, PDGFRB, DHRS2, and KLRD1. This study not only enhanced our understanding of bladder cancer at the molecular level but also provided promising avenues for the development of targeted therapies, representing a significant step toward the identification of effective treatments and addressing the urgent need for advancements in bladder cancer management.
Collapse
Affiliation(s)
- Wensheng Shi
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, Hunan, China
- Furong Laboratory, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jiaming Dong
- Department of Radiation, Cangzhou Central Hospital, Hebei, 061000, China
| | - Bowen Zhong
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, Hunan, China
- Furong Laboratory, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiheng Hu
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, Hunan, China
- Furong Laboratory, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chunguang Zhao
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
9
|
Vastrad SJ, Saraswathy GR, Dasari JB, Nair G, Madarakhandi A, Augustine D, Sowmya S. A comprehensive transcriptome based meta-analysis to unveil the aggression nexus of oral squamous cell carcinoma. Biochem Biophys Rep 2025; 42:102001. [PMID: 40271514 PMCID: PMC12016861 DOI: 10.1016/j.bbrep.2025.102001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Lymph node metastasis in oral cancer (OC) complicates management due to its aggressive nature and high risk of recurrence, underscoring the need for biomarkers for early detection and targeted therapies. However, the drivers of this aggressive phenotype remain unclear due to the variability in gene expression patterns. To address this, an integrative meta-analysis of six publicly available transcriptomic profiles, categorized by lymph nodal status, is conducted. Key determinants of disease progression are identified through functional characterization and the TopConfects ranking approach of nodal associated differentially expressed genes (DEGs). To explore the critical nexus between lymph node metastasis and OC recurrence, significant metastatic genes were cross-analysed with literature-derived genes exhibiting aberrant methylation patterns in OC recurrence. Their clinical relevance and expression patterns were then validated in an external dataset from the TCGA head and neck cancer cohort. The analysis identified elevated expression of genes involved in extracellular matrix remodelling and immune response, while the expression of genes related to cellular differentiation and barrier functions was reduced, driving the transition to nodal positivity. The highest-ranked gene, MMP1, showed a log-fold change (LFC) of 4.946 (95 % CI: 3.71, 6.18) in nodal-negative samples, which increased to 5.899 (95 % CI: 4.80, 6.99) in nodal-positive samples, indicating consistent elevation across disease stages. In contrast, TMPRSS11B was significantly downregulated, with an LFC of -5.512 (95 % CI: -6.63, -4.38) in nodal-negative samples and -5.898 (95 % CI: -7.15, -4.64) in nodal-positive samples. Furthermore, MEIS1, down-regulated in nodal-positive status, was found to exhibit hypermethylation at CpG sites associated with OC recurrence. This study represents the first transcriptomic meta-analysis to explore the intersection of lymph node metastasis and OC recurrence, identifying MEIS1 as a potential key contributor. These comprehensive insights into disease trajectories offer potential biomarkers and therapeutic targets for future treatment strategies.
Collapse
Affiliation(s)
- Soujanya J. Vastrad
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| | - Ganesan Rajalekshmi Saraswathy
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| | | | - Gouri Nair
- Department of Pharmacology, Faculty of Pharmacy, M. S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Ashok Madarakhandi
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, (A Constituent Unit of KAHER-Belagavi), 2nd Block, Rajajinagar, Bangalore, India
| | - Dominic Augustine
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| | - S.V. Sowmya
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| |
Collapse
|
10
|
Toubia J, Kusay Y, Maqsood M, Warnock N, Lawrence D, Bracken C, Gregory P, Kan W, Selth L, Conn S, Lopez A, Branford S, Scott H, Kok CH, Goodall G, Schreiber A. TRanscriptome ANalysis of StratifiEd CohorTs (TRANSECT) enables automated assessment of global gene regulation linked to disparate expression in user defined genes and gene sets. NAR Genom Bioinform 2025; 7:lqaf041. [PMID: 40225790 PMCID: PMC11992672 DOI: 10.1093/nargab/lqaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Publicly accessible expression data produced by large consortium projects like TCGA and GTEx are increasing in number and size at an unprecedented rate. Their utility cannot be underestimated given the diversity of valuable tools widely used to interrogate these data and the many discoveries of biological and clinical significance already garnered from these datasets. However, there remain undiscovered ways to mine these rich resources and a continuing need to provide researchers with easily accessible and user-friendly applications for complex or bespoke analyses. We introduce TRanscriptome ANalysis of StratifiEd CohorTs (TRANSECT), a bioinformatics application automating the stratification and subsequent differential expression analysis of cohort data to provide further insights into gene regulation. TRANSECT works by defining two groups within a cohort based on disparate expression of a gene or a gene set and subsequently compares the groups for differences in global expression. Akin to reverse genetics minus the inherent requirement of in vitro or in vivo perturbations, cell lines or model organisms and all the while working within natural physiological limits of expression, TRANSECT compiles information about global transcriptomic change and functional outcomes. TRANSECT is freely available as a command line application or online at https://transect.au.
Collapse
Affiliation(s)
- John Toubia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Data and Bioinformatics Innovation, Department of Genetics and Molecular Pathology, SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Yasir Kusay
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Data and Bioinformatics Innovation, Department of Genetics and Molecular Pathology, SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Muneeza Maqsood
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Nicholas I Warnock
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Data and Bioinformatics Innovation, Department of Genetics and Molecular Pathology, SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - David M Lawrence
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Data and Bioinformatics Innovation, Department of Genetics and Molecular Pathology, SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Cameron P Bracken
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
| | - Winnie L Kan
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide 5000, Australia
| | - Luke A Selth
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide 5042, South Australia
- Flinders University, College of Medicine and Public Health, Freemasons Centre for Male Health and Wellbeing, Adelaide 5042, Australia
| | - Simon J Conn
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide 5042, South Australia
| | - Angel F Lopez
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Cytokine Receptor Laboratory, Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide 5000, Australia
| | - Susan Branford
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
| | - Hamish S Scott
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
| | - Chung Hoow Kok
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Data and Bioinformatics Innovation, Department of Genetics and Molecular Pathology, SA Pathology, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
| | - Andreas W Schreiber
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia
- ACRF Genomics Facility, Centre for Cancer Biology, An alliance between SA Pathology and the University of South Australia, Adelaide 5000, Australia
- School of Biological Sciences, University of Adelaide, Adelaide 5000, Australia
| |
Collapse
|
11
|
Jiang Y, Li Y, Zheng D, Du X, Yang H, Wang C, Zhao M, Xiao H, Zhang L, Li X, Shi S. Nano-polymeric platinum activates PAR2 gene editing to suppress tumor metastasis. Biomaterials 2025; 317:123090. [PMID: 39799696 DOI: 10.1016/j.biomaterials.2025.123090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/18/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
Metastasis as the hallmark of cancer preferentially contributes to tumor recurrence and therapy resistance, aggrandizing the lethality of patients with cancer. Despite their robust suppressions of tumor progression, chemotherapeutics failed to attenuate cancer cell migration and even triggered pro-metastatic effects. In parallel, protease-activated receptor 2 (PAR2), a member of the G protein-coupled receptor subfamily, actively participates in cancer metastasis via multiple signal transduction pathways. CRISPR/Cas9 that is a dominating genome editing tool can evoke PAR2 knockout to inhibit cancer metastasis. However, the absence of valid delivery systems largely limits its efficacy. Herein, we nanosized polymeric platinum (NanoPt) as therapeutical drug carries to deliver CRISPR/Cas9 to elicit genome editing of PAR2, which drastically augmented anti-metastatic effects and alleviated systematic toxicity of platinum-based treatment in vitro and in vivo. More importantly, the NanoPt@Cas9-PAR2 initiated PAR2 deficiency to mechanistically attenuate EMT process and ferroptosis via RAGE/ERK signalling, consequently preventing cancer cell migration. Our findings indicate that NanoPt@Cas9-PAR2 that mitigated PAR2 signalling and cytotoxic effects of platinum could be a safe and powerful all-in-one combinatorial strategy for cancer treatment.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dongmei Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xin Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Huan Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chuan Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Science and Education Division, Public Health Clinical Center of Chengdu& Public Health Clinical Center of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, China.
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Beijing Institute of Technology Chongqing Innovation Center, Chongqing, 401120, China.
| |
Collapse
|
12
|
Li J, Yang C, Zhang Y, Hong X, Jiang M, Zhu Z, Li J. Deciphering aging-associated prognosis and heterogeneity in gastric cancer through a machine learning-driven approach. iScience 2025; 28:112316. [PMID: 40256325 PMCID: PMC12008712 DOI: 10.1016/j.isci.2025.112316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with a high mortality rate and limited treatment options. Aging significantly contributes to tumor progression, and GC was confirmed as an aging-related heterogeneous disease. This study established an aging-associated index (AAI) using a machine learning-derived gene panel to stratify GC patients. High AAI scores associated with poor prognosis and indicated potential benefits from adjuvant chemotherapy, while showing resistance to immunotherapy. Single-cell transcriptome analysis revealed that AAI was enriched in monocyte cells within the tumor microenvironment. Two distinct molecular subtypes of GC were identified through unsupervised clustering, leading to the development of a subtype-specific regulatory network highlighting SOX7 and ELK3 as potential therapeutic targets. Drug sensitivity analyses indicated that patients with high ELK3 expression may respond to FDA-approved drugs (axitinib, dacarbazine, crizotinib, and vincristine). Finally, a user-friendly Shiny application was created to facilitate access to the prognostic model and molecular subtype classifier for GC.
Collapse
Affiliation(s)
- Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chuanlai Yang
- Department of Science and Technology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Yunxiao Zhang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mingye Jiang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhongxu Zhu
- Biomics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Guangdong, Shenzhen, China
| |
Collapse
|
13
|
Liu K, Hu S, Wufuer R, Zhang Q, Qiu L, Zhang Z, Wang M, Zhang Y. Deficiency of DDI2 suppresses liver cancer progression by worsening cell survival conditions. Free Radic Biol Med 2025; 232:200-213. [PMID: 40049338 DOI: 10.1016/j.freeradbiomed.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
The levels of reactive oxygen species (ROS) and the extent of ensuing DNA damage significantly influence cancer initiation and progression. Of crucial importance, the aspartate protease DDI2 has been proposed to play a pivotal role in monitoring intracellular ROS levels (to trigger oxidative eustress or distress), as well as in the oxidative DNA damage repair, through redox homeostasis-determining factor Nrf1 (encoded by NFE2L1). However, the specific role of DDI2 in the multi-step process resulting in the development and progression of liver cancer remains elusive to date. In the present study, we employed the CRISPR/Cas9 gene editing system to create two nuanced lines of DDI2 knockout (i.e., DDI2-/- and DDI2insG/-) from liver cancer cells. Subsequent experiments indicate that the knockout of DDI2 leads to increased ROS levels in hepatoma cells by downregulating two major antioxidant transcription factors Nrf1 and Nrf2 (encoded by NFE2L2), exacerbating endogenous DNA damages caused by ROS and not-yet-identified factors, thereby inhibiting cell proliferation and promoting apoptosis, and ultimately hindering in vivo malignant growth of xenograft tumor cells. Conversely, the restoration of DDI2 expression reverses the accumulation of ROS and associated DNA damage caused by DDI2 knockout, eliminating the subsequent inhibitory effects of DDI2 deficiency on both in vitro and in vivo growth of liver cancer cells. Collectively, these findings demonstrate that DDI2 deficiency impedes liver tumor growth by disrupting its survival environment, suggesting that DDI2 may serve as a novel therapeutic target for anti-cancer strategies aimed at modulating ROS or DNA damage processes.
