251
|
Azadegan C, Santoro J, Whetstine JR. CONNECTING THE DOTS: EPIGENETIC REGULATION OF EXTRACHROMOSOMAL AND INHERITED DNA AMPLIFICATIONS. J Biol Chem 2025:108454. [PMID: 40154613 DOI: 10.1016/j.jbc.2025.108454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025] Open
Abstract
DNA amplification has intrigued scientists for decades. Since its discovery, significant progress has been made in understanding the mechanisms promoting DNA amplification and their associated function(s). While DNA copy gains were once thought to be regulated purely by stochastic processes, recent findings have revealed the important role of epigenetic modifications in driving these amplifications and their integration into the genome. Furthermore, advances in genomic technology have enabled detailed characterization of these genomic events in terms of size, structure, formation, and regulation. This review highlights how our understanding of DNA amplifications has evolved over time, tracing its trajectory from initial discovery to the contemporary landscape. We describe how recent discoveries have started to uncover how these genomic events occur by controlled biological processes rather than stochastic mechanisms, presenting opportunities for therapeutic modulation.
Collapse
Affiliation(s)
- Chloe Azadegan
- Drexel University, College of Medicine, Philadelphia, PA, 19111; Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111
| | - John Santoro
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111
| | - Johnathan R Whetstine
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111.
| |
Collapse
|
252
|
Yao L, Jiang B, Xu D. Strategies to combat cancer drug resistance: focus on copper metabolism and cuproptosis. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:15. [PMID: 40201308 PMCID: PMC11977383 DOI: 10.20517/cdr.2025.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
Cancer cells often develop tolerance to chemotherapy, targeted therapy, and immunotherapy drugs either before or during treatment. The significant heterogeneity among various tumors poses a critical challenge in modern cancer research, particularly in overcoming drug resistance. Copper, as an essential trace element in the body, participates in various biological processes of diseases, including cancers. The growth of many types of tumor cells exhibits a heightened dependence on copper. Thus, targeting copper metabolism or inducing cuproptosis may be potential ways to overcome cancer drug resistance. Copper chelators have shown potential in overcoming cancer drug resistance by targeting copper-dependent processes in cancer cells. In contrast, copper ionophores, copper-based nanomaterials, and other small molecules have been used to induce copper-dependent cell death (cuproptosis) in cancer cells, including drug-resistant tumor cells. This review summarizes the regulation of copper metabolism and cuproptosis in cancer cells and the role of copper metabolism and cuproptosis in cancer drug resistance, providing ideas for overcoming cancer resistance in the future.
Collapse
Affiliation(s)
- Leyi Yao
- Zhanjiang Institute of Clinical Medicine, Central People’s Hospital of Zhanjiang, Zhanjiang 524033, Guangdong, China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524033, Guangdong, China
- Authors contributed equally
| | - Baoyi Jiang
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
- Authors contributed equally
| | - Dacai Xu
- Zhanjiang Institute of Clinical Medicine, Central People’s Hospital of Zhanjiang, Zhanjiang 524033, Guangdong, China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524033, Guangdong, China
| |
Collapse
|
253
|
Zhou Z, Li X, Xiong M, He Y, Cheng X, Deng J, Li Y, Zhang X, Zhang Z, Zhou C, Yang X. Association between the dietary inflammatory index, bowel habits, and systemic serum inflammatory markers: insights from NHANES (2005-2010). Front Nutr 2025; 12:1543715. [PMID: 40206947 PMCID: PMC11978653 DOI: 10.3389/fnut.2025.1543715] [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: 12/12/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
Objective To examine the relationship between the Dietary Inflammatory Index (DII), abnormal bowel habits, and systemic serum inflammatory markers. Methods Data from 9,880 participants in the National Health and Nutrition Examination Survey (NHANES) 2005-2010 were analyzed. The DII was calculated from two 24-h dietary recalls. Bowel habits were assessed using the Bristol Stool Form Scale, and systemic inflammatory markers included AAPR, IBI, NLR, LMR, PNLR, LCR, LA, and PLR. Statistical analyses were performed using R, Zstats, and EmpowerStats to evaluate associations. Results Higher DII scores were positively associated with abnormal bowel habits, including constipation [β (95% CI): 0.11 (0.01-0.22)] and diarrhea [β (95% CI): 0.42 (0.32-0.53)], and with PNLR [β (95% CI): 0.01 (0.01-0.01)], PNLRQ4 [β (95% CI): 0.13 (0.05-0.20)], IBI [β (95% CI): 0.02 (0.01-0.02)], and IBIQ4 [β (95% CI): 0.33 (0.25-0.42)] (p < 0.05). Negative associations were found with AAPR [β (95% CI): -0.33 (-0.60 - -0.06)] and AAPRQ4 [β (95% CI): -0.18 (-0.34 - -0.01)], while no significant associations were observed with LA, LCR, or LMR. Subgroup analyses confirmed stable associations between DII and both chronic diarrhea and constipation across seven subgroups. Smoothed curve fitting revealed nonlinear relationships. A J-shaped association between DII and chronic constipation was identified in BMI and IBI subgroups. For BMI >30, the breakpoint (K) was 1.89, with ORs of 1.228 (95% CI: 1.097-1.375) below and 3.318 (95% CI: 1.531-7.191) above this point. In the IBI Q4 subgroup, the breakpoint was 1.96, with ORs of 1.145 (95% CI: 1.013-1.294) below and 5.794 (95% CI: 2.359-14.228) above. In the diarrhea group, a U-shaped association was observed in the AAPR Q4 population, with a breakpoint of -1.312 and ORs of 0.657 (95% CI: 0.478-0.901) below and 1.266 (95% CI: 1.057-1.518) above. Conclusion Higher DII scores are linked to an increased risk of chronic constipation and diarrhea and are associated with systemic inflammatory markers and factors such as BMI.
Collapse
Affiliation(s)
- Zeyang Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Xiangyong Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Mengya Xiong
- Department of Operating Room, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuee He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Xinmeng Cheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Jianbo Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Yanan Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Xiaoyang Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Zhengcao Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Chenxi Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Xiaodong Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
- National Center of Technology Innovation for Biopharmaceuticals, Suzhou, China
| |
Collapse
|
254
|
Lai G, Zhao X, Chen Y, Xie T, Su Z, Lin J, Chen Y, Chen K. The origin and polarization of Macrophages and their role in the formation of the Pre-Metastatic niche in osteosarcoma. Int Immunopharmacol 2025; 150:114260. [PMID: 39938167 DOI: 10.1016/j.intimp.2025.114260] [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: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Osteosarcoma, a primary malignant bone tumor commonly found in adolescents, is highly aggressive, with a high rate of disability and mortality. It has a profound negative impact on both the physical and psychological well-being of patients. The standard treatment approach, comprising surgery and chemotherapy, has seen little improvement in patient outcomes over the past several decades. Once relapse or metastasis occurs, prognosis worsens significantly. Therefore, there is an urgent need to explore new therapeutic approaches. In recent years, the successful application of immunotherapy in certain cancers has demonstrated its potential in the field of cancer treatment. Macrophages are the predominant components of the immune microenvironment in osteosarcoma and represent critical targets for immunotherapy. Macrophages exhibit dual characteristics; while they play a key role in maintaining tumor-promoting properties within the microenvironment, such as inflammation, angiogenesis, and immune suppression, they also possess antitumor potential as part of the innate immune system. A deeper understanding of macrophages and their relationship with osteosarcoma is essential for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Guisen Lai
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Xinyi Zhao
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanquan Chen
- Department of Orthopaedic Sun Yat-sen Memorial Hospital Sun Yat-sen University PR China
| | - Tianwei Xie
- The People's Hospital of Hezhou, No.150 Xiyue Street, Hezhou 542800 PR China
| | - Zepeng Su
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Jiajie Lin
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanhai Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Keng Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China.
| |
Collapse
|
255
|
Naldi L, Peri A, Fibbi B. Apelin/APJ: Another Player in the Cancer Biology Network. Int J Mol Sci 2025; 26:2986. [PMID: 40243599 PMCID: PMC11988549 DOI: 10.3390/ijms26072986] [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: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The apelinergic system exerts multiple biological activities in human pathologies, including cancer. Overactivation of apelin/APJ, which has been detected in many malignant tumors, and the strong correlation with progression-free and overall survival, suggested the role of an oncogene for the apelin gene. Emerging evidence sheds new light on the effects of apelin on cellular functions and homeostasis in cancer cells and supports a direct role for this pathway on different hallmarks of cancer: "sustaining proliferative signaling", "resisting cell death", "activating invasion and metastasis", "inducing/accessing vasculature", "reprogramming cellular metabolism", "avoiding immune destruction" and "tumor-promoting inflammation", and "enabling replicative immortality". This article reviews the currently available literature on the intracellular processes regulated by apelin/APJ, focusing on those pathways correlated with tumor development and progression. Furthermore, the association between the activity of the apelinergic axis and the resistance of cancer cells to oncologic treatments (chemotherapy, immunotherapy, radiation) suggests apelin/APJ as a possible target to potentiate traditional therapies, as well as to develop diagnostic and prognostic applications. This issue will be also covered in the review.
Collapse
Affiliation(s)
- Laura Naldi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Alessandro Peri
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Benedetta Fibbi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| |
Collapse
|
256
|
Mehta D, Dua C, Chakraborty R, Yadav P, Dasgupta U, Bajaj A. Docetaxel-conjugated bile acid-derived nanomicelles can inhibit tumour progression with reduced toxicity. NANOSCALE ADVANCES 2025; 7:2003-2010. [PMID: 39967859 PMCID: PMC11832056 DOI: 10.1039/d4na00715h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Docetaxel (DTX) is a highly effective chemotherapy drug commonly employed in the management of multiple cancers, such as breast, lung, and prostate cancer. However, its clinical usage is significantly hampered by its limited solubility, which limits its bioavailability, and its considerable toxic effects like neutropenia, neuropathy, and hypersensitive reactions. These limitations necessitate the development of innovative formulations to boost the therapeutic index of DTX. In this study, we aimed to enhance the tolerability and reduce the toxic effects of DTX by developing a novel hybrid scaffold (PIP-LCA-DTX), where we conjugated DTX to piperidine-derived lithocholic acid. This hybrid scaffold integrates the beneficial properties of bile acid-based drug conjugates and cationic amphiphiles to form stable and effective drug delivery systems. Our research demonstrates that PIP-LCA-DTX exhibits similar anticancer properties to DTX when tested against murine colon cancer (CT26) and melanoma (B16-F10) cell lines, indicating that the hybrid retains the therapeutic efficacy of the original drug. Our findings revealed that PIP-LCA-DTX forms stable nanomicelles (DTX-NMs) with an average hydrodynamic diameter of <150 nm and provides a promising delivery system by enhancing the solubility and stability of DTX. DTX-NMs showed significantly better tolerability and enhanced therapeutic efficacy (survival) compared to DTX alone. This improved tolerability, combined with the maintained therapeutic efficacy of DTX-NMs against murine cancer models, suggests that this hybrid scaffold could offer a more viable and safer option for cancer treatment.
Collapse
Affiliation(s)
- Devashish Mehta
- Amity Institute of Biotechnology, Amity University Haryana Panchgaon, Manesar Gurgaon 122413 Haryana India
| | - Chhavi Dua
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| | - Ruchira Chakraborty
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| | - Poonam Yadav
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| | - Ujjaini Dasgupta
- Amity Institute of Biotechnology, Amity University Haryana Panchgaon, Manesar Gurgaon 122413 Haryana India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| |
Collapse
|
257
|
Jiang H, Ye J. The Warburg effect: The hacked mitochondrial-nuclear communication in cancer. Semin Cancer Biol 2025; 112:93-111. [PMID: 40147702 DOI: 10.1016/j.semcancer.2025.03.006] [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: 07/31/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Mitochondrial-nuclear communication is vital for maintaining cellular homeostasis. This communication begins with mitochondria sensing environmental cues and transmitting signals to the nucleus through the retrograde cascade, involving metabolic signals such as substrates for epigenetic modifications, ATP and AMP levels, calcium flux, etc. These signals inform the nucleus about the cell's metabolic state, remodel epigenome and regulate gene expression, and modulate mitochondrial function and dynamics through the anterograde feedback cascade to control cell fate and physiology. Disruption of this communication can lead to cellular dysfunction and disease progression, particularly in cancer. The Warburg effect is the metabolic hallmark of cancer, characterized by disruption of mitochondrial respiration and increased lactate generation from glycolysis. This metabolic reprogramming rewires retrograde signaling, leading to epigenetic changes and dedifferentiation, further reprogramming mitochondrial function and promoting carcinogenesis. Understanding these processes and their link to tumorigenesis is crucial for uncovering tumorigenesis mechanisms. Therapeutic strategies targeting these disrupted pathways, including metabolic and epigenetic components, provide promising avenues for cancer treatment.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
258
|
Li YH, Zheng CR, Liu Y, Wang K, Zhou FF, Dong X, Yuan T, He QJ, Zhu H, Yang B. The role of calcium signaling in organotropic metastasis of cancer. Acta Pharmacol Sin 2025:10.1038/s41401-025-01537-3. [PMID: 40133629 DOI: 10.1038/s41401-025-01537-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025]
Abstract
Tumor metastasis is an important event in cancer progression, representing an enduring and irrevocable hallmark of cancers. The causes of tumor metastasis are complex and diverse. Arising evidence shows that the dysregulation of calcium signaling plays a crucial role in its initiation and progress. Calcium is an essential secondary messenger that regulates signaling pathways associated with tumor metastasis. The transient accumulation of calcium potentially promotes the advancement of tumor metastasis, while calcium-dependent proteins and calcium-related channels also significantly contribute to such malignant process. Thus, compounds specially targeting calcium channels, transporters or pumps may be therapeutic approaches prohibiting tumor metastasis. This review focuses on exploring the roles of calcium ions, calcium-dependent proteins and calcium-related channels in organotropic metastasis of cancer and its clinical applications in the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Yong-Hao Li
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chu-Run Zheng
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ke Wang
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 210000, China
| | - Fan-Fan Zhou
- Sydney Pharmacy School, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Xin Dong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310015, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310015, China
| | - Qiao-Jun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310015, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310015, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| |
Collapse
|
259
|
Alenezi A, Alhamad H, Alenezi A, Khan MU. Hypoxia Imaging in Lung Cancer: A PET-Based Narrative Review for Clinicians and Researchers. Pharmaceuticals (Basel) 2025; 18:459. [PMID: 40283896 PMCID: PMC12030053 DOI: 10.3390/ph18040459] [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: 03/01/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Hypoxia plays a critical role in lung cancer progression and treatment resistance by contributing to aggressive tumor behavior and poor therapeutic response. Molecular imaging, particularly positron emission tomography (PET), has become an essential tool for noninvasive hypoxia detection, providing valuable insights into tumor biology and aiding in personalized treatment strategies. Objective: This narrative review explores recent advancements in PET imaging for detecting hypoxia in lung cancer, with a focus on the development, characteristics, and clinical applications of various radiotracers. Findings: Numerous PET-based hypoxia radiotracers have been investigated, each with distinct pharmacokinetics and imaging capabilities. Established tracers such as 18F-Fluoromisonidazole (18F-FMISO) remain widely used, while newer alternatives like 18F-Fluoroazomycin Arabinoside (18F-FAZA) and 18F-Flortanidazole (18F-HX4) demonstrate improved clearance and image contrast. Additionally, 64Cu-ATSM has gained attention for its rapid tumor uptake and hypoxia selectivity. The integration of PET with hybrid imaging modalities, such as PET/CT and PET/MRI, enhances the spatial resolution and functional interpretation, making hypoxia imaging a promising approach for guiding radiotherapy, chemotherapy, and targeted therapies. Conclusions: PET imaging of hypoxia offers significant potential in lung cancer diagnosis, treatment planning, and therapeutic response assessment. However, challenges remain, including tracer specificity, quantification variability, and standardization of imaging protocols. Future research should focus on developing next-generation radiotracers with enhanced specificity, optimizing imaging methodologies, and leveraging multimodal approaches to improve clinical utility and patient outcomes.
Collapse
Affiliation(s)
- Ahmad Alenezi
- Radiologic Sciences Department, Kuwait University, Kuwait City 31470, Kuwait
| | - Hamad Alhamad
- Occupational Therapy Department, Kuwait University, Jabriya 31470, Kuwait
| | - Aishah Alenezi
- Radiologic Sciences Department, Kuwait University, Kuwait City 31470, Kuwait
| | - Muhammad Umar Khan
- Nuclear Medicine Department, Jahra Hospital, Ministry of Health, Al Jahra 03200, Kuwait
| |
Collapse
|
260
|
Pérez Escriva P, Correia Tavares Bernardino C, Letellier E. De-coding the complex role of microbial metabolites in cancer. Cell Rep 2025; 44:115358. [PMID: 40023841 DOI: 10.1016/j.celrep.2025.115358] [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: 08/09/2024] [Revised: 12/11/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
The human microbiome, an intricate ecosystem of trillions of microbes residing across various body sites, significantly influences cancer, a leading cause of morbidity and mortality worldwide. Recent studies have illuminated the microbiome's pivotal role in cancer development, either through direct cellular interactions or by secreting bioactive compounds such as metabolites. Microbial metabolites contribute to cancer initiation through mechanisms such as DNA damage, epithelial barrier dysfunction, and chronic inflammation. Furthermore, microbial metabolites exert dual roles on cancer progression and response to therapy by modulating cellular metabolism, gene expression, and signaling pathways. Understanding these complex interactions is vital for devising new therapeutic strategies. This review highlights microbial metabolites as promising targets for cancer prevention and treatment, emphasizing their impact on therapy responses and underscoring the need for further research into their roles in metastasis and therapy resistance.
Collapse
Affiliation(s)
- Pau Pérez Escriva
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catarina Correia Tavares Bernardino
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
261
|
Scerbo P, Tisserand B, Delagrange M, Debare H, Bensimon D, Ducos B. In vivo targeted and deterministic single-cell malignant transformation. eLife 2025; 13:RP97650. [PMID: 40130618 PMCID: PMC11936417 DOI: 10.7554/elife.97650] [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] [Indexed: 03/26/2025] Open
Abstract
Why does a normal cell possibly harboring genetic mutations in oncogene or tumor suppressor genes becomes malignant and develops a tumor is a subject of intense debate. Various theories have been proposed but their experimental test has been hampered by the unpredictable and improbable malignant transformation of single cells. Here, using an optogenetic approach we permanently turn on an oncogene (KRASG12V) in a single cell of a zebrafish brain that, only in synergy with the transient co-activation of a reprogramming factor (VENTX/NANOG/OCT4), undergoes a deterministic malignant transition and robustly and reproducibly develops within 6 days into a full-blown tumor. The controlled way in which a single cell can thus be manipulated to give rise to cancer lends support to the 'ground state theory of cancer initiation' through 'short-range dispersal' of the first malignant cells preceding tumor growth.
Collapse
Affiliation(s)
- Pierluigi Scerbo
- Laboratoire de Physique de l’Ecole Normale Supérieure LPENS, ENS, PSL Research University, CNRS, Sorbonne Université, Université de ParisParisFrance
- InovarionParisFrance
| | - Benjamin Tisserand
- Laboratoire de Physique de l’Ecole Normale Supérieure LPENS, ENS, PSL Research University, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Marine Delagrange
- Laboratoire de Physique de l’Ecole Normale Supérieure LPENS, ENS, PSL Research University, CNRS, Sorbonne Université, Université de ParisParisFrance
- High Throughput qPCR Core Facility of the ENS, Ecole Normale Supérieure, PSL Research University, IBENSParisFrance
| | - Héloise Debare
- Laboratoire de Physique de l’Ecole Normale Supérieure LPENS, ENS, PSL Research University, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - David Bensimon
- Laboratoire de Physique de l’Ecole Normale Supérieure LPENS, ENS, PSL Research University, CNRS, Sorbonne Université, Université de ParisParisFrance
- Dept. Chemistry and Biochemistry, UCLALos AngelesUnited States
| | - Bertrand Ducos
- Laboratoire de Physique de l’Ecole Normale Supérieure LPENS, ENS, PSL Research University, CNRS, Sorbonne Université, Université de ParisParisFrance
- High Throughput qPCR Core Facility of the ENS, Ecole Normale Supérieure, PSL Research University, IBENSParisFrance
| |
Collapse
|
262
|
Xiao H, Du X, Hou H, Guo W, Tao Z, Bao S, Wen Z, Jing N, Gao WQ, Dong B, Fang YX. ATOH8 confers the vulnerability of tumor cells to ferroptosis by repressing SCD expression. Cell Death Differ 2025:10.1038/s41418-025-01482-y. [PMID: 40133667 DOI: 10.1038/s41418-025-01482-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Emerging evidence indicates that transcriptional regulation plays pivotal roles in modulating cellular vulnerability to ferroptosis. However, the intricate mechanisms governing these processes remain poorly understood. In this study, we identify ATOH8, a basic helix-loop-helix (bHLH) transcription factor, as a key player in ferroptosis regulation. ATOH8 is significantly upregulated in tumor cells following treatment with a ferroptosis inducer. Overexpression of ATOH8 increases the susceptibility of tumor cells to ferroptosis, while deletion of ATOH8 promotes ferroptosis evasion. Mechanistically, ATOH8 confers the sensitivity of tumor cells to ferroptosis by suppressing the transcription of stearoyl-CoA desaturase (SCD). Additionally, another bHLH family member, TCF3, is found to functions as a co-factor with ATOH8 by forming a TCF3-ATOH8 transcriptional repressive complex that suppresses SCD transcription. Furthermore, searching for upstream element reveals that EZH2 epigenetically suppresses ATOH8 expression by promoting DNA methylation in the ATOH8 promoter region and increasing the level of H3K27 me3. Importantly, pharmacological inhibition of EZH2 in a combined with a ferroptosis inducer markedly impedes tumor growth both in vitro and in vivo. Collectively, our study elucidates a molecular link between ferroptosis and epigenetic and transcriptional regulation, highlighting the potential of EZH2 and ATOH8 as therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Huixiang Xiao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Hou
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Wenyun Guo
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenkeke Tao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shijia Bao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenzhen Wen
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Nan Jing
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yu-Xiang Fang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
263
|
Marques-Magalhães Â, Moreira-Silva F, Graça I, Dias PC, Correia MP, Alzamora MA, Henrique R, Lopez M, Arimondo PB, Miranda-Gonçalves V, Jerónimo C. Combination of MLo-1508 with sunitinib for the experimental treatment of papillary renal cell carcinoma. Front Oncol 2025; 15:1399956. [PMID: 40196736 PMCID: PMC11973455 DOI: 10.3389/fonc.2025.1399956] [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: 03/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Renal cell carcinoma (RCC) is the 14th most incident cancer worldwide, and no curative therapeutic options are available for advanced and metastatic disease. Hence, new treatment alternatives are urgently needed to tackle disease management and drug resistance. Herein, we explored the use of MLo-1508 as an anti-tumoral agent in RCC and further assessed its combination with sunitinib for the treatment of papillary RCC. For that, different RCC cell lines were treated with both drugs, alone or in combination, and different phenotypic assays were performed. Moreover, global DNA methylation levels and specific DNMT3a activity were measured, and gene-specific CpG methylation and transcript levels were quantified after treatment. Finally, the combinatory potential of MLo-1508 and sunitinib were asses both in vitro and in vivo using the ACHN cell line. We found that MLo-1508 significantly decreased RCC cell viability while inducing apoptosis in a dose-dependent manner without cytotoxicity for non-malignant cells. Moreover, the treatment induced morphometric alterations and DNA damage in all RCC cell lines. MLo-1508 decreased DNMT1 and DNMT3A transcript levels in 786-O and ACHN cells, inhibited DNMT3A activity, and reduced the global DNA methylation content of ACHN cells. When combined with sunitinib, a reduction in ACHN cell viability, as well as cell cycle arrest at G2/M was observed. Importantly, MLo-1508 decreased the sunitinib effective anti-tumoral concentration against ACHN cell viability. In an in vivo ACHN CAM model, the combination induced cell necrosis. Thus, MLo-1508 might improve sensitivity to sunitinib treatment by decreasing the required concentration and delaying resistance acquisition.