Collapse
Affiliation(s)
- Keli Liu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Shaofan Hu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Jinfeng Laboratory, No. 313 Jinyue Road, Chongqing High-tech District, 401329, China; Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Reziyamu Wufuer
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; School of Parmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Xinjiang, 830017, China
| | - Qun Zhang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Lu Qiu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Meng Wang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Jinfeng Laboratory, No. 313 Jinyue Road, Chongqing High-tech District, 401329, China; Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba, Chongqing, 400038, China.
| | - Yiguo Zhang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; School of Life and Health Sciences, Fuyao University of Science and Technology, No. 104 Wisdom Avenue, Nanyu Town, Minhou County High-Tech District, Fuzhou, 350109, Fujian, China.
| |
Collapse
|
14
|
Cunanan CJ, Amirfallah A, Sanders AB, Gallant KC, Cavallo MR, Homer EA, El Naggar OS, Farnan JK, Romano G, Hope JL, Jackson JG, Hartsough EJ. BAP1 Loss Affords Lipotoxicity Resistance in Uveal Melanoma. Pigment Cell Melanoma Res 2025; 38:e70021. [PMID: 40300851 PMCID: PMC12040534 DOI: 10.1111/pcmr.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
Uveal melanoma (UM) is an aggressive intraocular malignancy. Despite effective control of primary tumors, ~50% of UM patients develop metastases, with the liver being the predominant secondary site. BAP1 deficiency, present in ~80% of metastatic UM cases, is strongly associated with increased metastatic risk and poor prognosis. In silico analysis of UM patient samples suggests that reduced BAP1 is linked to enhanced expression of genes involved in fatty acid processing; therefore, we hypothesize that BAP1 deficiency primes UM cells for survival in the hepatic microenvironment by enhancing lipid tolerance and oxidative stress responses. Our findings demonstrate BAP1-mutant UM resist lipotoxicity, whereas BAP1-competent UM exhibit sensitivity due to lipid peroxide accumulation-a hallmark of ferroptotic-like stress, and a response that can be mitigated by ferroptosis inhibition. Using an ex vivo liver slice model, we found that disrupting lipid metabolism with atorvastatin, an HMG-CoA reductase inhibitor, reduced tumor burden of BAP1-mutant UM. Moreover, we demonstrate a positive correlation between BAP1 and an epigenetic regulator of lipid homeostasis, ASXL2. Notably, ASXL2 depletion in BAP1-competent UM phenocopies the lipotoxicity resistance observed in BAP1-mutant UM-an effect that may be mediated by altered PPAR expression. This study reveals a novel mechanism linking BAP1 expression to lipid sensitivity via ASXL2, providing insights into liver tropism and potential therapeutic avenues for metastatic uveal melanoma.
Collapse
Affiliation(s)
- C. J. Cunanan
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - A. Amirfallah
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - A. B. Sanders
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - K. C. Gallant
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - M. R. Cavallo
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - E. A. Homer
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - O. S. El Naggar
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - J. K. Farnan
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - G. Romano
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
- Sidney Kimmel Comprehensive Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - J. L. Hope
- Sidney Kimmel Comprehensive Cancer CenterPhiladelphiaPennsylvaniaUSA
- Department of Microbiology & ImmunologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - J. G. Jackson
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
- Sidney Kimmel Comprehensive Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - E. J. Hartsough
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
- Sidney Kimmel Comprehensive Cancer CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
15
|
Xing L, Wu S, Xue S, Li X. A Novel Neutrophil Extracellular Trap Signature Predicts Patient Chemotherapy Resistance and Prognosis in Lung Adenocarcinoma. Mol Biotechnol 2025; 67:1939-1957. [PMID: 38734842 DOI: 10.1007/s12033-024-01170-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024]
Abstract
Chemoresistance is a key obstacle in the long-term survival of patients with locally and advanced lung adenocarcinoma (LUAD). This study used bioinformatic analysis to reveal the chemoresistance of gene-neutrophil extracellular traps (NETs) associated with LUAD. RNA sequencing data and LUAD expression patterns were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, respectively. The GeneCards database was used to identify NETosis-related genes (NRGs). To identify hub genes with significant and consistent expression, differential analysis was performed using the TCGA-LUAD and GEO datasets. LUAD subtypes were determined based on these hub genes, followed by prognostic analysis. Immunological scoring and infiltration analysis were conducted using NETosis scores (N-scores) derived from the TCGA-LUAD dataset. A clinical prognostic model was established and analyzed, and its clinical applications explored. Twenty-two hub genes were identified, and consensus clustering was used to identify two subgroups based on their expression levels. The Kaplan-Meier (KM) curves demonstrated statistically significant differences in prognosis between the two LUAD subtypes. Based on the median score, patients were further divided into high and low N-score groups, and KM curves showed that the N-scores were more precise at predicting the prognosis of patients with LUAD for overall survival (OS). Immunological infiltration analysis revealed significant differences in the abundances of 10 immune cell infiltrates between the high and low N-score groups. Risk scores indicated significant differences in prognosis between the two extreme score groups. The risk scores for the prognostic model also indicated significant differences between the two groups. The results provide new insights into NETosis-related differentially expressed genes (NRDEGs) associated with chemotherapy resistance in patients with LUAD. The established prognostic model is promising and could help with clinical applications to evaluate patient survival and therapeutic efficiency.
Collapse
Affiliation(s)
- Long Xing
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China
- Department of Oncology, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shuangli Wu
- Department of Special Examination, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shiyue Xue
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Xingya Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
16
|
Gao H, Zheng S, Liang J, Wang Y, Chen L, Li H, Chen Y, Zhang F, Shi H, Han A. m6A-induced DEAD-box RNA helicase 21 enhances lipid metabolism via 3‑hydroxy-3-methylglutaryl-CoA synthases 1 in colorectal cancer. Transl Oncol 2025; 55:102373. [PMID: 40127603 PMCID: PMC11979938 DOI: 10.1016/j.tranon.2025.102373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Altered lipid metabolism is a well-known hallmark of cancer. However, the underlying mechanisms of altered lipid metabolism in colorectal cancer (CRC) progression requires further investigation. Previously we have revealed that DEAD-box RNA helicase 21 (DDX21) promotes CRC metastasis via liquid-liquid phase separation. In this study, we identify DDX21 as a novel regulator of lipid metabolism in CRC. METHODS In vitro and in vivo assays illustrated the biological role of DDX21 and YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) in CRC lipid metabolism and progression. Bioinformatics analysis, ChIP, meRIP, RIP, RNA stability assay and dual-luciferase reporter assay were applied to explore the underlying molecular mechanisms. The expression levels and prognostic role of YTHDF1/DDX21/HMGCS1 axis in CRC patients were analyzed by immunohistochemical staining and Kaplan-Meier plotter. RESULTS DDX21 enhanced CRC progression via promoting lipid metabolism. Mechanistically, YTHDF1 enhanced DDX21 mRNA stability by recognizing its m6A-modified sites. DDX21 further binded to 3‑hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) promoter region and directly activated HMGCS1 transcription. Moreover, our clinical data showed that a simultaneously high expression of YTHDF1, DDX21 and HMGCS1 predicted an unfavorable overall survival in CRC patients. CONCLUSIONS Our study demonstrates that the YTHDF1/DDX21/HMGCS1 axis promotes CRC progression via regulating lipid metabolism and DDX21 might be a promising target for CRC therapy.
Collapse
Affiliation(s)
- Huabin Gao
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuai Zheng
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiangtao Liang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuting Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Lin Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hui Li
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yongyu Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Fenfen Zhang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Anjia Han
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
17
|
Bal Albayrak MG, Simsek T, Akpinar G, Kasap M, Canturk NZ. Proteomic insights into lymph node metastasis in breast cancer subtypes: Key biomarkers and pathways. Pathol Res Pract 2025; 269:155938. [PMID: 40179440 DOI: 10.1016/j.prp.2025.155938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/27/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Breast cancer (BC) is a significant global cause of death in women, primarily due to its diversity and metastatic potential. METHODS BC, healthy lymph node (HL), and metastatic lymph node (ML) tissues were collected from 19 patients diagnosed with infiltrating ductal carcinoma. Protein isolation was performed, followed by two-dimensional gel electrophoresis (2DE) and mass spectrometry (MALDI-TOF/TOF) to identify differentially expressed proteins. Bioinformatic analyses, including protein-protein interaction networks and molecular pathways, were conducted using STRING. Kaplan-Meier analysis was performed with KM plotter to evaluate the prognostic significance of identified proteins. Receiver operating characteristic (ROC) curves were generated using TCGA and GTEx data from UCSC Xena and easyROC to assess diagnostic relevance. RESULTS Distinct pathways related to cytoskeletal regulation, immune modulation, and oxidative stress response were enriched in each subtype. Key proteins such as TUBA1C, CCT6A, and Vimentin (LNA), CAPZB and ENO1 (LNB), GSTO1 (HER2 OE), and CORO1A and LAP3 (TNBC) were identified as significant in driving metastatic behavior. KM survival analysis showed that CAPZB (LNB) and CORO1A (TNBC) were associated with patient outcomes, while GSTO1 was linked to improved distant metastasis-free survival in HER2 OE. ROC analysis highlighted CAPZB as a strong diagnostic marker. CONCLUSIONS These findings form a basis for comprehending the molecular mechanisms underlying metastasis in different subtypes of breast cancer. They may lead to the identification of new therapeutic targets for customized interventions against invasion and metastasis. Further validation is required to confirm their clinical utility in larger cohorts.
Collapse
Affiliation(s)
| | - Turgay Simsek
- Department of General Surgery, Medical School, Kocaeli University, Kocaeli 41001, Turkiye
| | - Gurler Akpinar
- Department of Medical Biology, Medical School, Kocaeli University, Kocaeli 41001, Turkiye.
| | - Murat Kasap
- Department of Medical Biology, Medical School, Kocaeli University, Kocaeli 41001, Turkiye
| | - Nuh Zafer Canturk
- Department of General Surgery, Medical School, Kocaeli University, Kocaeli 41001, Turkiye
| |
Collapse
|
18
|
Padam KSR, Basavarajappa DS, Kumar NAN, Gadicherla S, Chakrabarty S, Hunter KD, Radhakrishnan R. Epigenetic regulation of HOXA3 and its impact on oral squamous cell carcinoma progression. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 139:550-563. [PMID: 39658479 DOI: 10.1016/j.oooo.2024.11.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVE The role of homeobox A3 (HOXA3) in cancer progression is gaining prominence, however, to date, no studies have investigated its regulatory function in oral cancer. In this study, we explored the role of HOXA3 through epigenetic mechanisms. METHODS Clinical samples were collected from 25 potentially malignant oral lesions and 50 oral squamous cell carcinoma (OSCC) patients, categorized into low-stage and high-stage tumors. The promoter activity of HOXA3 was determined through cloning and luciferase assays. CpG methylation patterns across the gene were identified using methyl-capture sequencing. Gene expression was analyzed using RT‒qPCR. The Survminer R package was used to assess the clinical significance of 3' UTR methylation associated with overall survival. RNA‒RNA interactions were analyzed using RNAInter and TargetScan v8.0. RESULTS HOXA3 expression was upregulated in dysplasia and downregulated in advanced cancer stages, showing an inverse correlation with promoter methylation, suggesting epigenetic regulation by DNA methylation. Hypermethylation of the 3' UTR was associated with poor overall survival in advanced stages. Long noncoding RNAs and microRNAs may post-transcriptionally modulate HOXA3 in oral carcinogenesis. CONCLUSION CpG-specific hypermethylation in the 3' UTR may serve as a potential biomarker in OSCC.
Collapse
Affiliation(s)
- Kanaka Sai Ram Padam
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Dhanraj Salur Basavarajappa
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Naveena A N Kumar
- Department of Surgical Oncology, Kasturba Medical College and Hospital, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Srikanth Gadicherla
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Public Health & Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Keith D Hunter
- Liverpool Head and Neck Centre, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India; Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, ST10 2TN, UK; Academic Unit of Oral Biology and Oral Pathology, Oman Dental College, P.O Box 835, Mina Al Fahal, Muscat, Wattayah 116, Oman.
| |
Collapse
|
19
|
Nakano T, Okita K, Okazaki S, Yoshimoto S, Masuko S, Yagi H, Kato K, Tomioka Y, Imai K, Hamada Y, Masuko K, Shimada-Takaura K, Nagai N, Saya H, Arai T, Ishiwata T, Masuko T. CD44v, S1PR1, HER3, MET and cancer-associated amino acid transporters are promising targets for the pancreatic cancers characterized using mAb. FEBS Open Bio 2025; 15:867-884. [PMID: 39757718 DOI: 10.1002/2211-5463.13963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/09/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Effective therapies have yet to be established for pancreatic ductal adenocarcinomas (PDAC) even though it is the most aggressive cancer. In the present study, PDAC was analyzed using novel rat mAbs against membrane proteins in conjunction with flow cytometry and immunohistochemistry. Human epidermal growth receptor (HER)1-4, mesenchymal to epithelial transition factor (MET), sphingosine-1-phospahate receptor 1 (S1PR1), l-type amino acid transporter 1 (LAT1), system x- c transporter (xCT), alanine-serine-cysteine transporter (ASCT2), cationic amino acid transporter 1 (CAT1) and variant CD44 (CD44v) were expressed at high frequencies in both in vitro and in vivo PDAC. Internalization of membrane proteins by mAbs and growth inhibition by toxin-linked mAbs were demonstrated in many PDAC cell lines, and mAbs against S1PR1, ASCT2, HER3 and CD44v inhibited the growth of xenografted MIA PaCa-2 PDAC cells. Furthermore, CD44v-high PDAC showed high mRNA expression of HER1-3, MET and CD44v, and was correlated with poor prognosis. Taken together, our results suggest that CD44v, S1PR1, HER3, MET and the above-mentioned cancer-associated amino acid transporters might be promising targets for the diagnosis and treatment of PDAC.