Collapse
Affiliation(s)
- Ângela Marques-Magalhães
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Inês Graça
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Paula C. Dias
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Margareta P. Correia
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Maria Ana Alzamora
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| | - Marie Lopez
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS-Université de Montpellier-ENSCM, Montpellier, France
| | - Paola B. Arimondo
- Epigenetic Chemical Biology, Institut Pasteur, UMR 3523CNRS, Paris, France
| | - Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
264
|
Rubiera-Valdés M, Corte-Torres MD, Navarro-López A, Blanco-Agudín N, Fernández-Menéndez S, Piña-Batista KM, Santos-Juanes J, Merayo-Lloves J, Quirós LM, Fernández-Velasco AA, Fernández-Vega I. PI3K and PINK1 Immunoexpression as Predictors of Survival in Patients Undergoing Resection of Brain Metastases from Lung Adenocarcinoma. Int J Mol Sci 2025; 26:2945. [PMID: 40243539 PMCID: PMC11988690 DOI: 10.3390/ijms26072945] [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: 02/26/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Phosphoinositide 3-kinase (PI3K) and PTEN-induced kinase 1 (PINK1) are key regulators of metabolism and mitochondrial quality control. This study assessed their immunoexpression in 22 patients with lung adenocarcinoma and resected brain metastases who underwent curative treatment between 2007 and 2017 and evaluated their prognostic significance. Tissue microarrays of primary tumors and matched metastases were analyzed using the H-score method. PI3K expression was significantly higher in primary tumors (96.8 ± 57.9 vs. 43.5 ± 62.3; p = 0.003) and in stage IV adenocarcinomas (113.3 ± 56.3 vs. 61.4 ± 47.1; p = 0.043). PINK1 expression showed no significant variation across disease stages. Univariate analysis identified older age (>55 years), PI3K overexpression (HR = 7.791, 95% CI 1.718-36.432; >50 points), and PINK1 overexpression (>100 points) in primary tumors as predictors of poor overall survival (HR = 2.236, 95% CI 1.109-4.508; p = 0.025). Multivariate analysis confirmed PINK1 overexpression in primary tumors as an independent prognostic factor (HR = 4.328, 95% CI 1.264-14.814; p = 0.020). These findings suggest that PI3K and PINK1 may serve as prognostic biomarkers in lung adenocarcinoma with resected brain metastases, emphasizing the need for research on their role in tumor progression and therapeutic response.
Collapse
Affiliation(s)
- Miriam Rubiera-Valdés
- Department of Pathology, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
| | - Mª Daniela Corte-Torres
- Biobank of Principality of Asturias (BioPA), 33011 Oviedo, Spain; (M.D.C.-T.); (A.N.-L.); (S.F.-M.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
| | - Andrea Navarro-López
- Biobank of Principality of Asturias (BioPA), 33011 Oviedo, Spain; (M.D.C.-T.); (A.N.-L.); (S.F.-M.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
| | - Noelia Blanco-Agudín
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, 33012 Oviedo, Spain
| | - Santiago Fernández-Menéndez
- Biobank of Principality of Asturias (BioPA), 33011 Oviedo, Spain; (M.D.C.-T.); (A.N.-L.); (S.F.-M.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
- Department of Neurology, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain
| | - Kelvin M. Piña-Batista
- Department of Neurosurgery, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
| | - Jorge Santos-Juanes
- Department of Dermatology, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
| | - Jesús Merayo-Lloves
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, 33012 Oviedo, Spain
- Department of Surgery and Medical-Surgical Specialties, University of Oviedo, 33006 Oviedo, Spain
| | - Luis M. Quirós
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, 33012 Oviedo, Spain
| | - Adela A. Fernández-Velasco
- Department of Pathology, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
- Biobank of Principality of Asturias (BioPA), 33011 Oviedo, Spain; (M.D.C.-T.); (A.N.-L.); (S.F.-M.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
- Department of Surgery and Medical-Surgical Specialties, University of Oviedo, 33006 Oviedo, Spain
| | - Iván Fernández-Vega
- Department of Pathology, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
- Biobank of Principality of Asturias (BioPA), 33011 Oviedo, Spain; (M.D.C.-T.); (A.N.-L.); (S.F.-M.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain; (N.B.-A.); (J.M.-L.); (L.M.Q.)
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, 33012 Oviedo, Spain
- Department of Surgery and Medical-Surgical Specialties, University of Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
265
|
Thomsen MT, Busk M, Zhang D, Chiu CL, Zhao H, Garcia-Marques FJ, Bermudez A, Pitteri S, Borre M, Brooks JD, Nyengaard JR. The olfactory receptor OR51E2 regulates prostate cancer aggressiveness and modulates STAT3 in prostate cancer cells and in xenograft tumors. BMC Cancer 2025; 25:535. [PMID: 40128715 PMCID: PMC11934788 DOI: 10.1186/s12885-025-13928-0] [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: 08/30/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Despite advancements in the detection and treatment of prostate cancer, the molecular mechanisms underlying its progression remain unclear. This study aimed to investigate the role of the receptor OR51E2, which is commonly upregulated in prostate cancer, in the progression of this disease. METHODS We investigated the physiological effects of OR51E2 through CRISPR-Cas9-induced monoclonal OR51E2 knockout. We assessed in vitro and in vivo tumorigenicity and conducted transcriptomic and proteomic analyses of xenograft tumors derived from these knockout cells. Furthermore, we analyzed the effects of differences in OR51E2-expression levels in patients from a TCGA cohort. RESULTS OR51E2-knockout cells exhibited increased proliferation, migration, adhesion, anchorage-independent colony formation, and tumor growth rates, resulting in a more aggressive cancer phenotype. Omics analyses revealed several potential pathways associated with significant molecular changes, notably an aberration in the STAT3 pathway linked to IL-6 signaling, highlighting a connection to inflammatory pathways. TCGA cohort analysis revealed that prostate cancer patients with low tumor OR51E2 expression had a worse prognosis and a higher average Gleason grade than those with higher expression levels. Additionally, this analysis supported the putative OR51E2-related modulation of the STAT3 pathway. CONCLUSIONS OR51E2 is regulated throughout prostate cancer progression and actively influences cancer cell physiology affecting cancer aggressiveness. Reduced OR51E2 expression may adversely affect patient outcomes, potentially through alterations in the STAT3 pathway that impact cellular responses to inflammatory signaling.
Collapse
Affiliation(s)
- Mikkel Thy Thomsen
- Core Center for Molecular Morphology, Department of Clinical Medicine, Aarhus University, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, Aarhus N C113 8200, Denmark.
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Morten Busk
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fernando Jose Garcia-Marques
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Abel Bermudez
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sharon Pitteri
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Borre
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
- Department of Urology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jens Randel Nyengaard
- Core Center for Molecular Morphology, Department of Clinical Medicine, Aarhus University, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, Aarhus N C113 8200, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
266
|
Jing F, Mu J, Liu J, Hu C, Wu F, Gao Q. Senescent vascular endothelial cells promote oral squamous cell carcinoma progression through complement C3 activation. Arch Oral Biol 2025; 174:106242. [PMID: 40158302 DOI: 10.1016/j.archoralbio.2025.106242] [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: 01/11/2025] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE The tumour microenvironment (TME) plays a critical role in therapeutic response and clinical outcomes in cancer. Senescent stromal cells have been shown to promote tumour progression; however, the role of senescent vascular endothelial cells (VECs) in oral squamous cell carcinoma (OSCC) remains largely unknown. In this study, we aimed to explore the effects and potential mechanisms of senescent VECs in OSCC progression. DESIGN Cisplatin was used to induce senescence in two endothelial cell lines. Senescence-associated β-galactosidase (SA-β-gal) staining, immunoblotting, cell cycle and proliferation assays, and migration and invasion assays were performed to access senescence development and biological behavior. Additionally, RNA sequencing analysis, multiplex immunohistochemical staining, immunoblotting, and xenograft mouse models were used to investigate the senescence-associated secretory phenotype of senescent VECs during OSCC progression and its potential molecular mechanisms. RESULTS Cisplatin-induced senescent VECs exhibited senescence-related changes, including positive SA-β-gal expression and upregulation of p16, p21, and p53, along with attenuated proliferation and migration. Notably, cisplatin-induced VEC senescence promoted OSCC cell proliferation, migration, and invasion by activating complement C3. Increased gene and protein levels of C3 were observed in cisplatin-treated senescent VECs. Inhibition of C3 in vitro and in vivo reduced OSCC cell proliferation and invasion. CONCLUSION Senescent VECs induced by cisplatin promote OSCC proliferation and invasion through complement C3 activation. Targeting complement C3 in senescent VECs may offer a novel therapeutic strategy for OSCC treatment.
Collapse
Affiliation(s)
- Fangqi Jing
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Can Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fanglong Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Qinghong Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
267
|
Wu J, Li R, Wang J, Zhu H, Ma Y, You C, Shu K. Reactive Astrocytes in Glioma: Emerging Opportunities and Challenges. Int J Mol Sci 2025; 26:2907. [PMID: 40243478 PMCID: PMC11989224 DOI: 10.3390/ijms26072907] [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: 02/14/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Gliomas are the most prevalent malignant tumors in the adult central nervous system (CNS). Glioblastoma (GBM) accounts for approximately 60-70% of primary gliomas. It is a great challenge to human health because of its high degree of malignancy, rapid progression, short survival time, and susceptibility to recurrence. Owing to the specificity of the CNS, the glioma microenvironment often contains numerous glial cells. Astrocytes are most widely distributed in the human brain and form reactive astrocyte proliferation regions around glioma tissue. In addition, astrocytes are activated under pathological conditions and regulate tumor and microenvironmental cells through cell-to-cell contact or the secretion of active substances. Therefore, astrocytes have attracted attention as important components of the glioma microenvironment. Here, we focus on the mechanisms of reactive astrocyte activation under glioma conditions, their contribution to the mechanisms of glioma genesis and progression, and their potential value as targets for clinical intervention in gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| |
Collapse
|
268
|
Eduardo MB, Cottone G, McCloskey CW, Liu S, Palma FR, Zappia MP, Islam AB, Gao P, Setya J, Dennis S, Gao H, Zhang Q, Xuei X, Luo Y, Locasale J, Bonini MG, Khokha R, Frolov MV, Benevolenskaya EV, Chandel NS, Khan SA, Clare SE. A metabolic shift to the serine pathway induced by lipids fosters epigenetic reprogramming in nontransformed breast cells. SCIENCE ADVANCES 2025; 11:eads9182. [PMID: 40117373 PMCID: PMC11927636 DOI: 10.1126/sciadv.ads9182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025]
Abstract
Lipid metabolism and the serine, one-carbon, glycine (SOG) and methionine pathways are independently and significantly correlated with estrogen receptor-negative breast cancer (ERneg BC). Here, we propose a link between lipid metabolism and ERneg BC through phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in the de novo serine pathway. We demonstrate that the metabolism of the paradigmatic medium-chain fatty acid octanoic acid leads to a metabolic shift toward the SOG and methionine pathways. PHGDH plays a role in both the forward direction, contributing to the production of S-adenosylmethionine, and the reverse direction, generating the oncometabolite 2-hydroxyglutarate, leading to epigenomic reprogramming and phenotypic plasticity. The methionine cycle is closely linked to the transsulfuration pathway. Consequently, we observe that the shift increases the antioxidant glutathione, which mitigates reactive oxygen species (ROS), enabling survival of a subset of cells that have undergone DNA damage. These metabolic changes contribute to several hallmarks of cancer.
Collapse
Affiliation(s)
| | - Gannon Cottone
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Curtis W. McCloskey
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shiyu Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Flavio R. Palma
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maria Paula Zappia
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Abul B.M.M.K. Islam
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joel Setya
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Saya Dennis
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Hongyu Gao
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qian Zhang
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaoling Xuei
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Jason Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Marcelo G. Bonini
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Maxim V. Frolov
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Elizaveta V. Benevolenskaya
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Navdeep S. Chandel
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Seema A. Khan
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Susan E. Clare
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
269
|
Wu Y, Qu Z, Wu Z, Zhuang J, Wang Y, Wang Z, Chu J, Qi Q, Han S. Multiple primary malignancies and gut microbiome. BMC Cancer 2025; 25:516. [PMID: 40114168 PMCID: PMC11927136 DOI: 10.1186/s12885-025-13894-7] [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: 02/03/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Multiple primary malignancies (MPM) are two or more independent primary malignancies. Recently, the relationship between microbiome and various tumors has been gradually focused on. OBJECTIVE To describe the relationship between MPM patients (MPMs) and gut microbiome. METHODS A total of 27 MPMs, 30 colorectal cancer patients (CRCs), and 30 healthy individuals were included to obtain metagenomic sequencing data. The knowledge graphs of gut bacteria and enteroviruses were plotted based on metagenomics. Wilcoxon rank-sum test was used to screen the characteristic gut microbiome. RESULTS The knowledge graph of gut microbiome in MPM patients was plotted. A total of 26 different gut bacteria, including Dialister, Fecalibacterium and Mediterraneibacter, were found between MPMs and healthy individuals. Twenty gut bacteria, including Parvimonas, Dialister and Mediterraneibacter, were more abundant in MPM complicated by CRC compared with CRCs. Twenty-one different enterovirus, including Triavirus, Punavirus and Lilyvirus, were screened between MPMs and healthy individuals. Triavirus, Punavirus and Lilyvirus were less abundant in MPM than healthy individuals. The abundance of Triavirus, Punavirus and Lilyvirus in CRC patients were also lower than MPM complicated by CRC patients. CONCLUSION The knowledge graph of gut microbiome in MPM patients was plotted. It may provide basic data support for future research of MPM.
Collapse
Affiliation(s)
- Yinhang Wu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China
| | - Zhanbo Qu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China
| | - Zheng Wu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China
| | - Jing Zhuang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China
| | - Yingchen Wang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China
| | - Zefeng Wang
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France
- Huzhou University, Huzhou, China
| | - Jian Chu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China
| | - Quan Qi
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China.
| | - Shuwen Han
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, Huzhou, China.
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France.
| |
Collapse
|
270
|
Santos-Sousa DC, da Rosa S, Filippi-Chiela E. Molecular signatures of cellular senescence in cancer: a critical review of prognostic implications and therapeutic opportunities. Mech Ageing Dev 2025; 225:112052. [PMID: 40120861 DOI: 10.1016/j.mad.2025.112052] [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: 12/16/2024] [Revised: 03/01/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Cellular senescence is a state of permanent loss of proliferative capacity. Therefore, cells that reach a senescent state prevent tumor initiation, acting as an anti-tumor mechanism. However, despite not being proliferative, senescent cells have high secretory activity, constituting the Senescence-Associated Secretory Phenotype (SASP). SASP includes thousands of soluble molecules and extracellular vesicles, through which senescent cells can affect other cells and the extracellular matrix. In advanced tumors, the enrichment of senescent cells can have anti- or pro-tumor effects depending on features like SASP composition, tumor microenvironment (TME) composition, the anatomic site, histopathologic characteristics of malignancy, and tumor molecular background. We reviewed the studies assessing the impact of the senescence status, measured by mRNA or lncRNA molecular signatures, in the prognosis and other clinically relevant information in cancer, including anti-tumor immunity and response to therapy. We discussed the pros and cons of different strategies to define those molecular signatures and the main limitations of the studies. Finally, we also raised clinical challenges regarding the crossroad between cellular senescence and cancer prognosis, including some therapeutic opportunities in the field.
Collapse
Affiliation(s)
- Débora C Santos-Sousa
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil.
| | - Solon da Rosa
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil.
| | - Eduardo Filippi-Chiela
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil; Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil.
| |
Collapse
|
271
|
Peng Z, Ayad MA, Jing Y, Chou T, Cooper LA, Goldstein JA. Benchmarking pathology foundation models for non-neoplastic pathology in the placenta. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.19.25324282. [PMID: 40166578 PMCID: PMC11957174 DOI: 10.1101/2025.03.19.25324282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Machine learning (ML) applications within diagnostic histopathology have been extremely successful. While many successful models have been built using general-purpose models trained largely on everyday objects, there is a recent trend toward pathology-specific foundation models, trained using histopathology images. Pathology foundation models show strong performance on cancer detection and subtyping, grading, and predicting molecular diagnoses. However, we have noticed lacunae in the testing of foundation models. Nearly all the benchmarks used to test them are focused on cancer. Neoplasia is an important pathologic mechanism and key concern in much of clinical pathology, but it represents one of many pathologic bases of disease. Non-neoplastic pathology dominates findings in the placenta, a critical organ in human development, as well as a specimen commonly encountered in clinical practice. Very little to none of the data used in training pathology foundation models is placenta. Thus, placental pathology is doubly out of distribution, representing a useful challenge for foundation models. We developed benchmarks for estimation of gestational age, classifying normal tissue, identifying inflammation in the umbilical cord and membranes, and in classification of macroscopic lesions including villous infarction, intervillous thrombus, and perivillous fibrin deposition. We tested 5 pathology foundation models and 4 non-pathology models for each benchmark in tasks including zero-shot K-nearest neighbor classification and regression, content-based image retrieval, supervised regression, and whole-slide attention-based multiple instance learning. In each task, the best performing model was a pathology foundation model. However, the gap between pathology and non-pathology models was diminished in tasks related to inflammation or those in which a supervised task was performed using model embeddings. Performance was comparable among pathology foundation models. Among non-pathology models, ResNet consistently performed worse, while models from the present decade showed better performance. Future work could examine the impact of incorporating placental data into foundation model training.
Collapse
Affiliation(s)
| | | | | | | | | | - Jeffery A. Goldstein
- Corresponding author: Jeffery A. Goldstein, MD, PhD, Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Ward 3-140, Chicago IL, 60611,
| |
Collapse
|
272
|
Cao L, Li T, Xu X, Sun M, Teng W, Zhu M. Texasin, A main product from Caragana Jubata (Pall.) Poir, induces proliferation arrest and protective autophagy in lung adenocarcinoma. BMC Cancer 2025; 25:513. [PMID: 40114111 PMCID: PMC11924753 DOI: 10.1186/s12885-025-13933-3] [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: 11/18/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Lung cancer, a leading cause of mortality worldwide, necessitates effective therapeutic strategies. Caragana jubata, a traditional Chinese medicinal plant, harbors Texasin, a potential anti-tumor agent. This study aimed to evaluate the anti-cancer effects of Texasin on lung cancer cells, while assessing its impact on normal lung cells. METHODS The study utilized cell lines H1299 and A549, alongside normal lung embryonic cells, to investigate Texasin's effects through Cell Counting Kit-8, Transwell, and wound healing assays. Transcriptome sequencing and analysis were performed to identify potential mechanisms. β-galactosidase activity and Retinoblastoma(RB) protein expression were assessed, and autophagy and apoptosis were explored through chloroquine co-treatment. Mice bearing H1299 cell-derived tumors were treated with Texasin. Tumor changes were assessed through in vivo imaging, and autophagy levels within the tumors were analyzed. RESULTS Texasin inhibited lung cancer cell proliferation, migration, and invasion without harming normal cells. It promoted cell senescence, arrested the cell cycle in G1 phase, and upregulated β-galactosidase and RB protein expression. Texasin induced protective autophagy, which could be converted to apoptosis by chloroquine co-treatment. Texasin inhibits the proliferation of lung adenocarcinoma cells in vivo, as evidenced by immunohistochemistry showing an increase in autophagy levels within the tumors. CONCLUSIONS Texasin emerges as a promising non-cytotoxic anti-lung adenocarcinoma cancer compound, significantly inhibiting malignant phenotypes, highlighting its potential for lung adenocarcinoma cancer therapy.
Collapse
Affiliation(s)
- Liuzhao Cao
- Department of pulmonary and critical care medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Tiantian Li
- Department of Ultrasound, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Xingxiang Xu
- Department of pulmonary and critical care medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Mei Sun
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Weiyun Teng
- Department of pulmonary and critical care medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China.
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China.
| | - Miao Zhu
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China.