Collapse
Affiliation(s)
- Takashi Nakano
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- Faculty of Health and Sports Sciences, Toyo University, Kita-ku, Japan
| | - Kouki Okita
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- Advanced Design for Pharmaceuticals, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
| | - Shogo Okazaki
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- Department of Microbiology, Division of Immunology and Pathobiology, School of Dentistry, Nihon University, Chiyoda-ku, Japan
| | - Soshi Yoshimoto
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- PIERAS Co., Ltd, Osaka-shi, Japan
| | - Sachiko Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- PIERAS Co., Ltd, Osaka-shi, Japan
| | - Hideki Yagi
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- Department of Pharmaceuticals, Faculty of Pharmacy, International University of Health and Welfare, Otawara-shi, Japan
| | - Kazunori Kato
- Faculty of Health and Sports Sciences, Toyo University, Kita-ku, Japan
| | - Yoshihisa Tomioka
- Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-shi, Japan
| | - Kenichi Imai
- Department of Microbiology, Division of Immunology and Pathobiology, School of Dentistry, Nihon University, Chiyoda-ku, Japan
| | | | - Kazue Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
| | | | - Noriaki Nagai
- Advanced Design for Pharmaceuticals, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
| | - Hideyuki Saya
- Oncology Innovation Center, Fujita Health University, Toyoake-shi, Japan
| | - Tomio Arai
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
| | - Toshiyuki Ishiwata
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
| | - Takashi Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- Advanced Design for Pharmaceuticals, School of Pharmacy, Kindai University, Higashiosaka-shi, Japan
- Oncology Innovation Center, Fujita Health University, Toyoake-shi, Japan
| |
Collapse
|
20
|
Zhang X, Di Y, Wang Y, Qin J, Ye L, Wen X, Ke Z, Wang Z, He W. SIRT5-mediated desuccinylation of PPA2 enhances HIF-1alpha-dependent adaptation to hypoxic stress and colorectal cancer metastasis. EMBO J 2025; 44:2514-2540. [PMID: 40164945 PMCID: PMC12048626 DOI: 10.1038/s44318-025-00416-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Metastasis is the primary cause of death in patients with colorectal cancer (CRC). Hypoxia is a hallmark of solid tumors that promotes cellular metabolic adaptation and dissemination. However, the mechanisms linking hypoxia-regulated metabolic adaptation to CRC metastasis remain unclear. Here, we found that inorganic pyrophosphatase 2 (PPA2) suppresses metastatic progression of CRC via its phosphatase function. PPA2 expression levels are reduced in CRC specimen and correlate with enhanced response to hypoxia by promoting hypoxia-inducible factor-1 (HIF-1) signaling to promote CRC cell glycolysis and dissemination. Mechanistically, PPA2 decreases HIF-1alpha stability through non-canonical ubiquitin-mediated proteasomal degradation via recruitment of E3 ligase NEDD4. Furthermore, PPA2 directly dephosphorylates NEDD4 at threonine 758 residue, resulting in its activation. Under hypoxic stress, NAD-dependent protein deacetylase sirtuin-5 promotes the dissociation of PPA2 and NEDD4 by inducing PPA2 desuccinylation at lysine 176, contributing to the improved stability of HIF-1alpha under hypoxic conditions. Our findings reveal a tumor-suppressive role of PPA2 in HIF-1alpha-dependent colorectal cancer, providing a potential therapeutic target and prognostic strategy.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuqin Di
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Youpeng Wang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiale Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lvlan Ye
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiangqiong Wen
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Medical College of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zunfu Ke
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Ziyang Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China.
| |
Collapse
|
21
|
Varisli L, Zoumpourlis P, Spandidos DA, Zoumpourlis V, Vlahopoulos S. ALDH1A1 in breast cancer: A prospective target to overcome therapy resistance (Review). Oncol Lett 2025; 29:213. [PMID: 40093866 PMCID: PMC11905208 DOI: 10.3892/ol.2025.14959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
The expression of cytosolic aldehyde dehydrogenases (ALDHs), which mediate the last step in the pathway of the synthesis of all-trans retinoic acid, is dysregulated in various types of human cancer, and has been associated with the development of cancer stem cells (CSCs) in solid tumors and hematological malignancies. CSCs are considered a minor fraction of cancer cells with the capacity to initiate neoplastic tumors. ALDH1A1 serves a crucial role in the emergence of the CSC phenotype, induces the malignant behavior of cancer cells and promotes treatment resistance. Notably, ALDH1A1-induced therapy resistance is not exclusive to just one group of drugs, but affects diverse types of drugs that use different mechanisms to kill cells. This diversity of drug resistance-inducing effects is associated with the stemness-supporting functions of ALDH1A1. The inhibition of ALDH1A1 activity using chemicals or the depletion of ALDH1A1 via genetic approaches, such as the use of small interfering RNA, can overcome diverse pathways of therapy resistance. In the context of breast cancer, it is critical that only a fraction of malignant cells are expected to manifest stem-like features, which include increased expression of ALDH1A1. From the angle of disease prognosis, the extent of the association of ALDH1A1 with increased malignant behavior and drug resistance remains to be determined through the application of cutting-edge methods that detect the expression of tracked biomarkers within tumors.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
22
|
Yang L, Ren S, Lou L, He J, Huang Q, Wu X, Zhao R. A bioinformatics analysis and experimental validation of PDGFD as a promising diagnostic biomarker for acute myeloid leukemia. Sci Rep 2025; 15:14862. [PMID: 40295666 PMCID: PMC12037738 DOI: 10.1038/s41598-025-99038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Acute myeloid leukemia (AML) is a malignant blood cancer resulting from leukemia stem cells (LSCs) supplanting normal stem cells. Platelet-derived growth factors (PDGFs) are important for LSCs but have not been studied in the development of AML. In this study, transcriptome data of PDGFs were sourced from The Cancer Genome Atlas (TCGA) and GTEx databases, and relevant differential expression and prognosis analysis were performed using R software packages and online tools (UCSC-Xena Shiny tools, GEPIA2, Kaplan-Meier Plotter databases, etc.). Then, we focused on PDGFD expression in AML, along with its clinical and diagnostic importance, drug resistance studies, and association with immunotherapy. The real-time quantitative polymerase chain reaction (RT-qPCR) was performed to verify the expression and clinical characteristics of PDGFD. Analyses of public data and clinical samples revealed that PDGFD expression was upregulated compared with other PDGF genes, and only this upregulation was associated with poor prognosis in AML. High expression of PDGFD showed a significant positive correlation with intermediate-high cytogenetic risk, NPM1 mutation, FLT3-ITD mutation, and unfavorable prognosis. ROC curve analysis indicated that PDGFD holds substantial diagnostic potential for AML patients. Functional enrichment analysis revealed the role of PDGFD in calcium and Rap1 signaling pathways. Additionally, PDGFD expression exhibited a significant positive correlation with natural killer cells and dendritic cells. Furthermore, we propose that MiR-203-3p targeting PDGFD has potential anti-leukemic effects in AML. In conclusion, PDGFD serves as a possible diagnostic and prognostic biomarker, as well as a target for cellular immunotherapy in AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Computational Biology/methods
- Nucleophosmin
- Prognosis
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
- Lymphokines/genetics
- Lymphokines/metabolism
- Male
- Female
- Gene Expression Regulation, Leukemic
- Middle Aged
- Gene Expression Profiling
- Mutation
Collapse
Affiliation(s)
- Lulu Yang
- Department of Hematology, Ninghai First Hospital, Ningbo, 315600, Zhejiang, China
| | - Shuangshuang Ren
- Department of Ultrasound, Dongyang People's Hospital, Jinhua, 322100, Zhejiang, China
| | - Lijiang Lou
- Department of Hematology, Ninghai First Hospital, Ningbo, 315600, Zhejiang, China
| | - Jiasu He
- Department of Anesthesiology, Ninghai First Hospital, Ningbo, 315600, Zhejiang, China
| | - Qianlei Huang
- Department of Hematology, The First Affiliated Hospital of Hainan Medical College, Haikou, 570102, Hainan, China
| | - Xiaojin Wu
- Department of Hematology, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Ranran Zhao
- Department of Hematology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.
| |
Collapse
|
23
|
Ju G, Lin Q, Lu L, Lin Z, Huang D, Lin Y, Huang X, Lin Q, Xu H, Bai Y, Miao S, Li J, Song J, Chen J. TFDP1 drives triple-negative breast Cancer development through senescence suppression and serves as a therapeutic target for topotecan. Int J Biol Macromol 2025; 310:143543. [PMID: 40300683 DOI: 10.1016/j.ijbiomac.2025.143543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Triple-negative breast cancer (TNBC) constitutes the molecular subtype exhibiting the poorest prognosis. Targeted therapy emerges as a pivotal strategy to enhance the clinical outcomes of individuals with TNBC. Identifying targets and corresponding therapeutic agents is essential for reducing TNBC-related mortality. Topotecan, a chemotherapeutic agent approved for treating metastatic breast cancer, remains under investigation regarding its specific targets and molecular mechanisms in TNBC. Data procured from CRISPR/Cas9 library screenings showed that TFDP1 may be a therapeutic target in TNBC, and the L1000FWD database suggested that TFDP1 serves as a potential target of topotecan. The overexpression of TFDP1 was observed in TNBC tissues, correlating with poorer prognosis. Knockdown of TFDP1 inhibited the cell growth, clonal expansion, and tumorigenicity of TNBC cells. Mechanistically, TFDP1 inhibited cellular senescence in TNBC cells. In vitro experiments demonstrated that topotecan inhibited TNBC cell growth and promoted cellular senescence, counteracting the effects of TFDP1 overexpression on TNBC cells. These findings suggest that topotecan impedes TNBC cell growth by targeting TFDP1. This interaction provides valuable insights into the molecular mechanisms governing TNBC cell senescence, presenting TFDP1 as a potential therapeutic target. Combining topotecan with senolytic therapies may offer a promising strategy for TNBC treatment.
Collapse
Affiliation(s)
- Gaoda Ju
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China.
| | - Qingliang Lin
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Lihu Lu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Zhuangbin Lin
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Daxin Huang
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Yaobin Lin
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Xiaoxue Huang
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Qiong Lin
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Huan Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Yue Bai
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Sen Miao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Junjian Li
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianyuan Song
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China.
| | - Jianwu Chen
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China; Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China.
| |
Collapse
|
24
|
Ou G, Gao T, Hu S, Zhang S, Song S, Sun Y, Wang Y, Zhong R, Hu K, Gao L, Jiang M. Comprehensive pan-cancer analysis of CHRDL1 and experimental validation of its role in lung adenocarcinoma. BMC Cancer 2025; 25:783. [PMID: 40287623 PMCID: PMC12034113 DOI: 10.1186/s12885-025-14174-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Chordin-like 1 (CHRDL1) is a secreted antagonist of bone morphogenetic proteins, and has been implicated in various biological processes and cancer prognosis. This study offered a detailed examination of CHRDL1 expression across 33 diverse cancer types, leveraging data from The Cancer Genome Atlas (TCGA) and supplementary public datasets. We demonstrated that, for the majority of cancer types, CHRDL1 expression was reduced in tumor tissues compared to normal adjacent tissues. Notably, lower CHRDL1 expression led to negative prognosis in malignancies such as lung adenocarcinoma (LUAD), melanoma (SKCM), and mesothelioma (MESO). Furthermore, CHRDL1 expression was positively correlated with the infiltration of CD4⁺ T cells, CD8⁺ T cells, B cells, neutrophils, macrophages, and dendritic cells in most tumors. Higher CHRDL1 expression correlated with more favorable immune profiles and a reduction in tumor stemness. To assess the effect of CHRDL1 overexpression on LUAD progression, we conducted CCK-8, wound healing, and invasion assays in vitro, along with subcutaneous tumor formation experiments in nude mice. The results showed that the proliferation, migration, and invasion abilities of A549 and H1299 cells with high CHRDL1 expression were reduced, and the growth of A549 cells was also significantly inhibited in nude mice. These findings underscored CHRDL1's potential as a prognostic biomarker and its influence on tumor immunology and cellular dynamics.
Collapse
Affiliation(s)
- Guangyin Ou
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangke Gao
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaopu Hu
- The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shuixiu Zhang
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shuo Song
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Sun
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wang
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruikang Zhong
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaiwen Hu
- Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Gao
- Beijing University of Chinese Medicine, Beijing, China.
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Min Jiang
- Beijing University of Chinese Medicine, Beijing, China.
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
25
|
Wang Y, Wang Y, Bao L, Vale G, McDonald JG, Fang Y, Peng Y, Kumar A, Xing C, Brasó-Maristany F, Prat A, Arteaga CL, Wang Y, Luo W. ZMYND8 drives HER2 antibody resistance in breast cancer via lipid control of IL-27. Nat Commun 2025; 16:3908. [PMID: 40281007 PMCID: PMC12032076 DOI: 10.1038/s41467-025-59184-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Anti-HER2 antibodies are effective but often lead to resistance in patients with HER2+ breast cancer. Here, we report an epigenetic crosstalk with aberrant glycerophospholipid metabolism and inflammation as a key resistance mechanism of anti-HER2 therapies in HER2+ breast cancer. Histone reader ZMYND8 specifically confers resistance to cancer cells against trastuzumab and/or pertuzumab. Mechanistically, ZMYND8 enhances cPLA2α expression in resistant tumor cells through inducing c-Myc. cPLA2α inactivates phosphatidylcholine-specific phospholipase C to inhibit phosphatidylcholine breakdown into diacylglycerol, which diminishes protein kinase C activity leading to interleukin-27 secretion. Supplementation with interleukin-27 protein counteracts cPLA2α loss to reinforce trastuzumab resistance in HER2+ tumor cells and patient-derived organoids. Upregulation of ZMYND8, c-Myc, cPLA2α, and IL-27 is prevalent in HER2+ breast cancer patients following HER2-targeted therapies. Targeting c-Myc or cPLA2α effectively overcomes anti-HER2 therapy resistance in patient-derived xenografts. Collectively, this study uncovers a druggable signaling cascade that drives resistance to HER2-targeted therapies in HER2+ breast cancer.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yanan Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lei Bao
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Goncalo Vale
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yisheng Fang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yan Peng
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Cancer Institute and Blood Diseases, Hospital Clínic of Barcelona, Barcelona, Spain
- Reveal Genomics, S.L., Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Cancer Institute and Blood Diseases, Hospital Clínic of Barcelona, Barcelona, Spain
- Reveal Genomics, S.L., Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Institute of Oncology-Hospital Quirónsalud, Barcelona, Spain
| | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
26
|
Thankachan S, Bhardwaj BK, Patel D, Kp K, Kabekkodu SP, Suresh PS. Clinicopathological correlation of PTPN3 expression in breast cancer and in silico drug screening against PTPN3 for therapeutics. Cancer Genet 2025; 294-295:111-122. [PMID: 40315635 DOI: 10.1016/j.cancergen.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 05/04/2025]
Abstract
PTPN3 regulates cellular signaling and is dysregulated in cancer. There has been less research about the oncogenic impact of PTPN3 in breast cancer patients. This study analyzed PTPN3 mRNA levels and their prognostic significance in breast cancer using TCGA datasets. qRT-PCR was used to assess PTPN3 expression in formalin-fixed, paraffin-embedded Indian breast cancer patient samples (tumor-74, control-36). PTPN3 protein levels (ER-positive 15; ER-negative: 15; distant normal breast tissues: 20) were also immunohistochemically assessed using the H-score method. The biomarker potential was examined using a receiver operating characteristic (ROC) analysis. Docking and molecular dynamics (MD) simulations were used to find PTPN3 inhibitors (PDB ID: 2B49) from 892 FDA-approved natural chemicals in the ZINC database. PTPN3 mRNA and protein expression were significantly higher in breast cancers and associated with clinicopathological variables such as age, ER status, tumor stage, grade, Ki-67 index, menopause, and lymph node metastasis (p < 0.05). ROC analysis revealed an AUC of 0.7654, indicating PTPN3's biomarker potential. Docking yielded three high-affinity inhibitors: Cyclocort (ZINC000003977777), Toposar (ZINC000003938684), and Tetracycline (ZINC000084441937), with binding energies of -9.3, -8.73, and -8.66 kcal/mol, respectively. MD simulations confirmed stable connections via hydrogen bonds and hydrophobic interactions under minimal constraints. In conclusion, PTPN3 overexpression supports its role as a prognostic biomarker, and Cyclocort, Toposar, and Tetracycline need further confirmation as potential PTPN3 inhibitors.
Collapse
Affiliation(s)
- Sanu Thankachan
- Department of Bioscience and Engineering, National Institute of Technology, Calicut 673601, Kerala, India
| | - Boddapati Kalyani Bhardwaj
- Department of Bioscience and Engineering, National Institute of Technology, Calicut 673601, Kerala, India
| | - Dimple Patel
- Department of Bioscience and Engineering, National Institute of Technology, Calicut 673601, Kerala, India
| | - Kavitha Kp
- Department of Pathology, Aster Malabar Institute of Medical Sciences (MIMS), Calicut 673016, Kerala, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Padmanaban S Suresh
- Department of Bioscience and Engineering, National Institute of Technology, Calicut 673601, Kerala, India.
| |
Collapse
|
27
|
Zheng G, Wei P, Li D, Li X, Zafereo M, Li C, Yu W, Chen X, Zheng H, Song X, Li G. A Deep Learning Survival Model for Evaluating the Survival Prognosis of Papillary Thyroid Cancer: A Population-Based Cohort Study. Ann Surg Oncol 2025:10.1245/s10434-025-17290-0. [PMID: 40254654 DOI: 10.1245/s10434-025-17290-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/24/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Deep learning can assess the individual survival prognosis in sizeable datasets with intricate underlying processes. However, studies exploring the performance of deep learning survival in papillary thyroid cancer (PTC) are lacking. This study aimed to construct a deep learning model based on clinical risk factors for survival prediction in patients with PTC. METHODS A Cox proportional hazards deep neural network (DeepSurv) was developed and validated by using consecutive patients with PTC from 17 US Surveillance, Epidemiology, and End Results Program (SEER) cancer registries (2000-2020). The performance of the DeepSurv model was further validated on two external test datasets from the University of Texas MD Anderson Cancer Center (MDACC) and The Cancer Genome Atlas (TCGA). Using the survival risk scores at 10 years predicted by the DeepSurv model, we classified patients with PTC into low-risk and high-risk groups and explored their overall survival (OS). RESULTS The concordance index of the DeepSurv model for predicting OS was 0.798 in the SEER test dataset, 0.893 in the MDACC dataset, and 0.848 in the TCGA dataset. The DeepSurv model was capable of classifying patients with PTC into low-risk and high-risk groups according to the survival risk scores at 10 years. Patients in the high-risk group had significantly worse OS than patients in the low-risk group in all three test datasets (all P < 0.001). CONCLUSION The DeepSurv model was capable of classifying patients with PTC into low-risk and high-risk groups, which may provide important prognostic information for personalized treatment in patients with PTC.
Collapse
Affiliation(s)
- Guibin Zheng
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danxia Li
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xinna Li
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Mark Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chao Li
- Department of Head and Neck Surgery, Schoolof Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of ElectronicScience and Technology of China, Chengdu, 610041, China
| | - Wenbin Yu
- Department of Hand and Neck Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaohong Chen
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Haitao Zheng
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, the Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China.
| | - Guojun Li
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
28
|
Feng B, Guo HY, Ning Y, Zhao YY, Wang X, Cui R. LPCAT3 regulates the immune infiltration and prognosis of ccRCC patients by mediating ferroptosis and endoplasmic reticulum stress. Discov Oncol 2025; 16:574. [PMID: 40253575 PMCID: PMC12009263 DOI: 10.1007/s12672-025-02283-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) accounts for 70% of renal cell carcinoma (RCC) cases. Although surgery remains the mainstay treatment, renal injury and high metastasis rates after nephrectomy dramatically reduce patient quality of life. Drugs that stimulate the immune system by targeting checkpoint pathways improve overall survival in patients with RCC. Here, we investigated the applicability of lysophosphatidylcholine acyltransferase 3 (LPCAT3) as a target for immunotherapy. METHODS In the present study, high LPCAT3 expression in ccRCC was identified using The Cancer Genome Atlas (TCGA) data and validated in two external cohorts from the Gene Expression Omnibus (GEO) database. qRT-PCR was performed to identify the mRNA level of LPCAT3 in tumors and adjacent normal tissues. And immunohistochemistry was used to evaluate the protein level of LPCAT3 between two groups of samples. Furthermore, gene set enrichment analysis was performed to explore the biological processes and pathways related to LPCAT3 expression. Key gene expression and correlation analyses were performed to determine the crosstalk among LPCAT3 expression, ferroptosis, and endoplasmic reticulum stress (ERS). Subsequently, CIBERSORT was used to analyze the immune infiltration status of patients with high and low LPCAT3 expression. RESULTS TCGA and GEO data revealed that LPCAT3 expression in ccRCC tumor tissues was higher than that in adjacent normal tissues; moreover, patients with high LPCAT3 expression had better survival outcomes. qRT-PCR and immunohistochemistry verified the high LPCAT3 expression in tumor tissue. Pathways related to ferroptosis and ERS were upregulated in patients with high LPCAT3 expression. Univariate and multivariate regression analyses revealed that low LPCAT3 levels represent an independent risk factor for ccRCC. LPCAT3 expression was positively correlated with M2 macrophage infiltration levels but negatively correlated with the memory B cell, CD8+ T cell, follicular helper T cell, regulatory T cell, activated natural killer cell, and activated memory CD4+ T cell infiltration levels. CONCLUSIONS LPCAT3was identified as a ccRCC biomarker and may regulate immune infiltration and prognosis in ccRCC by mediating ferroptosis and ERS. Thus, it has potential for exploitation as a prognostic and immune therapeutic target for patients with ccRCC.
Collapse
Affiliation(s)
- Bei Feng
- Department of Nephrology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hai-Ying Guo
- Department of Nephrology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Ning
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu-Ying Zhao
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiang Wang
- Department of Nephrology, The First People's Hospital in Jinzhou, Dalian, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Cui
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
29
|
Zhu X, Cui Z, Li S, She Y, Wu Z. ADAMTSL2 is an independent predictor for the prognosis of gastric cancer. Discov Oncol 2025; 16:570. [PMID: 40252157 PMCID: PMC12009256 DOI: 10.1007/s12672-025-02259-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/28/2025] [Indexed: 04/21/2025] Open
Abstract
AIMS To explore novel biomarkers capable of predicting the prognosis of gastric cancer (GC) and investigate the mechanisms underlying the development of GC. METHODS Firstly, differentially expressed genes (DEGs) in GC tumors and adjacent tissues were analyzed using transcriptome sequencing data. Then, the DEGs significantly associated with the prognosis of GC were selected. From this subset, genes with high protein expression levels in tumor tissues were focused. Multivariate hazard analysis was performed to further identify DEGs with independent prognostic value for GC patients. Eventually, the potential mechanisms involving DEGs that underlie the development of GC were investigated. RESULTS Altogether, 25 previously DEGs that have not been reported before were discovered in the context of GC. Among these genes, ADAMTSL2, DSCC1, COL5A3, F2RL2, GRIN2D, IGSF6, IER5L, PLA2G7, PODNL1, RCN3 and RTN4RL2 were significantly associated with the overall survival, first progression and post progression survival of GC patients. Moreover, protein levels of ADAMTSL2, COL5A3, DSCC1, GRIN2D, PODNL1 and RCN3 were consistently highly expressed in clinical GC specimens. Furthermore, multivariate hazard analysis identified ADAMTSL2 as an independent predictor of GC prognosis. Further exploration revealed a potential regulatory connection between ADAMTSL2 and hsa-miR-7-2-3p. hsa-miR-7-2-3p was significantly down-regulated in GC and GC patients with low expression of hsa-miR-7 had a poor overall survival. Additionally, ADAMTSL2 was significantly co-expressed with key molecules (NOTCH1, NOTCH3, NOTCH4 and HEY1) in Notch signaling pathway. CONCLUSIONS ADAMTSL2 stands out as an independent predictor for the prognosis of GC and may play a crucial pathological role in the development of GC.