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China.
| |
Collapse
|
273
|
Gao X, Shen X, Huang S, Huang S. Classification of lung adenocarcinoma based on senescence-related genes identifies a cluster with immunotherapy resistance and poor prognosis. Discov Oncol 2025; 16:363. [PMID: 40111675 PMCID: PMC11925839 DOI: 10.1007/s12672-025-02127-9] [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: 09/09/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Lung adenocarcinoma is one of the major contributors to cancer-related mortality, with immunotherapy emerging as a key treatment. However, many patients exhibit resistance to immune checkpoint inhibitors. Cellular senescence has been linked to tumor progression and drug resistance, influencing the tumor microenvironment. This study applied consensus clustering to classify lung adenocarcinoma patients into two clusters based on senescence-related gene expression, revealing differing immune characteristics. One of the identified clusters exhibited immunosuppressive characteristics and showed resistance to immunotherapy. A senescence-related risk score was developed using machine learning to predict immunotherapy response and prognosis. High senescence-related risk score correlated with poorer survival and increased immunotherapy resistance across multiple cancer types. The senescence-related risk score model showed robust predictive ability in both the training and validation cohorts. These findings suggest a link between senescence and immunotherapy resistance, and further investigation into their relationship could reveal new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Xinrui Gao
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiang Shen
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shasha Huang
- Department of Ultrasound, Zhongjiang People's Hospital, Deyang, Sichuan, China.
| | - Shangke Huang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
274
|
Zhang X, Gao Y, Zhang S, Wang Y, Du Y, Hao S, Ni T. The Regulation of Cellular Senescence in Cancer. Biomolecules 2025; 15:448. [PMID: 40149983 PMCID: PMC11940315 DOI: 10.3390/biom15030448] [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] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Cellular senescence is a stable state of cell cycle arrest caused by telomere shortening or various stresses. After senescence, cells cease dividing and exhibit many age-related characteristics. Unlike the halted proliferation of senescence cells, cancer cells are considered to have unlimited growth potential. When cells display senescence-related features, such as telomere loss or stem cell failure, they can inhibit tumor development. Therefore, inducing cells to enter a senescence state can serve as a barrier to tumor cell development. However, many recent studies have found that sustained senescence of tumor cells or normal cells under certain circumstances can exert environment-dependent effects of tumor promotion and inhibition by producing various cytokines. In this review, we first introduce the causes and characteristics of induced cellular senescence, analyze the senescence process of immune cells and cancer cells, and then discuss the dual regulatory role of cell senescence on tumor growth and senescence-induced therapies targeting cancer cells. Finally, we discuss the role of senescence in tumor progression and treatment opportunities, and propose further studies on cellular senescence and cancer therapy.
Collapse
Affiliation(s)
- Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Siyu Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China;
| | - Yixiong Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| |
Collapse
|
275
|
Domingues ACM, de Oliveira SB, Tessarollo NG, Lepique AP, Rodrigues O, Sharova T, Lawless A, Li D, Basnet M, Boland GM, Cohen S, Jenkins RW, Strauss BE. Use of patient-derived organotypic tumor spheroids for testing of viral vector gene therapy in combination with checkpoint blockade. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200942. [PMID: 40034966 PMCID: PMC11874566 DOI: 10.1016/j.omton.2025.200942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
Checkpoint inhibitors have revolutionized cancer treatment, but a significant proportion of patients do not respond to these therapies, underscoring the need for alternative strategies. Although gene therapy has made substantial strides, its application in solid tumors remains underexplored, with limited treatments approved. Here, we further investigated a gene therapy approach with non-replicating adenoviral vectors encoding the alternate reading frame (ARF) and interferon beta (IFNb) and tested it in a clinically relevant setting. We previously showed that this combined gene therapy induces immunogenic cell death in melanoma models, and now, we utilize patient-derived organotypic tumor spheroids (PDOTS), a model that closely recapitulates the immune environment of tumors, to test its effects using patient tumors. Our results demonstrate, for the first time, the effectiveness of using PDOTS to evaluate viral-vector-based gene therapies. While the addition of anti-PD-1 did not enhance therapeutic outcomes, the gene therapy alone suppressed tumor growth and triggered antitumor immune responses across different cancer models, notably those with low immunogenicity and specific genome profiles. These findings suggest that this gene therapy could serve as a valuable alternative for patients not responsive to checkpoint inhibitors and who have solid tumors with limited treatment and impaired p53-ARF-MDM2 pathways, such as liposarcomas.
Collapse
Affiliation(s)
- Ana Carolina M. Domingues
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Soraia B. de Oliveira
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Nayara G. Tessarollo
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Ana Paula Lepique
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
- Department of Immunology, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Otavio Rodrigues
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Tatyana Sharova
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Aleigha Lawless
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David Li
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Madak Basnet
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Genevieve M. Boland
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sonia Cohen
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Russell W. Jenkins
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bryan E. Strauss
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
276
|
Chandrasekaran J, Gopal D, Sureshkumar LV, Santhiyagu IX, Senthil Kumar V, Munuswamy B, Gani BFHMY, Agrawal M. Integrated AI and machine learning pipeline identifies novel WEE1 kinase inhibitors for targeted cancer therapy. Mol Divers 2025:10.1007/s11030-025-11157-y. [PMID: 40106128 DOI: 10.1007/s11030-025-11157-y] [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: 01/28/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
The dysregulation of the cell cycle in cancer underscores the therapeutic potential of targeting WEE1 kinase, a key regulator of the G2/M checkpoint. This study harnessed artificial intelligence (AI)-driven methodologies, particularly the MORLD platform, to identify novel WEE1 inhibitors. Starting with clinically validated WEE1 inhibitors as references, we generated 20,000 structurally diverse compounds optimized for binding affinity, synthetic accessibility, and drug-likeness. A rigorous cheminformatics pipeline-comprising PAINS filtering, physicochemical property assessments, and molecular fingerprinting-refined this library to 242 promising candidates. Dimensionality reduction using UMAP and clustering via K-means enabled the prioritization of structurally unique leads. Molecular docking studies highlighted two compounds, MORLD5036 and MORLD6305, with exceptional binding affinities and interactions with key WEE1 active site residues. Molecular dynamics simulations and MM-GBSA binding free energy calculations further validated MORLD5036 as the most stable and potent inhibitor. Scaffold analysis revealed novel chemotypes distinct from existing inhibitors, enhancing potential for intellectual property. Comprehensive ADME profiling confirmed favorable pharmacokinetics, while synthetic accessibility evaluations indicated practicality for experimental validation. The identified lead compound, MORLD5036, exhibits favorable pharmacokinetics and novel chemotypes, positioning it as a potential therapeutic candidate for cancers reliant on WEE1-mediated cell cycle control. This integrated, AI-driven pipeline expedites the identification of next-generation WEE1 inhibitors, paving the way for advancements in precision oncology. Unlike traditional methods reliant on pre-existing datasets, this study leverages MORLD's reinforcement learning framework to autonomously generate inhibitors, enabling exploration of uncharted chemical space. These findings establish MORLD5036 as a computationally promising WEE1 inhibitor candidate warranting further experimental validation.
Collapse
Affiliation(s)
- Jaikanth Chandrasekaran
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India.
| | - Dhanushya Gopal
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Lokesh Vishwa Sureshkumar
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Infant Xavier Santhiyagu
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Varsha Senthil Kumar
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Bhuvaneshwari Munuswamy
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Beevi Fathima Harshatha Mohamed Yousuf Gani
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, 600116, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram, Haryana, India
| |
Collapse
|
277
|
Wei W, Dang Y, Chen G, Han C, Zhang S, Zhu Z, Bie X, Xue J. Comprehensive analysis of senescence-related genes identifies prognostic clusters with distinct characteristics in glioma. Sci Rep 2025; 15:9540. [PMID: 40108265 PMCID: PMC11923138 DOI: 10.1038/s41598-025-93482-8] [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: 08/17/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
Cellular senescence, defined as a state of permanent arrest in cell growth, is regarded as a crucial tumor suppression mechanism. However, accumulating scientific evidence suggests that senescent cells play a detrimental role in the progression of cancer. Unfortunately, the current lack of reliable markers that specifically reflect the level of senescence in cancer greatly hinders our in-depth understanding of this important biological foundation. Therefore, the search for more specific and reliable markers to reveal the specific role of senescent cells in cancer progression is particularly urgent and important. To uncover the role of senescence in gliomas, we collected senescence-related genes for integrated analysis. Consensus clustering was used to subtype gliomas based on the senescence gene set, and we identified two robust prognostic clusters of gliomas with distinct survival outcomes, multi-omics landscapes, immune characteristics, and differential drug responses. Multiple external datasets were used to validate the stability of our subtypes. Various computational and experimental methods, including WGCNA (Weighted Gene Co-expression Network Analysis), ssGSEA (single-sample Gene Set Enrichment Analysis), and machine learning algorithms (lasso regression, support vector machines, random forests), were employed for analysis. We found that CEBPB and LMNA are associated with poor prognosis in gliomas and may mediate immunosuppression and tumor proliferation. Drug prediction indicated that dasatinib is a potential therapeutic agent. Our findings provide insights into the role of the senescence gene set in patient stratification and precision medicine.
Collapse
Affiliation(s)
- Wenyuan Wei
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ying Dang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Gang Chen
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Chao Han
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Siwei Zhang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ziqiang Zhu
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Xiaohua Bie
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| | - Jungang Xue
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
278
|
Wijayasiri P, Astbury S, Needham G, Kaye P, Bhat M, Piccinini AM, Aravinthan AD. Role of hepatocellular senescence in the development of hepatocellular carcinoma and the potential for therapeutic manipulation. Hum Cell 2025; 38:70. [PMID: 40100482 PMCID: PMC11920335 DOI: 10.1007/s13577-025-01201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Accumulation of senescent hepatocytes is universal in chronic liver disease (CLD). This study investigates an association between hepatocyte senescence and hepatocellular carcinoma (HCC) and explores the therapeutic role of sirolimus. Background liver biopsies from 15 patients with cirrhosis and HCC and 45 patients with cirrhosis were stained for p16, a marker of cell senescence. STAM™ mice were randomized into 3 groups of 5 at 4 weeks of age and administered vehicle ± sirolimus intraperitoneally, thrice weekly, from 4 to 18 weeks of age. Placebo group was an administered vehicle, early sirolimus group was an administered vehicle with sirolimus, late sirolimus group was an administered vehicle from 4 to 12 weeks then vehicle with sirolimus from 12 to 18 weeks. The primary outcome was HCC nodule development. Senescent hepatocyte burden and senescence-associated secretory phenotype (SASP) factors were assessed in mice livers. In the human study, age (OR 1.282, 95% CI 1.086-1.513, p = 0.003) and p16 (OR 1.429, 95% CI 1.112-1.838, p = 0.005) were independently associated with HCC. In the animal study, all three groups exhibited similar MASLD activity scores (p = 0.39) and fibrosis area (p = 0.92). The number and the maximum diameter of HCC nodules were significantly lower in the early sirolimus group compared to placebo and late sirolimus group. The gene expression of SASP factors was similar in all groups. Protein levels of some SASP factors (TNFα, IL1β, IL-2, CXCL15) were significantly lower in sirolimus administered groups compared to placebo group. The study demonstrates an independent association between senescent hepatocyte burden and HCC. It indicates a potential chemoprophylactic role for sirolimus through SASP factor inhibition. These early results could inform a future human clinical trial.