Collapse
Affiliation(s)
- Xiuling Zhu
- Department of Hepatobiliary, The 900th Hospital of Joint Service Support Force (Fuzong Clinical Medical College), Fujian Medical University, 156, North Xi'erhuan Rd, Fuzhou, 350025, Fujian, China
| | - Zhongyuan Cui
- Department of Hepatobiliary, The 900th Hospital of Joint Service Support Force (Fuzong Clinical Medical College), Fujian Medical University, 156, North Xi'erhuan Rd, Fuzhou, 350025, Fujian, China
| | - Shasha Li
- Department of Hepatobiliary, The 900th Hospital of Joint Service Support Force (Fuzong Clinical Medical College), Fujian Medical University, 156, North Xi'erhuan Rd, Fuzhou, 350025, Fujian, China
| | - Yingzhen She
- Department of Hepatobiliary, The 900th Hospital of Joint Service Support Force (Fuzong Clinical Medical College), Fujian Medical University, 156, North Xi'erhuan Rd, Fuzhou, 350025, Fujian, China
| | - Zhixian Wu
- Department of Hepatobiliary, The 900th Hospital of Joint Service Support Force (Fuzong Clinical Medical College), Fujian Medical University, 156, North Xi'erhuan Rd, Fuzhou, 350025, Fujian, China.
| |
Collapse
|
30
|
Karthikeyan SK, Chandrashekar DS, Sahai S, Shrestha S, Aneja R, Singh R, Kleer CG, Kumar S, Qin ZS, Nakshatri H, Manne U, Creighton CJ, Varambally S. MammOnc-DB, an integrative breast cancer data analysis platform for target discovery. NPJ Breast Cancer 2025; 11:35. [PMID: 40251157 PMCID: PMC12008238 DOI: 10.1038/s41523-025-00750-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/27/2025] [Indexed: 04/20/2025] Open
Abstract
Breast cancer (BCa), a leading malignancy among women, is characterized by morphological and molecular heterogeneity. While early-stage, hormone receptor, and HER2-positive BCa are treatable, triple-negative BCa and metastatic BCa remains largely untreatable. Advances in sequencing and proteomic technologies have improved our understanding of the molecular alterations that occur during BCa initiation and progression and enabled identification of subclass-specific biomarkers and therapeutic targets. Despite the availability of abundant omics data in public repositories, user-friendly tools for multi-omics data analysis and integration are scarce. To address this, we developed a comprehensive BCa data analysis platform called MammOnc-DB ( http://resource.path.uab.edu/MammOnc-Home.html ), comprising data from more than 20,000 BCa samples. MammOnc-DB facilitates hypothesis generation and testing, biomarker discovery, and therapeutic targets identification. The platform also includes pre- and post-treatment data, which can help users identify treatment resistance markers and support combination therapy strategies, offering researchers and clinicians a comprehensive tool for BCa data analysis and visualization.
Collapse
Affiliation(s)
| | | | - Snigdha Sahai
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadeep Shrestha
- Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Ritu Aneja
- School of Health Professions, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sidharth Kumar
- Department of Computer Science, University of Illinois Chicago, Chicago, IL, USA
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | | | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chad J Creighton
- Department of Medicine and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
31
|
Tang K, Liu M, Zhang C. Construction of a prognostic model and identification of key genes in liver hepatocellular carcinoma based on multi-omics data. Sci Rep 2025; 15:13393. [PMID: 40251374 PMCID: PMC12008308 DOI: 10.1038/s41598-025-98038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
Liver hepatocellular carcinoma (LIHC) strongly contributes to global cancer mortality, highlighting the need for a deeper understanding of its molecular mechanisms to enhance patient prognosis and treatment approaches. We aimed to investigate the differential expression of immunogenic cell death-related genes (ICDRGs) and cellular senescence-related genes (CSRGs) in LIHC and their effects on patient prognosis. We combined the GSE25097, GSE46408, and GSE121248 datasets by eliminating batch effects and standardizing the data. After processing, 16 genes were identified as ICDR&CSR differentially expressed genes (ICDR&CSRDEGs), including UBE2T, HJURP, PTTG1, CENPA, and FOXM1. Gene set enrichment analysis indicated a strong enrichment of these genes in pre-Notch expression and processing. Gene set variation analysis revealed 20 pathways with significant differences between the LIHC and control groups. Mutation analysis identified TP53 as the most commonly mutated gene in LIHC samples. A prognostic risk model integrating 12 ICDR&CSRDEGs was developed, showing high precision at 1 year but diminished accuracy at 2 and 3 years. Our constructed prognostic risk model provides valuable insights for predicting patient outcomes and may guide future therapeutic interventions targeting these specific genes. Further research is needed to explore the mechanistic roles of these genes in LIHC progression and treatment response.
Collapse
Affiliation(s)
- Kun Tang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China
| | - Mingjiang Liu
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China
| | - Cuisheng Zhang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China.
| |
Collapse
|
32
|
Lin Z, Long JW, Zhao MC, Guo P, Wen J, Chen GL. Purinosomes as a therapeutic target in hepatocellular carcinoma: insights and opportunities. Discov Oncol 2025; 16:564. [PMID: 40251459 PMCID: PMC12008087 DOI: 10.1007/s12672-025-02366-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 04/11/2025] [Indexed: 04/20/2025] Open
Abstract
The formation of purinosomes, dynamic complexes involved in de novo purine biosynthesis, has been recognized as a critical process for cell growth. Although their upregulation in cancer cells suggests their potential as a therapeutic target, the specific role of purinosomes in hepatocellular carcinoma (HCC) remains uncertain. The purinosome score was found to have prognostic value. Enrichment analyses indicated a connection between purinosome-related genes and cell cycle regulation. Moreover, our research has demonstrated a correlation between the upregulation of genes associated with purinosomes and the enhanced formation of purinosomes in Huh-7 cells. Pyrimethamine has been identified as a promising therapeutic option for targeting purinosome to exert anti-cancer effects. Furthermore, the purinosome score exhibited an positive relationship with the response to immunotherapy. It may guide the stratification of liver cancer patients and screen for populations that may benefit from immunotherapy. This study examines the prognostic and predictive value of purinosome in liver cancer, suggesting that targeting purinosome formation with pyrimethamine or immunotherapy could benefit patients with high purinosome scores.
Collapse
Affiliation(s)
- Zhen Lin
- Department of Medical Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Jia-Wei Long
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Ming-Chun Zhao
- Department of Pathology, Guilin Hospital of Chinese Traditional and Western Medicine, Guilin, 541004, China
| | - Pin Guo
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jin Wen
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, 55905, USA.
| | - Guang-Liang Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China.
| |
Collapse
|
33
|
Wang Y, Gao H, Li X, Li D, Huang F, Sun Y, Liu X, Yang J, Sun F. PRC1 as an independent adverse prognostic factor in Wilms tumor via integrated bioinformatics and experimental validation. Sci Rep 2025; 15:13282. [PMID: 40247060 PMCID: PMC12006549 DOI: 10.1038/s41598-025-98030-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Wilms Tumor (WT), a prevalent pediatric renal malignancy, exhibits marked heterogeneity and variable clinical outcomes. Epithelial-mesenchymal transition (EMT), a biological process enabling epithelial cells to acquire mesenchymal traits associated with enhanced migratory and invasive capacities, plays a crucial role in cancer progression. Protein Regulator of Cytokinesis 1 (PRC1) is a critical protein in cell division, whose overexpression is linked to poor prognosis in various cancers. This study investigates the role of PRC1 as a key prognostic factor in WT and explore the mechanism through comprehensive bioinformatic and experimental approaches. Through bulk RNA-seq data from the TARGET database, we identified PRC1 as significantly up-regulated in WT and associated with poor overall survival. Functional enrichment analyses (GO, KEGG, GSEA) demonstrated PRC1's involvement in cell division, chromatin dynamics, and activation of oncogenic pathways including Wnt/β-catenin, PI3K/AKT/mTOR, and Hedgehog signaling. Immunological analysis showed that elevated PRC1 expression correlates with diminished immune cell activity, particularly in NK cells, suggesting potential immune evasion mechanisms. Single-cell RNA-seq analysis (GSE200256) confirmed PRC1's elevated expression in anaplastic Wilms tumor (AWT) compared to favorable Wilms tumor (FWT), and highlighted its involvement in intercellular communication and metastasis via the EMT process. Genomic analyses identified copy number variations (CNVs) and downregulated PRC1-targeting microRNAs as drivers of its overexpression. In vitro, PRC1 knockdown in WIT-49 cells significantly impaired migratory capacity, invasive potential, EMT progression, and glycolytic metabolism. These findings collectively position PRC1 as a promising therapeutic target and prognostic biomarker in WT.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Hongjie Gao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Xuetian Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Ding Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Fan Huang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yuqiang Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xingjian Liu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Junli Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.
| | - Fengyin Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
34
|
Zhang C, Chen L. 6-Methoxyflavone inhibits glycolytic energy metabolism in HeLa cells. BMC Cancer 2025; 25:719. [PMID: 40247232 PMCID: PMC12004806 DOI: 10.1186/s12885-025-14133-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Enhanced glycolytic levels in cancer cells are a common characteristic of many cancer types. Modulation of glycolytic metabolism is crucial for enhancing the efficacy of cancer therapy. The specific role of 6-methoxyflavone in regulating glycolytic metabolism in cancer cells remains unclear. This study aimed to elucidate the impact of 6-methoxyflavone on glycolytic metabolism in cervical cancer cells and its clinical relevance. METHODS The tandem mass tag (TMT) proteomic analysis was used to identify significantly enriched biological processes and pathways in HeLa cells after treatment with 6-methoxyflavone. Additionally, the differential expression of glycolysis-related proteins was validated using parallel reaction monitoring (PRM) proteomics. Untargeted and targeted metabolomics analyses were used to identify differentially expressed glycolysis-related metabolites. Furthermore, alternative splicing, new transcripts, and domain analyses were used to detect the effects of 6-methoxyflavone on the structures of glycolysis-related genes and proteins. Subcellular localization, molecular docking, and non-covalent interaction analyses were used to detect the subcellular localization, affinity of 6-methoxyflavone for glycolysis-related proteins, and sites of non-covalent interactions. Clinical characteristics and immunological correlation analyses were used to elucidate the relationships between glycolysis-related genes and clinicopathological characteristics, survival, prognosis, and immune-related indicators of patients with cervical cancer. Finally, glycolysis stress tests and enzyme activity assays were used to verify the effect of 6-methoxyflavone on glycolysis in HeLa cells. RESULTS TMT and PRM proteomics, as well as untargeted and targeted metabolomics results, showed that 6-methoxyflavone downregulated the expression levels of glycolysis-related proteins and metabolites in HeLa cells, and that the structures and functions of glycolysis-related genes and proteins in the cytoplasm underwent changes. 6-Methoxyflavone had a good affinity for nine glycolysis-related proteins, all of which had non-covalent interaction sites. Clinical characteristics and immune correlation analyses showed relationships between 6-methoxyflavone and five clinical characteristics, survival prognosis, and four immune-related indicators in patients with cervical cancer. After treatment with 6-methoxyflavone, the basal glycolytic level, maximum glycolytic capacity, and glycolytic reserve of HeLa cells were downregulated. Additionally, 6-methoxyflavone inhibited the activity of pyruvate kinase. CONCLUSION 6-Methoxyflavone inhibited energy metabolism in HeLa cells through the glycolysis pathway. 6-Methoxyflavone may be related to five clinical characteristics, prognosis, tumor microenvironment, immune cells, immune checkpoints, and immunotherapy efficacy in patients with cervical cancer.
Collapse
Affiliation(s)
- Chaihong Zhang
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an City, Shaanxi Province, 710000, China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an City, Shaanxi Province, 710000, China.
| |
Collapse
|
35
|
Tan Y, Yang Y, Zhang M, Li N, Hu L, Deng M, Xiao Y, Wang Y, Tian F, Sun R. IRF4 as a molecular biomarker in pan-cancer through multiple omics integrative analysis. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:3183-3201. [PMID: 40176546 DOI: 10.1039/d4ay02269f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
IRF4, characterized by its unique helix-turn-helix DNA-binding motif, is a member of the interferon regulatory factor (IRF) family. It plays a critical role in regulating host defense mechanisms, including innate and adaptive immune responses, as well as oncogenesis. However, the precise role of IRF4 in malignant tumors remains poorly understood. In this study, we first investigated IRF4 gene expression across various cancer types and its distribution within different molecular and immunological subtypes, providing a comprehensive understanding of its expression patterns in pan-cancer. We further explored the interacting proteins, diagnostic significance, molecular characteristics, prognostic relevance, and biological functions of IRF4 in diverse cancers. Focusing on colorectal cancer (CRC), we conducted a detailed analysis of IRF4, examining its associations with clinical features and outcomes across multiple clinical subgroups and databases. Additionally, we assessed IRF4 expression at both transcriptional and translational levels in CRC tumor specimens using tissue microarrays. Our findings revealed that IRF4 expression varies significantly not only across cancer types but also among molecular and immunological subtypes. In CRC, elevated IRF4 expression was associated with poorer overall survival. Notably, IRF4 was predominantly expressed in immune cells and showed a strong correlation with tumor immune regulation. Given its high predictive accuracy for cancer outcomes and robust prognostic associations, IRF4 may serve as a valuable prognostic biomarker for CRC. In conclusion, IRF4 represents a unique molecular biomarker for pan-cancer prognosis and an independent prognostic risk factor for CRC. Its critical role in immune regulation also positions IRF4 as a promising target for immunotherapeutic strategies in CRC.
Collapse
Affiliation(s)
- Yiqing Tan
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiping Yang
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Mingjun Zhang
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Ni Li
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Lei Hu
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Mingyou Deng
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Yin Xiao
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Yingying Wang
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Fuhua Tian
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
| | - Ran Sun
- Department of Oncology, Chongqing Jiulongpo People's Hospital, Chongqing 400050, China.
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
36
|
Nesselbush MC, Luca BA, Jeon YJ, Jabara I, Meador CB, Garofalo A, Binkley MS, Hui AB, van 't Erve I, Xu N, Shi WY, Liu KJ, Sugio T, Kastelowitz N, Hamilton EG, Liu CL, Olsen M, Bonilla RF, Wang YP, Jiang A, Lau B, Eichholz J, Banwait M, Schroers-Martin J, Boegeholz J, King DA, Luikart H, Esfahani MS, Mehrmohamadi M, Stehr H, Raclin T, Tibshirani R, Khush K, Srinivas S, Yu H, Rogers AJ, Nair VS, Isbell JM, Li BT, Piotrowska Z, Sequist LV, Hata AN, Neal JW, Wakelee HA, Gentles AJ, Alizadeh AA, Diehn M. An ultrasensitive method for detection of cell-free RNA. Nature 2025:10.1038/s41586-025-08834-1. [PMID: 40240612 DOI: 10.1038/s41586-025-08834-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
Sensitive methods for detection of cell-free RNA (cfRNA) could facilitate non-invasive gene expression profiling and monitoring of diseases1-6. Here we describe RARE-seq (random priming and affinity capture of cfRNA fragments for enrichment analysis by sequencing), a method optimized for cfRNA analysis. We demonstrate that platelet contamination can substantially confound cfRNA analyses and develop an approach to overcome it. In analytical validations, we find RARE-seq to be approximately 50-fold more sensitive for detecting tumour-derived cfRNA than whole-transcriptome RNA sequencing (RNA-seq), with a limit of detection of 0.05%. To explore clinical utility, we profiled 437 plasma samples from 369 individuals with cancer or non-malignant conditions and controls. Detection of non-small-cell lung cancer expression signatures in cfRNA increased with stage (6 out of 20 (30%) in stage I; 5 out of 8 (63%) in stage II; 10 out of 15 (67%) in stage III; 80 out of 96 (83% sensitivity) in stage IV at 95% specificity) and RARE-seq was more sensitive than tumour-naive circulating tumour DNA (ctDNA) analysis. In patients with EGFR-mutant non-small-cell lung cancer who developed resistance to tyrosine kinase inhibitors, we detected both histological transformation and mutation-based resistance mechanisms. Finally, we demonstrate the potential utility of RARE-seq for determination of tissue of origin, assessing benign pulmonary conditions and tracking response to mRNA vaccines. These results highlight the potential value of ultrasensitive cfRNA analysis and provide proof of concept for diverse clinical applications.
Collapse
Affiliation(s)
- Monica C Nesselbush
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Bogdan A Luca
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Isabel Jabara
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Catherine B Meador
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrea Garofalo
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Program in Cancer Biology, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michael S Binkley
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Angela B Hui
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Iris van 't Erve
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Nova Xu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - William Y Shi
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Kevin J Liu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Takeshi Sugio
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Noah Kastelowitz
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Emily G Hamilton
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Chih Long Liu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mari Olsen
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Rene F Bonilla
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Yi Peng Wang
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Alice Jiang
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Brianna Lau
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Jordan Eichholz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mandeep Banwait
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph Schroers-Martin
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Jan Boegeholz
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Daniel A King
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Helen Luikart
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mohammad S Esfahani
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mahya Mehrmohamadi
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Henning Stehr
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Tyler Raclin
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Kiran Khush
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Sandy Srinivas
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Helena Yu
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Angela J Rogers
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Viswam S Nair
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pulmonary, Critical Care & Sleep Medicine, University of Washington, Seattle, WA, USA
| | - James M Isbell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bob T Li
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zofia Piotrowska
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron N Hata
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Joel W Neal
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Heather A Wakelee
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | | | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
37
|
Li Y, Yang W, Chen H, Jin Z, Dong J, Ma L, Ji Z. Comprehensive pan-cancer single-cell analysis reveals glycolysis-related signatures as predictive biomarkers for immunotherapy response and their role in bladder cancer. Int Immunopharmacol 2025; 152:114381. [PMID: 40058104 DOI: 10.1016/j.intimp.2025.114381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Glycolysis is a vital metabolic biological process in tumor progression and immune modulation. This study comprehensively investigated the roles of glycolysis in pan-cancer, especially in bladder cancer. Exploration of 34 single-cell RNA sequencing (scRNA-seq) cohorts, eight ICI-treated bulk RNA-seq cohorts, and TCGA bulk pan-cancer RNA-seq cohorts uncovered a Glycolysis.Sig which strongly correlated with immunotherapy response and demonstrated excellent predictive performance in prognosis and immune response. Hub-Glycolysis.Sig exhibited varying interactions with the immune microenvironment based on cancer type. In bladder cancer, higher glycolysis risk scores correlated with poorer prognosis, with distinct immune infiltration characteristics between subtypes. scRNA-seq revealed high glycolysis levels in bladder epithelial cells. COPB2 was highly expressed in bladder cancer, promoting cell proliferation, migration, and glycolytic activity in vitro and in vivo. Our large-scale data analysis confirmed the negative correlation between glycolysis and immunotherapy outcomes, identifying Glycolysis.Sig as a novel predictive biomarker. Hub-Glycolysis.Sig provides clinical insights for bladder cancer therapy strategies, while COPB2 and other potential therapeutic targets facilitate personalized cancer treatment.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Wenjie Yang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Hualin Chen
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhaoheng Jin
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Jie Dong
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Lin Ma
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China.
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China.
| |
Collapse
|
38
|
Cui Q, Fu S, Yu D, Li M, Li Y. Impact of Non-SMC Condensin I Complex Subunit D2 Upregulation on Oral Squamous Cell Carcinoma Prognosis. Int Dent J 2025; 75:1818-1827. [PMID: 40245749 PMCID: PMC12022477 DOI: 10.1016/j.identj.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/23/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE To explore the influence of non-SMC condensin I complex subunit D2 (NCAPD2) on the prognosis of oral squamous cell carcinoma (OSCC) and the correlation between NCAPD2 and OSCC. METHODS In this study, NCAPD2 gene expression profiles of OSCC and normal tissues were collected from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The real-time quantitative polymerase chain reaction (RT-qPCR) was employed to preliminarily validate OSCC cell strains and normal epithelial cell strains. Besides EdU, cell scratch, and transwell assays were performed to assess the proliferation, migration, and invasion of OSCC cell strains with the silence of NCAPD2. Moreover, immunohistochemistry (IHC) staining was utilised to measure the expression of NCAPD2 and tumour-related markers in 74 OSCC specimens. Finally, the Kaplan-Meier analysis was performed to evaluate the influence of NCAPD2 in the prognosis of OSCC. RESULTS The expression of NCAPD2 in OSCC tissues was higher than that in normal tissues. Inhibiting NCAPD2 can reduce the proliferation and migration of OSCC cell lines and inhibit the invasion of these cells. The IHC staining results indicated that the high expression of NCAPD2 in OSCC tissues was positively correlated with T stages, Ki67 expression, and affected sites. The Kaplan-Meier analysis results validated that the up-regulated expression of NCAPD2 was significantly correlated with the poor overall survival (OS) of OSCC patients. CONCLUSION NCAPD2 is a potential molecular marker for the poor prognosis of OSCC, and it is expected to become a target for the treatment of this carcinoma.
Collapse
Affiliation(s)
- Qingying Cui
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shuai Fu
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Diping Yu
- Department of Pathology, Hospital of Pu'er, Kunming University of Science and Technology, Kunming, China
| | - Ming Li
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Yong Li
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
39
|
Zhang S, Hu X, Sun M, Chen X, Le S, Wang X, Wang J, Hu Z. Potential role of hypobaric hypoxia environment in treating pan-cancer. Sci Rep 2025; 15:12942. [PMID: 40234469 PMCID: PMC12000279 DOI: 10.1038/s41598-024-84561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/24/2024] [Indexed: 04/17/2025] Open
Abstract
Cancer incidence and mortality are lower among high-altitude residents, suggesting that hypobaric hypoxia (HH) might protect against cancer. Our study aimed to develop a pan-cancer prognosis risk model using ADME genes, which are influenced by low oxygen, to explore HH's impact on overall survival (OS) across various cancers. We constructed and validated the model with gene expression and survival data from 8628 samples, using three gene expression databases. AltitudeOmics confirmed HH's significant effects. We employed single-gene prognostic analysis, weighted gene co-expression network analysis, and stepwise Cox regression to identify biomarkers and refine the model. Drugs interacting with the model were explored using LINCS L1000, AutoDockTools, and STITCH. Eight ADME genes significantly altered by HH were identified, revealing their prognostic value across cancers. The model showed lower risk scores linked to better prognosis in 25 cancers, with reduced overall gene expression and decreased tumor mortality risk. Higher T cell infiltration was observed in the low-risk group. Additionally, three potential drugs to modulate our model were identified. This study presents a novel pan-cancer survival prognosis model based on ADME genes influenced by HH, offering new insights into cancer prevention and treatment.
Collapse
Affiliation(s)
- Shixuan Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Xiaoxi Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Mengzhen Sun
- Zhangjiang Fudan International Innovation Centre, Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinrui Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Shiguan Le
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Xilu Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China.
| | - Zixin Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200438, China.
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, China.
| |
Collapse
|
40
|
Zhou M, Zhao W, Zhang X, Cheng Y, Wang M, Chen Y, Zhao L. Nicotinamide metabolism affects the prognosis of hepatocellular carcinoma by influencing the tumor microenvironment. Cytokine 2025; 191:156939. [PMID: 40228405 DOI: 10.1016/j.cyto.2025.156939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/15/2025] [Accepted: 04/05/2025] [Indexed: 04/16/2025]
Abstract
In this study, we utilized the public database along with single-cell genomics techniques to systematically analyze the expression patterns and clinical significance of key genes in the nicotinamide metabolism pathway in liver cancer samples. The findings indicate that differential nicotinamide metabolism-related key genes are expressed in liver cancer samples. The liver cancer samples were put into separate subgroups using consistency clustering analysis based on differential gene expression levels observed. Additionally, immune infiltration and drug sensitivity analysis also revealed differences between the two subgroups. Survival analysis suggested that the key genes were associated with prognosis. Finally, a prognostic model was established using the key genes, offering a fresh viewpoint on the molecular mechanism investigating liver cancer. This study demonstrated the significant correlation between key genes in the nicotinamide metabolism pathway and the occurrence and progression of liver cancer and indicated that these key genes could serve as prognostic markers and tailored treatment targets for liver cancer.