Collapse
Affiliation(s)
- Pramudi Wijayasiri
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, E Floor, West Block, QMC Campus, Derby Road, Nottingham, NG7 2UH, UK
| | - Stuart Astbury
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, E Floor, West Block, QMC Campus, Derby Road, Nottingham, NG7 2UH, UK
| | - Grace Needham
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Philip Kaye
- Department of Pathology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Mamatha Bhat
- Multiorgan Transplant Program, Toronto General Hospital, University of Toronto, Toronto, Canada
| | | | - Aloysious D Aravinthan
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, E Floor, West Block, QMC Campus, Derby Road, Nottingham, NG7 2UH, UK.
| |
Collapse
|
279
|
Ramchatesingh B, Martinez Villarreal A, Lefrançois P, Gantchev J, Sivachandran S, Abou Setah S, Litvinov IV. Targeting PRAME directly or via EZH2 inhibition overcomes retinoid resistance and represents a novel therapy for keratinocyte carcinoma. Mol Oncol 2025. [PMID: 40101298 DOI: 10.1002/1878-0261.13820] [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: 10/22/2024] [Revised: 12/11/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025] Open
Abstract
Retinoids have demonstrated efficacy as preventative/treatment agents for keratinocyte carcinomas (KCs): basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (SCC). However, retinoid resistance mechanisms limit the efficacy of these compounds. A subset of KCs expresses Preferentially Expressed Antigen in Melanoma (PRAME): a retinoid signaling corepressor. PRAME is proposed to repress retinoid signaling by guiding enhancer of zeste homolog 2 (EZH2) to retinoic acid response elements (RARE) in promoters. We investigated the effects of PRAME on KC pathogenesis and retinoid response. High-PRAME expression in tumors was negatively correlated with epidermal differentiation gene signatures. PRAME overexpression downregulated epidermal differentiation gene signatures and impaired differentiation in 3D culture. PRAME overexpression attenuated retinoid-induced RARE activation, growth suppression, and differentiation responses. Conversely, low-PRAME tumors and PRAME-depleted KC cells demonstrated enriched epidermal differentiation gene signatures. PRAME downregulation restored retinoid-induced RARE activation, growth suppression, keratinization in SCC, and cell death signaling in BCC. Furthermore, combined retinoid and EZH2 inhibitor treatment augmented RARE activation and suppressed PRAME-expressing KC cell growth. Hence, PRAME confers retinoid resistance in KC, which may be overcome by EZH2 inhibition.
Collapse
Affiliation(s)
- Brandon Ramchatesingh
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Amelia Martinez Villarreal
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Philippe Lefrançois
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Division of Dermatology, McGill University Health Center, Montreal, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University Montreal, Canada
| | - Jennifer Gantchev
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Sriraam Sivachandran
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Samy Abou Setah
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Ivan V Litvinov
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Division of Dermatology, McGill University Health Center, Montreal, Canada
| |
Collapse
|
280
|
Damiano OM, Stevens AJ, Kenwright DN, Seddon AR. Chronic Inflammation to Cancer: The Impact of Oxidative Stress on DNA Methylation. FRONT BIOSCI-LANDMRK 2025; 30:26142. [PMID: 40152377 DOI: 10.31083/fbl26142] [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: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025]
Abstract
The genomic landscape of cancer cells is complex and heterogeneous, with aberrant DNA methylation being a common observation. Growing evidence indicates that oxidants produced from immune cells may interact with epigenetic processes, and this may represent a mechanism for the initiation of altered epigenetic patterns observed in both precancerous and cancerous cells. Around 20% of cancers are linked to chronic inflammatory conditions, yet the precise mechanisms connecting inflammation with cancer progression remain unclear. During chronic inflammation, immune cells release oxidants in response to stimuli, which, in high concentrations, can cause cytotoxic effects. Oxidants are known to damage DNA and proteins and disrupt normal signalling pathways, potentially initiating a sequence of events that drives carcinogenesis. While research on the impact of immune cell-derived oxidants on DNA methylation remains limited, this mechanism may represent a crucial link between chronic inflammation and cancer development. This review examines current evidence on inflammation-associated DNA methylation changes in cancers related to chronic inflammation.
Collapse
Affiliation(s)
- Olivia M Damiano
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Aaron J Stevens
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Diane N Kenwright
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Annika R Seddon
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, 8011 Christchurch, New Zealand
| |
Collapse
|
281
|
Garibaldi-Ríos AF, Figuera LE, Zúñiga-González GM, Gómez-Meda BC, Puebla-Pérez AM, Rivera-Cameras A, Magaña-Torres MT, García-Ortíz JE, Dávalos-Rodríguez IP, Rosales-Reynoso MA, García-Verdín PM, Carrillo-Dávila IA, Torres-Mendoza BM, Ávalos-Navarro G, Gallegos-Arreola MP. Genomic Landscape of Breast Cancer: Study Across Diverse Ethnic Groups. Diseases 2025; 13:86. [PMID: 40136626 PMCID: PMC11941751 DOI: 10.3390/diseases13030086] [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: 02/06/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Breast cancer (BC) is the most common cancer among women worldwide, with incidence and mortality rates varying across ethnic groups due to sociodemographic, clinicopathological, and genomic differences. This study aimed to characterize the genomic landscape of BC in diverse ethnic groups using computational tools to explore these variations. Methodology: cBioPortal was used to analyze genomic, clinicopathological, and sociodemographic data from 1084 BC samples. Mutated genes were classified based on GeneCards platform data. Enrichment analysis was performed with CancerHallmarks, and genes not found were compared with MSigDB's Hallmark Gene Sets. Genes absent from both were further analyzed using NDEx through Cytoscape.org to explore their role in cancer. Results: Significant differences (p < 0.05) were observed in sex, tumor subtypes, genetic ancestry, median of the fraction of the altered genome, mutation count, and mutation frequencies of genes across ethnic groups. We identified the most frequently mutated genes. Some of these genes were found to be associated with classic cancer hallmarks, such as replicative immortality, sustained proliferative signaling, and the evasion of growth suppressors. However, the exact role of some of these genes in cancer remains unclear, highlighting the need for further research to better understand their involvement in tumor biology. Conclusions: This study identified significant clinicopathological and genomic variations in BC across ethnic groups. While key genes associated with cancer hallmarks were found, the incomplete characterization of some highlights the need for further research, especially focusing on ethnic groups, to understand their role in tumor biology and improve personalized treatments.
Collapse
Affiliation(s)
- Asbiel Felipe Garibaldi-Ríos
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis E. Figuera
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Guillermo Moisés Zúñiga-González
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara 44340, Jalisco, Mexico; (G.M.Z.-G.); (M.A.R.-R.)
| | - Belinda Claudia Gómez-Meda
- Departamento de Biología Molecular y Genómica, Instituto de Genética Humana “Dr. Enrique Corona Rivera”, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| | - Ana María Puebla-Pérez
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara 44430, Jalisco, Mexico;
| | - Alicia Rivera-Cameras
- Departamento Ciclo de Vida, Genética y Medicina Genómica, Unidad Académica de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Guadalajara 45129, Jalisco, Mexico;
| | - María Teresa Magaña-Torres
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
| | - José Elías García-Ortíz
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
| | - Ingrid Patricia Dávalos-Rodríguez
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
| | - Mónica Alejandra Rosales-Reynoso
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara 44340, Jalisco, Mexico; (G.M.Z.-G.); (M.A.R.-R.)
| | - Patricia Montserrat García-Verdín
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Irving Alejandro Carrillo-Dávila
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Blanca Miriam Torres-Mendoza
- Laboratorio de Inmunodeficiencias Humanas y Retrovirus, División de Neurociencias, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico;
- Departamento de Disciplinas Filosófico Metodológicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Guadalupe Ávalos-Navarro
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega, Universidad de Guadalajara, Av. Universidad 1115, Lindavista, Ocotlán 47820, Jalisco, Mexico;
| | - Martha Patricia Gallegos-Arreola
- División de Genética, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Guadalajara 44340, Jalisco, Mexico; (A.F.G.-R.); (L.E.F.); (M.T.M.-T.); (J.E.G.-O.); (I.P.D.-R.); (P.M.G.-V.); (I.A.C.-D.)
| |
Collapse
|
282
|
Shi C, Ma X, Zhang R, Lin Z, Zhou X. The synergistic antitumour effect of Carrimycin combined with 5-fluorouracil on colorectal cancer. Sci Rep 2025; 15:9155. [PMID: 40097541 PMCID: PMC11914086 DOI: 10.1038/s41598-025-94306-5] [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: 01/13/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025] Open
Abstract
5-Fluorouracil (5-FU)-based chemotherapy often leads to drug resistance and adverse reactions in individuals with colorectal cancer (CRC). Carrimycin (CAM), a drug with notable antitumour effects across various tumour types, including hepatocellular carcinoma, glioblastoma, and small-cell lung carcinoma, offers an alternative owing to its limited side effects. Combination therapy is a common strategy to mitigate the negative effects of 5-FU and enhance its therapeutic efficacy. This study aimed to investigate the potential synergistic antitumour effects of CAM and 5-FU on HCT-15 and HT-29 CRC cell lines. Using computational analysis, we identified and quantified the synergistic effects of CAM and 5-FU. The combination therapy significantly outperformed 5-FU alone in inhibiting cell proliferation, colony formation, cell cycle progression, and migration. Additionally, it markedly increased the levels of reactive oxygen species and induced DNA damage. Furthermore, RNA-seq analysis revealed that the JNK and p38 MAPK signalling pathways were activated by this combination. In addition, the synergistic effects of the combination therapy were validated in a mouse subcutaneous tumour graft model. In conclusion, CAM enhances the sensitivity of CRC cells to 5-FU both in vitro and in vivo, suggesting its potential as a promising candidate for combination cancer therapy.
Collapse
Affiliation(s)
- Chaoyue Shi
- Central Laboratory, Yanbian University Hospital, Yanji, Jilin, China
| | - Xiao Ma
- Central Laboratory, Yanbian University Hospital, Yanji, Jilin, China
| | - Ruixue Zhang
- Central Laboratory, Yanbian University Hospital, Yanji, Jilin, China
| | - Zhenhua Lin
- Central Laboratory, Yanbian University Hospital, Yanji, Jilin, China
| | - Xianchun Zhou
- Central Laboratory, Yanbian University Hospital, Yanji, Jilin, China.