Collapse
Affiliation(s)
- Min Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Wenhui Zhao
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Xiaobo Zhang
- School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Ye Cheng
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Mengxiang Wang
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Yan Chen
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China.
| | - Lingrui Zhao
- School of Life Sciences, Westlake University, Hangzhou, 310024, China.
| |
Collapse
|
41
|
Zhao B, Wu J, Zhang T, Han M, Zhang C, Rong X, Zhang R, Chen X, Peng F, Jin J, Liu S, Dong X, Zhao S. A spatial transcriptomics study of MES-like and mono/macro cells in gliomas. Sci Rep 2025; 15:12730. [PMID: 40222970 PMCID: PMC11994772 DOI: 10.1038/s41598-025-95277-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Gliomas, including both glioblastoma multiforme (GBM) and lower-grade glioma (LGG), present a substantial challenge in neuro-oncology because of genetic heterogeneity and unsatisfactory prognosis. This study aimed to conduct a comprehensive multi-omics analysis of gliomas using various bioinformatics approaches to identify potential therapeutic targets and prognostic markers. A comprehensive analysis was conducted on 1327 sequencing data samples alongside their relevant clinical information sourced from The Cancer Genome Atlas (TCGA) pertaining to glioblastoma (GBM), low-grade glioma (LGG), the Chinese Glioma Genome Atlas (CCGA) and University of California Santa Cruz Xena (UCSC Xena) datasets. These tools were employed for gene expression profiling, survival analysis, and cell communication mapping. Spatial transcriptomics revealed the localization of mesenchymal (MES)-like malignant tumors, and drug sensitivity analysis was performed to evaluate responses to quinpirole and meropenem. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) framework was utilized to gauge the responsiveness to immunotherapy. The MES-like malignant and monocyte/macrophage (mono/macro) cell subsets showed high hallmark scores, playing key roles in the tumor microenvironment. MES-like malignant marker gene scores correlated with overall survival across datasets, whereas mono/macro marker gene scores were significant in the TCGA-LGG and CCGA datasets. Key interactions between these cell types were found, especially with CD14-ITGB2, LGALS1-CD69, and APOE-TREM2. The mono/macro cell subset demonstrated better immune therapy responsiveness, as indicated by lower TIDE scores. Spatial transcriptomics revealed that MES-like malignant tumors are predominantly localized in four distinct regions, with the marker genes CHI3L1 and ADM confirming these locations. Drug sensitivity analysis revealed differential responses of the MES-like malignant cell subset to quinpirole and meropenem. Our results offer fresh perspectives on the differential roles of MES-like malignant and monocyte/macrophage cell subsets in tumor progression and immune modulation, providing novel insights into glioma biology.
Collapse
Affiliation(s)
- Boyan Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Jianing Wu
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Tiehui Zhang
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong, China
| | - Mingyang Han
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Cheng Zhang
- University of Toronto Scarborough 1265 Military Trail, Scarborough, ON, M1C 1A4, Canada
| | - Xuan Rong
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, 150001, Heilongjiang, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Jin
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Shiya Liu
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Xingli Dong
- Central Laboratory, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
| | - Shiguang Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China.
- Department of Neurosurgery, Shenzhen University General Hospital, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
42
|
Li ZY, Li MF, He YY, Zheng GS, Chen JR, Guo YM, Lian Q, Yue CF. Construction of a Prognostic Model based on CSC-related Genes in Patients with Colorectal Cancer. J Cancer 2025; 16:2375-2387. [PMID: 40302814 PMCID: PMC12036084 DOI: 10.7150/jca.108188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common and deadly malignancies. Lack of efficient biomarkers for prognosis has limited the improvement of survival outcome in patients with CRC. Numerous studies have demonstrated the important roles of cancer stem cells (CSCs) in both treatment resistance and disease recurrence of CRC. Thus, the current study aims to construct a prognostic model based on expression level of CSC-related genes for precise molecular subtyping of CRC patients with different prognoses, TME infiltration patterns and therapeutic responses. The RNA sequencing data and clinical information were obtained from UCSC Xena database, followed by identification of differential expressed genes, univariate Cox regression, and LASSO regression to identify prognostic CSC-related genes and construct a novel prognostic risk scoring model consisting of 21 CSC-related genes. The patients in high-risk group suffered poor survival outcome (P<0.0001). Moreover, the performance of CSC-related prognostic model was validated in individual GEO datasets including GSE41258 and GSE39582 (P<0.05). Furthermore, patients with high-risk score exhibited lower response rate to immune checkpoint inhibitors as compared to those in low-risk group (17.4% vs. 28.2%), indicating the potential of CSC-related prognostic model to predict the immunotherapy response. Collectively, our findings provide an effective model to predict the immunotherapy response and survival outcome in patients with CRC.
Collapse
Affiliation(s)
- Zi-Yue Li
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang 524045, China
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ming-Feng Li
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang 524045, China
| | - Ying-Ying He
- Department of Anesthesiology, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang 524045, P. R. China
| | - Guan-Sheng Zheng
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jie-Rong Chen
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yun-Miao Guo
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang 524045, China
| | - Qizhou Lian
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Cai-Feng Yue
- Department of Laboratory Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang 524045, China
| |
Collapse
|
43
|
Yang C, Cheng D, Wang S, Wang B, Li Y, Wang G, Wang X, Shi C, Tian Y, Zhu K, Feng J. Identification of the role of MED6 in the development and prognosis of lung adenocarcinoma based on multi-omics profiling. J Cancer 2025; 16:2362-2374. [PMID: 40302793 PMCID: PMC12036094 DOI: 10.7150/jca.110981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/23/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Lung adenocarcinoma (LUAD) is the most common subtype of lung cancer. Recent studies have highlighted the importance of Mediator complex subunits in cancer, but their specific roles in LUAD are still unclear. Methods: The CRISPR-Cas9 loss-of-function data was used to assess gene dependency in cell growth. RNA-seq data were analyzed to evaluate the prognostic value of Mediator subunits and explore their downstream pathways. Single-cell sequencing data were utilized to examine the tumor microenvironment in LUAD. A drug sensitivity analysis was performed to identify potential therapeutic options. Results: Mediator complex subunit 6 (MED6) was found to influence tumor cell growth in LUAD. Additionally, MED6 expression levels were associated with patient prognosis. MED6-positive tumor cells showed more active interactions with other cells in the LUAD microenvironment, promoting tumor progression. Based on MED6 expression, drugs such as paclitaxel, afatinib, and brivanib were identified as potential treatments. Conclusions: This study revealed the role of MED6 in LUAD and its potential as a biomarker. Our findings suggest that MED6 has an effect on LUAD progression and provide valuable insights for patient stratification and personalized treatment strategies.
Collapse
Affiliation(s)
- Changqing Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ding Cheng
- Department of Plastic Surgery and Medical Aesthetics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shuo Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Baichuan Wang
- Anhui Chest Hospital, Hefei, Anhui Province, 23002, China
| | - Yingxi Li
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, China
| | - Guixin Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Xingkai Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Cangchang Shi
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Yao Tian
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Keyun Zhu
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang Province, 315040, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| |
Collapse
|
44
|
Tian Y, Yang Y, He L, Yu X, Zhou H, Wang J. Exploring the tumor microenvironment of breast cancer to develop a prognostic model and predict immunotherapy responses. Sci Rep 2025; 15:12569. [PMID: 40221624 PMCID: PMC11993623 DOI: 10.1038/s41598-025-97784-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Breast cancer is the most prevalent malignancy in women and exhibits significant heterogeneity. The tumor microenvironment (TME) plays a critical role in tumorigenesis, progression, and response to therapy. However, its impact on the prognosis and immunotherapy responses is incompletely understood. Using public databases, we conducted a comprehensive investigation of transcriptome and single-cell sequencing data. After performing immune infiltration analysis, we conducted consensus clustering, weighted gene co-expression network analysis (WGCNA), Cox regression, and least absolute shrinkage and selection operator (Lasso) regression to identify independent prognostic genes in breast cancer. Subsequently, we developed a prognostic model for patients with breast cancer. Tumor Immune Dysfunction and Exclusion (TIDE) values were used to assess patient's responsiveness to breast cancer. Based on single-cell RNA-sequencing data, we identified various cell types through cluster analysis and investigated the expression of prognostic model genes in each cell type. The drug sensitivity of targeted therapeutic agents for breast cancer treatment was analyzed in different cell types. We identified 12 independent prognostic genes associated with breast cancer and used these genes to construct a prognostic model. The prognostic model accurately discriminated between patients classified as high- and low-risk, providing precise prognostic predictions for individual patients. Additionally, our model exhibited a robust capacity to predict the immunotherapeutic response in breast cancer patients. Our investigation revealed a notable association between the proportion of endothelial cells (ECs) and patient prognosis in breast cancer. A prognostic model for breast cancer was formulated that showed close associations between prognosis and response to immunotherapy. For patients predicted by our model to not respond effectively to immunotherapeutic agents, it may be considered to combine immunotherapeutic agents with targeted therapeutic agents identified through our drug sensitivity analysis, which could potentially enhance treatment efficacy.
Collapse
Affiliation(s)
- Ye Tian
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei He
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaocheng Yu
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Zhou
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Wang
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
45
|
Zhao LL, Liu YJ, Guo QJ, Yan N, Yang J, Han JQ, Xie XH, Luo YS. TPM4 influences the initiation and progression of gastric cancer by modulating ferroptosis via SCD1. Clin Exp Med 2025; 25:115. [PMID: 40214825 PMCID: PMC11991984 DOI: 10.1007/s10238-025-01629-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Gastric cancer (GC) is a deadly disease with poor prognosis and few treatment options. Tropomyosin 4 (TPM4) is an actin-binding protein that stabilizes the cytoskeleton of cells and has an unclear role in GC. This study aimed to elucidate the role and underlying mechanisms of TPM4 in GC pathogenesis. The expression and diagnostic and prognostic value of TPM4 in GC were analyzed using bioinformatics. A nomogram based on TPM4 expression was created and validated with an external cohort. TPM4-knockdown GC cells and xenograft models in nude mice were used to study the function of TPM4 in vitro and in vivo. Proteomic and rescue experiments confirmed the regulatory effect of TPM4 on stearoyl-CoA desaturase 1 (SCD1) in GC. Immunohistochemistry verified the expression and correlation of the TPM4 and SCD1 proteins in GC tissues. Our study identified TPM4 as an oncogene in GC, suggesting its potential diagnostic and prognostic value. The TPM4-based nomogram showed potential prognostic value for clinical use. TPM4 knockdown inhibited GC cell proliferation, induced ferroptosis, and slowed tumor growth in vivo, which is achieved by inhibiting SCD1 expression. Immunohistochemical analysis of GC tissues revealed elevated expression levels of both TPM4 and SCD1 proteins, with a positive correlation observed between their expression. TPM4 is a promising target for new diagnostic, prognostic, and therapeutic strategies for GC. Downregulation of TPM4 inhibits GC cell growth and induces ferroptosis by suppressing SCD1 expression.
Collapse
Affiliation(s)
- Ling-Lin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Jun Liu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Qi-Jing Guo
- Department of Oncology, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jie Yang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jing-Qi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Xiao-Hong Xie
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Shuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China.
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China.
| |
Collapse
|
46
|
Markey M, Kim J, Goldstein Z, Gerardin Y, Brosnan-Cashman J, Javed SA, Juyal D, Pagidela H, Yu L, Rahsepar B, Abel J, Hennek S, Khosla A, Taylor-Weiner A, Parmar C. Spatial mapping of gene signatures in H&E-stained images: a proof of concept for interpretable predictions using additive multiple instance learning. Mod Pathol 2025:100772. [PMID: 40222651 DOI: 10.1016/j.modpat.2025.100772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/06/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
The relative abundance of cancer-associated fibroblast (CAF) subtypes influences a tumor's response to treatment, especially immunotherapy. However, the gene expression signatures associated with these CAF subtypes have yet to realize their potential as clinical biomarkers. Here, we describe an interpretable machine learning (ML) approach, additive multiple instance learning (aMIL), to predict bulk gene expression signatures from hematoxylin and eosin (H&E)-stained whole slide images (WSI), focusing on an immunosuppressive LRRC15+ CAF-enriched TGFβ-CAF signature. aMIL models accurately predicted TGFβ-CAF across various cancer types. Tissue regions contributing most highly to slide-level predictions of TGFβ-CAF were evaluated by ML models characterizing spatial distributions of diverse cell and tissue types, stromal subtypes, and nuclear morphology. In breast cancer, regions contributing most to TGFβ-CAF-high predictions ("excitatory") were localized to cancer stroma with high fibroblast density and mature collagen fibers. Regions contributing most to TGFβ-CAF-low predictions ("inhibitory") were localized to cancer epithelium and densely inflamed stroma. Fibroblast and lymphocyte nuclear morphology also differed between excitatory and inhibitory regions. Thus, aMIL enables a data-driven link between histologic features and transcription, offering biological interpretability beyond typical black-box models.