- Yanbian University Hospital, Yanji, 133000, Jilin Province, China.
| |
Collapse
|
283
|
Gomaa S, Nassef M, Tabl G, Gabry SE. Immunoenhancing of the anti-cancer therapy and anti-oxidative stress by co-administration of granulocyte-colony stimulating factor-mobilized stem cells or cells derived from bone marrow and/or spleen plus vaccination with chemotherapeutic cyclophosphamide. Immunol Res 2025; 73:62. [PMID: 40091102 DOI: 10.1007/s12026-025-09610-z] [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: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The combination of immunotherapy and chemotherapy, referred to as chemo-immunotherapy, represents a promising regimen for developing new cancer treatments that target the local tumor microenvironment and target tumors in their early stages. However, this approach carries potential risks, including myelo- and immunosuppression, as well as the emergence of chemo-resistant tumor cells. The purpose of this study was to investigate how well mobilizing hematopoietic stem cells (HSCs) work when used alongside chemotherapy and immunotherapy to enhance and modulate the immune response, thereby overcoming immunosuppression and eliminating distant cancer cells. Ehrlich ascetic carcinoma (EAC) tumor-bearing mice were intraperitoneal (i.p.) preconditioned with CTX (4 mg/mouse). EAC-bearing mice that were preconditioned with CTX were intravenous (i.v.) administered with adoptive transferred naive mice-derived bone marrow cells (nBMCs) at 5 × 106 through lateral tail vein (nBMCs group), adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) at 5 × 106 cell/mouse (tBMCs group), a combination of adoptive transferred naïve mice-derived bone marrow cells (nBMCs) and naïve mice-derived splenocytes (nSPs) at 5 × 106 (nBMCs/nSPs group), a combination of adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) and tumor-bearing mice derived-splenocytes (tSPs) at 5 × 106 cell/mouse (tBMCs/tSPs group), or G-CSF administrated subcutaneously (s.c.) at 5 µg/mouse (G-CSF group). Subsequently, all mice groups were vaccinated with tumor lysate at a dosage of 100 µg/mouse. Treating EAC tumor-bearing mice with G-CSF, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, adoptive transferred tBMCs/tSPs, resulted in a significantly enhanced anti-tumor effect that was evidenced by increased anti-proliferative activity and growth inhibition against EAC tumor cells, increased necrosis and apoptosis rates among EAC tumor cells, restricted tumor growth in EAC tumor-bearing mice, and reduced levels of carcinoembryonic antigen (CEA) tumor marker. Furthermore, there was an improvement in serum levels of antioxidant enzyme superoxide dismutase (SOD) and malondialdehyde (MDA) in EAC tumor-bearing mice receiving G-CSF, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, and adoptive transferred tBMCs/tSPs. Notably, this treatment regimen ameliorates liver and kidney damage associated with CTX administration in EA tumor-bearing mice. The integration of G-CSF-mobilized HSCs, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs combination, and adoptive transferred tBMCs/tSPs combination may yield powerful anti-cancer therapy, thereby facilitating more effective anti-tumor immunotherapy strategies when align with anti-tumor responses. This research may propose a novel therapeutic approach that combines chemotherapy and immunotherapy for addressing early-stage cancer. Further research is necessary to connect the biomedical application and heterogeneity of human tumors and immune systems of this regimen to both diagnostic and therapeutic methodologies.
Collapse
Affiliation(s)
- Soha Gomaa
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt.
| | - Mohamed Nassef
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Ghada Tabl
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Shaimaa El Gabry
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| |
Collapse
|
284
|
Wang M, Xu S, Xu J, Wei J, Wu Y. WTAP contributes to platinum resistance in high-grade serous ovarian cancer by up-regulating malic acid: insights from liquid chromatography and mass spectrometry analysis. Cancer Metab 2025; 13:14. [PMID: 40098185 PMCID: PMC11916999 DOI: 10.1186/s40170-025-00383-5] [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: 06/22/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
High-grade serous cancer (HGSC) is the most prevalent and aggressive subtype of ovarian cancer. In this study, we utilized liquid chromatography and mass spectrometry analysis to investigate metabolic alterations in HGSC. Among the 1353 metabolites identified, 140 were significantly differed between HGSC and normal ovarian tissue. KEGG pathway enrichment analysis revealed 23 distinct metabolic pathways, including the alanine/aspartate/glutamate metabolism, pyruvate metabolism, biosynthesis of amino acids, and citrate cycle, etc. Of the significantly differentiated metabolites, malic acid, fumarate, and phosphoenolpyruvate were found in the citrate cycle and glycolysis. In further analysis, 22 differentially expressed genes (DEGs) of glucose metabolism were found between HGSC and normal controls. Multivariate Cox analysis of the 22 DEGs showed that ME1, ALDOC, and RANBP2 were associated with overall survival in the TCGA cohort.Bioinformatic analysis indicated WTAP is strongly correlated to the expression of ME1, which is a rate-limiting enzyme that regulates the shuttle of malic acid in mitochondria and cytoplasm. After the knockdown of WTAP in A2780 and OVCAR-3 cells, the activity of the malic enzyme decreased which led to the accumulation of malic acid and citric acid, and the reduction of pyruvate and lactic acid. In A2780 and OVCAR-3 cells, the IC50 to platinum was increased after the knockdown of WTAP. After the knockdown of WTAP, the expression of ME1 was down-regulated and the m6A modification was down-regulated in ovarian cell lines. On the SRAMP website, there were two binding sites with high m6A scores at the 5 '-UTR 177 and 970 of ME1 mRNA. WTAP contributes to the platinum resistance through regulating the conversion from aerobic glycolysis to OXPHOS by upregulating the expression of ME1.
Collapse
Affiliation(s)
- Ming Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Shuiqing Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jianqing Xu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jiahui Wei
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Yumei Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China.
| |
Collapse
|
285
|
Ding M, Ma C, Lin Y, Fang H, Xu Y, Wang S, Chen Y, Zhou J, Gao H, Shan Y, Yang L, Sun H, Tang Y, Wu X, Zhu L, Zheng L, Assaraf YG, Zhou BBS, Gu S, Li H. Therapeutic targeting de novo purine biosynthesis driven by β-catenin-dependent PPAT upregulation in hepatoblastoma. Cell Death Dis 2025; 16:179. [PMID: 40097378 PMCID: PMC11914223 DOI: 10.1038/s41419-025-07502-6] [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: 09/06/2024] [Revised: 02/07/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
De novo purine biosynthesis (DNPS) was previously shown to be aberrantly activated in many cancers. However, the activity of DNPS pathway and its underlying regulatory mechanism in hepatoblastoma (HB) remain poorly understood. Herein, we discovered that the expression of PPAT, the rate-limiting enzyme in DNPS, was markedly upregulated in HB, leading to an augmented purine flux via DNPS, thereby promoting both HB cell proliferation and migration. Furthermore, we found that activated mutant β-catenin, a dominant driver of HB, transcriptionally activated PPAT expression, hence stimulating DNPS and constituting a druggable metabolic vulnerability in HB. Consistently, pharmacological targeting using a DNPS inhibitor lometrexol or genetic repressing the enhanced DNPS markedly blocked HB progression in vitro and in vivo. Our findings suggest that HB patients harboring activated β-catenin mutations and consequent DNPS upregulation, may be treated efficaciously with DNPS enzyme inhibitors like lometrexol. These novel findings bear major therapeutic implications for targeted precision medicine of HB.
Collapse
Affiliation(s)
- Ming Ding
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Fuzhou, 350014, China
| | - Chunshuang Ma
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yanyan Lin
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Houshun Fang
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yan Xu
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shuxuan Wang
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yao Chen
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jiquan Zhou
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hongxiang Gao
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yuhua Shan
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Liyuan Yang
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Huiying Sun
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yabin Tang
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Shanghai Collaborative Innovation Center for Translational Medicine Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyu Wu
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Shanghai Collaborative Innovation Center for Translational Medicine Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Zheng
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Fuzhou, 350014, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel.
| | - Bin-Bing S Zhou
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Fuzhou, 350014, China.
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Shanghai Collaborative Innovation Center for Translational Medicine Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Song Gu
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Hui Li
- Pediatric Translational Medicine Institute, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Fuzhou, 350014, China.
| |
Collapse
|
286
|
Feng T, Li P, Li S, Wang Y, Lv J, Xia T, Lee HJ, Piao HL, Chen D, Ma Y. Metabolic state uncovers prognosis insights of esophageal squamous cell carcinoma patients. J Transl Med 2025; 23:342. [PMID: 40098145 PMCID: PMC11912770 DOI: 10.1186/s12967-025-06087-0] [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: 10/23/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Metabolite-protein interactions (MPIs) are crucial regulators of cancer metabolism; however, their roles and coordination within the esophageal squamous cell carcinoma (ESCC) microenvironment remain largely unexplored. This study is the first to comprehensively map the metabolic landscape of the ESCC microenvironment by integrating an MPI network with multi-scale transcriptomics data. METHODS First, we characterized the metabolic states of cells in ESCC using single-cell transcriptome profiles of key metabolite-interacting proteins. Next, we determined the metabolic patterns of each ESCC patient based on the composition of different metabolic states within bulk samples. Finally, the ESCC samples were clustered into unique subtypes. RESULTS Sixteen ESCC metabolic states across 7 cell types were identified based on the re-analysis of single-cell RNA-sequencing data of 208,659 cells in 64 ESCC samples. Each of the 7 cell types within the tumor microenvironment exhibited distinct metabolic states, highlighting the high metabolic heterogeneity of ESCC. Based on differences in the compositions of the metabolic states, 4 ESCC subtypes were identified in two independent cohorts (n = 79 and 119), which were associated with significant variations in prognosis, clinical features, gene expression, and pathways. Notably, the inactivation of cellular detoxification processes may contribute to the poor prognosis of ESCC patients. CONCLUSIONS Overall, we redefined robust ESCC prognostic subtypes and identified key MPI pathways that link metabolism to tumor heterogeneity. This study provides the first comprehensive mapping of the ESCC metabolic microenvironment, offering novel insights into ESCC metabolic diversity and its clinical applications.
Collapse
Affiliation(s)
- Tingze Feng
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Pengfei Li
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China
| | - Siyi Li
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yuhan Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Jing Lv
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Tian Xia
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Hoy-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hai-Long Piao
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China.