Collapse
|
47
|
Huang J, Zhang H, Lin X, Wu X, Chen X, Chen W, Liang S, Chen Y, Luo Q, Xu C, Liu S, Liu X, Zhang S. Regulatory T Cell Infiltration-Driven Single-Cell Transcriptomic Analysis Identifies SAP18 as a Prognostic Marker for Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer 2025; 56:97. [PMID: 40208395 DOI: 10.1007/s12029-025-01174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Advanced esophageal squamous cell carcinoma (ESCC) is characterized by molecular heterogeneity and distinct patterns of immune cell infiltration. Regulatory T cells (Tregs), in particular, play a critical role in shaping an immunosuppressive tumor microenvironment (TME), which is associated with poor clinical outcomes. METHODS We developed a prognostic model by integrating GEO-derived bulk RNA sequencing data and single-cell transcriptome. Model predictions were confirmed through RT-qPCR, Western blot, and immunohistochemistry on clinical specimens, while in vitro assays (CCK8, transwell invasion, scratch, colony formation, and immunofluorescence) validated the function of SAP18 in cell proliferation, invasion, and ECM remodeling. RESULTS Expression patterns of the 5 Tregs-associated genes in clinical specimens aligned with model predictions, underscoring the model's robustness. The high-risk subgroup was associated with upregulated extracellular matrix (ECM) remodeling, an abundance of immune-suppressive cells, higher TP53 mutation rate, and limited benefit from immunotherapy. In contrast, the low-risk subgroup exhibited anti-tumor immunity. Cell-cell communication analysis also implicated the collagen pathway in Tregs-mediated immune evasion in ESCC. Functional assays indicated that SAP18 in the prognostic model significantly promotes proliferation, invasion, and ECM reconstruction, further highlighting its potential as a therapeutic target. CONCLUSION Our findings elucidate the role of Tregs in the TME, underscoring significant potential of SAP18, which is essential for assessing patient prognosis and may facilitate the development of personalized therapies for ESCC.
Collapse
Affiliation(s)
- Jianxiang Huang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Hanshuo Zhang
- Gastrointestinal Anorectal Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Xinyue Lin
- Department of Pharmacology, Medical College of Shantou University, Shantou, 515063, PR China
| | - Xiaolong Wu
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Xiaoshan Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Shanshan Liang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Qianhua Luo
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Chengcheng Xu
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Shaojie Liu
- Gastrointestinal Anorectal Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Xingmei Liu
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China.
- Department of Nursing, Guangzhou Red Cross Hospital of Jinan University, Haizhu District, No. 396, Tongfuzhong Road, Guangzhou, 510220, PR China.
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China.
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China.
| |
Collapse
|
48
|
Osborn G, López-Abente J, Adams R, Laddach R, Grandits M, Bax HJ, Chauhan J, Pellizzari G, Nakamura M, Stavraka C, Chenoweth A, Palhares LCGF, Evan T, Lim JHC, Gross A, Moise L, Jatiani S, Figini M, Bianchini R, Jensen-Jarolim E, Ghosh S, Montes A, Sayasneh A, Kristeleit R, Tsoka S, Spicer J, Josephs DH, Karagiannis SN. Hyperinflammatory repolarisation of ovarian cancer patient macrophages by anti-tumour IgE antibody, MOv18, restricts an immunosuppressive macrophage:Treg cell interaction. Nat Commun 2025; 16:2903. [PMID: 40210642 PMCID: PMC11985905 DOI: 10.1038/s41467-025-57870-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/06/2025] [Indexed: 04/12/2025] Open
Abstract
Ovarian cancer is the most lethal gynaecological cancer and treatment options remain limited. In a recent first-in-class Phase I trial, the monoclonal IgE antibody MOv18, specific for the tumour-associated antigen Folate Receptor-α, was well-tolerated and preliminary anti-tumoural activity observed. Pre-clinical studies identified macrophages as mediators of tumour restriction and pro-inflammatory activation by IgE. However, the mechanisms of IgE-mediated modulation of macrophages and downstream tumour immunity in human cancer remain unclear. Here we study macrophages from patients with epithelial ovarian cancers naive to IgE therapy. High-dimensional flow cytometry and RNA-seq demonstrate immunosuppressive, FcεR-expressing macrophage phenotypes. Ex vivo co-cultures and RNA-seq interaction analyses reveal immunosuppressive associations between patient-derived macrophages and regulatory T (Treg) cells. MOv18 IgE-engaged patient-derived macrophages undergo pro-inflammatory repolarisation ex vivo and display induction of a hyperinflammatory, T cell-stimulatory subset. IgE reverses macrophage-promoted Treg cell induction to increase CD8+ T cell expansion, a signature associated with improved patient prognosis. On-treatment tumours from the MOv18 IgE Phase I trial show evidence of this IgE-driven immune signature, with increased CD68+ and CD3+ cell infiltration. We demonstrate that IgE induces hyperinflammatory repolarised states of patient-derived macrophages to inhibit Treg cell immunosuppression. These processes may collectively promote immune activation in ovarian cancer patients receiving IgE therapy.
Collapse
Affiliation(s)
- Gabriel Osborn
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Jacobo López-Abente
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Rebecca Adams
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Roman Laddach
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, UK
| | - Melanie Grandits
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Heather J Bax
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Jitesh Chauhan
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Giulia Pellizzari
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Mano Nakamura
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Chara Stavraka
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Alicia Chenoweth
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| | - Lais C G F Palhares
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | - Theodore Evan
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
| | | | | | | | | | - Mariangela Figini
- ANP2, Department of Advanced Diagnostics, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Rodolfo Bianchini
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
- Center of Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University Vienna, Vienna, Austria
| | - Sharmistha Ghosh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ana Montes
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ahmad Sayasneh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Rebecca Kristeleit
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, Bush House, London, UK
| | - James Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Debra H Josephs
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, Guy's Hospital, London, UK.
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK.
| |
Collapse
|
49
|
Pîrlog LM, Pătrășcanu AA, Ona MD, Cătană A, Rotar IC. HOXA10 and HOXA11 in Human Endometrial Benign Disorders: Unraveling Molecular Pathways and Their Impact on Reproduction. Biomolecules 2025; 15:563. [PMID: 40305321 PMCID: PMC12024888 DOI: 10.3390/biom15040563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
HOX genes, a family of conserved transcription factors, are critical for reproductive tract development and endometrial functionality. This review highlights the molecular underpinnings of HOXA10/HOXA11 in reproductive health and their dysregulation in benign pathologies associated with infertility, such as endometriosis, adenomyosis, and endometrial polyps. These genes are dynamically regulated by estrogen and progesterone, with peak expression during the secretory phase of the menstrual cycle when implantation takes place. The molecular mechanisms underlying their action include the modulation of extracellular matrix (ECM) remodeling via metalloproteinases, cytokines like leukemia inhibitory factor, and cell adhesion molecules such as β3-integrin, all of which are essential for the differentiation of epithelial and stromal cells, as well as for trophoblast invasion. Aberrant HOX gene expression, driven by DNA hypermethylation or altered histone acetylation, compromises endometrial receptivity and implantation. For instance, reduced HOXA10 expression in endometriosis stems from hypermethylation and chronic inflammation, disrupting immune modulation and cytokine signaling. Similarly, adenomyosis alters HOXA11-regulated ECM remodeling and β3-integrin expression, impairing embryo attachment. Furthermore, regulatory pathways involving vitamin D and retinoic acid offer promising therapeutic avenues pathways, as they enhance HOXA10/HOXA11 expression and endometrial receptivity. This review underscores the critical molecular roles of HOXA10/HOXA11 genes as biomarkers and therapeutic targets to optimize fertility outcomes and address reproductive pathologies.
Collapse
Affiliation(s)
- Lorin-Manuel Pîrlog
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (L.-M.P.); (M.-D.O.)
| | - Andrada-Adelaida Pătrășcanu
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (L.-M.P.); (M.-D.O.)
| | - Mara-Diana Ona
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (L.-M.P.); (M.-D.O.)
| | - Andreea Cătană
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (L.-M.P.); (M.-D.O.)
- Regional Laboratory Cluj-Napoca, Department of Medical Genetics, Regina Maria Health Network, 400363 Cluj-Napoca, Romania
- Department of Oncogenetics, “Prof. Dr. I. Chiricuță” Institute of Oncology, 400015 Cluj-Napoca, Romania
| | - Ioana Cristina Rotar
- 1st Department of Obstetrics and Gynecology, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400006 Cluj-Napoca, Romania;
| |
Collapse
|
50
|
Jerabkova-Roda K, Peralta M, Huang KJ, Mousson A, Bourgeat Maudru C, Bochler L, Busnelli I, Karali R, Justiniano H, Lisii LM, Carl P, Mittelheisser V, Asokan N, Larnicol A, Lefebvre O, Lachuer H, Pichot A, Stemmelen T, Molitor A, Scheid L, Frenger Q, Gros F, Hirschler A, Delalande F, Sick E, Carapito R, Carapito C, Lipsker D, Schauer K, Rondé P, Hyenne V, Goetz JG. Peripheral positioning of lysosomes supports melanoma aggressiveness. Nat Commun 2025; 16:3375. [PMID: 40204688 PMCID: PMC11982396 DOI: 10.1038/s41467-025-58528-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Emerging evidence suggests that the function and position of organelles are pivotal for tumor cell dissemination. Among them, lysosomes stand out as they integrate metabolic sensing with gene regulation and secretion of proteases. Yet, how their function is linked to their position and how this controls metastasis remains elusive. Here, we analyze lysosome subcellular distribution in patient-derived melanoma cells and patient biopsies and show that lysosome spreading scales with melanoma aggressiveness. Peripheral lysosomes promote matrix degradation and cell invasion which is directly linked to the lysosomal and cell transcriptional programs. Using chemo-genetical control of lysosome positioning, we demonstrate that perinuclear clustering impairs lysosome secretion, matrix degradation and invasion. Impairing lysosome spreading significantly reduces invasive outgrowth in two in vivo models, mouse and zebrafish. Our study provides a direct demonstration that lysosome positioning controls cell invasion, illustrating the importance of organelle adaptation in carcinogenesis and suggesting its potential utility for diagnosis of metastatic melanoma.
Collapse
Affiliation(s)
- Katerina Jerabkova-Roda
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
- Institut Curie, PSL, CNRS, UMR144, Paris, France.
| | - Marina Peralta
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, 00015, Rome, Italy
| | - Kuang-Jing Huang
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Antoine Mousson
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Clara Bourgeat Maudru
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Louis Bochler
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Ignacio Busnelli
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Rabia Karali
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Hélène Justiniano
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Lucian-Mihai Lisii
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Philippe Carl
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Vincent Mittelheisser
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Nandini Asokan
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Annabel Larnicol
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Olivier Lefebvre
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Hugo Lachuer
- Institut Curie, PSL, CNRS, UMR144, Paris, France
- Institut Gustave Roussy, INSERM UMR1279, Université Paris-Saclay, Villejuif, France
- Université de Paris, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Angélique Pichot
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Tristan Stemmelen
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Anne Molitor
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | - Léa Scheid
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Quentin Frenger
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Frédéric Gros
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Aurélie Hirschler
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - François Delalande
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Emilie Sick
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Raphaël Carapito
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Dan Lipsker
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Kristine Schauer
- Institut Curie, PSL, CNRS, UMR144, Paris, France.
- Institut Gustave Roussy, INSERM UMR1279, Université Paris-Saclay, Villejuif, France.
| | - Philippe Rondé
- Université de Strasbourg, Strasbourg, France.
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France.
| | - Vincent Hyenne
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
- CNRS, SNC5055, Strasbourg, France.
| | - Jacky G Goetz
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|