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - Di Chen
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - Yegang Ma
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China.
| |
Collapse
|
287
|
Wang Y, Xu N, Ndzie Noah ML, Chen L, Zhan X. Pyruvate Kinase M1/2 Proteoformics for Accurate Insights into Energy Metabolism Abnormity to Promote the Overall Management of Ovarian Cancer Towards Predictive, Preventive, and Personalized Medicine Approaches. Metabolites 2025; 15:203. [PMID: 40137167 PMCID: PMC11944880 DOI: 10.3390/metabo15030203] [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: 02/02/2025] [Revised: 03/01/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Ovarian cancer (OC) is a global health problem that frequently presents at advanced stages, is predisposed to recurrence, readily develops resistance to platinum-based drugs, and has a low survival rate. Predictive, preventive, and personalized medicine (PPPM/3PM) offers an integrated solution with the use of genetic, proteomic, and metabolic biomarkers to identify high-risk individuals for early detection. Metabolic reprogramming is one of the key strategies employed by tumor cells to adapt to the microenvironment and support unlimited proliferation. Pyruvate kinases M1 and M2 (PKM1/2) are encoded by the PKM gene, a pivotal enzyme in the last step of the glycolytic pathway, which is at the crossroads of aerobic oxidation and the Warburg effect to serve as a potential regulator of glucose metabolism and influence cellular energy production and metabolic reprogramming. Commonly, the ratio of PKM1-to-PKM2 is changed in tumors compared to normal controls, and PKM2 is highly expressed in OC to induce a high glycolysis rate and participate in the malignant invasion and metastatic characteristics of cancer cells with epithelial/mesenchymal transition (EMT). PKM2 inhibitors suppress the migration and growth of OC cells by interfering with the Warburg effect. Proteoforms are the final structural and functional forms of a gene/protein, and the canonical protein PKM contains all proteoforms encoded by the same PKM gene. The complexity of PKM can be elucidated by proteoformics. The OC-specific PKM proteoform might represent a specific target for therapeutic interventions against OC. In the framework of PPPM/3PM, the OC-specific PKM proteoform might be the early warning and prognosis biomarker. It is important to clarify the molecular mechanisms of PKM proteoforms in cancer metabolism. This review analyzes the expression, function, and molecular mechanisms of PKM proteoforms in OC, which help identify specific biomarkers for OC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China
- Department of Gynecology, Gaotang County Medical Center, Liaocheng 252800, China
| | - Nuo Xu
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
| | - Marie Louise Ndzie Noah
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China
| | - Xianquan Zhan
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China
| |
Collapse
|
288
|
Frigerio C, Galli M, Castelli S, Da Prada A, Clerici M. Control of Replication Stress Response by Cytosolic Fe-S Cluster Assembly (CIA) Machinery. Cells 2025; 14:442. [PMID: 40136691 PMCID: PMC11941123 DOI: 10.3390/cells14060442] [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: 02/19/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Accurate DNA replication is essential for the maintenance of genome stability and the generation of healthy offspring. When DNA replication is challenged, signals accumulate at blocked replication forks that elicit a multifaceted cellular response, orchestrating DNA replication, DNA repair and cell cycle progression. This replication stress response promotes the recovery of DNA replication, maintaining chromosome integrity and preventing mutations. Defects in this response are linked to heightened genetic instability, which contributes to tumorigenesis and genetic disorders. Iron-sulfur (Fe-S) clusters are emerging as important cofactors in supporting the response to replication stress. These clusters are assembled and delivered to target proteins that function in the cytosol and nucleus via the conserved cytosolic Fe-S cluster assembly (CIA) machinery and the CIA targeting complex. This review summarizes recent advances in understanding the structure and function of the CIA machinery in yeast and mammals, emphasizing the critical role of Fe-S clusters in the replication stress response.
Collapse
Affiliation(s)
| | | | | | | | - Michela Clerici
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano-Bicocca, 20126 Milano, Italy; (C.F.); (M.G.); (S.C.); (A.D.P.)
| |
Collapse
|
289
|
Ye QY, Wang YY, Wang ZJ, Lu M, Peng HX, Wang X, Cheng XX, Ying HQ. Robust Predictive Performance of MLPAS and CCMLP for Clinical Outcome and Risk Stratification in Patients with Colorectal Cancer. J Inflamm Res 2025; 18:3889-3900. [PMID: 40109656 PMCID: PMC11921802 DOI: 10.2147/jir.s498028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND There is no recognized biomarker is recommended to monitor or predict the prognosis of colorectal cancer (CRC) patients with negative detection of carcinoembryonic antigen (CEA) or carbohydrate antigen 19-9 (CA19-9) and to classify high recurrence-risk cases. METHODS Discovery and two-stage validation cohorts, which included 2111 radically resected patients with stage II-III CRC, were enrolled in this study. We detected preoperative peripheral monocyte, platelet, albumin (Alb), pre-albumin (pAlb), CEA, and CA19-9 and investigated the prognostic and risk-stratified roles of twelve new inflammatory biomarkers in the three cohorts. RESULTS In our study, monocyte-to-pAlb ratio (MPAR), monocyte-to-lymphocyte -to-Alb ratio (MLAR), monocyte-to-lymphocyte-to-pAlb ratio (MLPAR), monocyte- to-pAlb score (MPAS), lymphocyte-to-monocyte-Alb score (MLAS), lymphocyte-to monocyte-pAlb score (MLPAS), and platelet-to-lymphocyte-Alb score (PLAS) were significantly associated with both RFS and OS in three cohorts. MLPAS showed the best performance in predicting RFS and OS, and it was related to right-tumor location and significant cancer burden (≥5cm) in the overall population. Moreover, MLPAS is a robust prognostic biomarker in subgroups stratified by CEA or CA19-9. Patients with scores zero and two of the CEA-CA19-9-MLPAS score (CCMLP) showed the lowest and highest recurrence and death rates, respectively, and significant survival differences were observed between them. CONCLUSION MLPAS is an optimal, independent, and robust prognostic biomarker in the stage II-III CRC population, especially with negative CEA or CA19-9. The CCMLP could effectively classify high recurrence-risk patients who require more focus, monitoring, and treatment for the clinic.
Collapse
Affiliation(s)
- Qiu-Ying Ye
- Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
- Department of Medical Technology, Jiangxi Medical College, Shangrao, 334000, People’s Republic of China
- Department of Laboratory Medicine, Central Hospital of Shangrao City, Shangrao, 334000, People’s Republic of China
| | - Yuan-Yuan Wang
- Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Zhi-Jie Wang
- Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Min Lu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Hong-Xin Peng
- Department of Clinical Laboratory, Nanjing First Hospital, Nanjing, 210006, People’s Republic of China
| | - Xin Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, People’s Republic of China
| | - Xue-Xin Cheng
- Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Hou-Qun Ying
- Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
- Department of Laboratory Medicine, Central Hospital of Shangrao City, Shangrao, 334000, People’s Republic of China
- Shangrao Medical Center, The Second Affiliated Hospital of Nanchang University, Shangrao, 334000, People’s Republic of China
| |
Collapse
|
290
|
Zhang YX, Wan H, Shan GY, Cheng JY, Liu YY, Shi WN, Li HJ. Pharmacological role of Herba Patriniae and Coix seed in colorectal cancer. World J Gastrointest Oncol 2025; 17:99673. [PMID: 40092956 PMCID: PMC11866235 DOI: 10.4251/wjgo.v17.i3.99673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/31/2024] [Accepted: 12/25/2024] [Indexed: 02/14/2025] Open
Abstract
Colorectal cancer (CRC) is the most prevalent cancer globally, and its traditional treatment modalities commonly encompass radiation therapy, chemotherapy, surgery and the administration of cytotoxic drugs. Currently, novel chemotherapy drugs that combine traditional Chinese medicine (TCM) with herbal extracts exhibit superior comprehensive benefits. Herein, we delved into an article authored by Wang et al, focusing specifically on the pharmacological effects of "Herba Patriniae and Coix seed (HC)" and their targeted mechanisms in combating CRC. From the perspective of TCM philosophy, damp-heat stagnation and toxicity are the cardinal pathogenic factors underlying CRC. HC, renowned for their abilities to antipyretic and enhance diuresis, have demonstrated promising efficacy in preliminary studies for the treatment of CRC. These findings offer potential insights in favor of fostering anti-cancer medications.
Collapse
Affiliation(s)
- Yu-Xin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hui Wan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Guan-Yue Shan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Jun-Ya Cheng
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130061, Jilin Province, China
| | - Yi-Ying Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Wen-Na Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hai-Jun Li
- Institute of Liver Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
291
|
Li Y, Zhao J, Tang K, Yin J, Song Y, Pan W, Li N, Tang B. Doxorubicin prodrug for γ-glutamyl transpeptidase imaging and on-demand cancer therapy. Biosens Bioelectron 2025; 272:117127. [PMID: 39778243 DOI: 10.1016/j.bios.2025.117127] [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: 10/16/2024] [Revised: 11/28/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025]
Abstract
The γ-glutamyl transpeptidase (γ-GGT) is an important tumor marker, which has been reported to be firmly associated with the developmental stage of liver cancer. Therefore, it makes sense to image and monitor γ-GGT level and design γ-GGT-responsive prodrug for integrated diagnosis and treatment of liver cancer. Herein, we prepare a doxorubicin (Dox) prodrug for imaging γ-GGT and on-demand treating liver cancer. When γ-GGT exists, the γ-glutamyl group will be cut off to liberate free Dox for monitoring cancer progression and killing tumor cells. Fortunately, little Dox is released due to the low level of γ-GGT in normal cells, which improves the safety and efficiency of chemotherapy. To further improve the tumor targeted ability, Dox prodrug is loaded in hyaluronic acid modified liposome nanoparticles to form the nano-prodrug. Then nano-prodrug is enriched in the tumor by binding to the high expressed CD44 on cancer cells. With the assistance of anti-PD-L1, nano-prodrug effectively inhibits the growth of proximal and distal tumors.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China
| | - Jiexiang Zhao
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China
| | - Kun Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China
| | - Jiaqi Yin
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China
| | - Yingying Song
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, PR China; Laoshan Laboratory, Qingdao, 266237, PR China.
| |
Collapse
|
292
|
Medhi K, Mukherjee S, Dagar A, Tiwari AK, Daffara S, Bana S, Uttam V, Ansari MR, Yadav V, Tuli HS, Jain A. MYOSLID: A Critical Modulator of Cancer Hallmarks. Genes (Basel) 2025; 16:341. [PMID: 40149492 PMCID: PMC11942567 DOI: 10.3390/genes16030341] [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: 02/06/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Despite being the leading cause of death worldwide, cancer still lacks precise biomarkers for effective targeting, limiting efforts to reduce mortality rates. This review explores the role and clinical significance of a newly identified long non-coding RNA, MYOSLID, in cancer progression. MYOSLID has emerged as a critical modulator in cancer progression by influencing key hallmarks such as proliferation, immune evasion, metastasis, and metabolic reprogramming. It promotes tumor cell growth by stabilizing hypoxia-inducible factor 1 and acting as a competing endogenous RNA (ceRNA) to sequester tumor-suppressive microRNAs like miR-29c-3p, thereby enhancing oncogene expression. It facilitates immune evasion by upregulating PD-L1, suppressing T cell activation, and modulating necroptosis pathways involving RIPK1 and RIPK3. Additionally, MYOSLID drives metastasis by regulating epithelial-mesenchymal transition markers such as LAMB3 and Slug while promoting RAB13-mediated cytoskeletal remodeling and enhancing cancer cell invasion. We have obtained the expression of MYOSLID from TCGA and the ENCORI database. The expression of colorectal adenocarcinoma (COAD) and head and neck squamous cell carcinoma (HNSCC) is associated with poor prognosis and lower survival rate. Given its significant potential as a diagnostic biomarker and therapeutic target, further research is required to elucidate its precise molecular mechanisms and therapeutic applications in cancer treatment.
Collapse
Affiliation(s)
- Kanupriya Medhi
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Sagarika Mukherjee
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Aastha Dagar
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Ashutosh Kumar Tiwari
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Sia Daffara
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Sanjana Bana
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Vivek Uttam
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| | - Md Rizwan Ansari
- 3rd Floor, Jyoti Pinnacle Building, Survey No.11, Kondapur Village, Serilingampally Mandal, Ranga Reddy District, Hyderabad 500081, Telangana, India;
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, 20213 Malmö, Sweden;
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Aklank Jain
- Non-Coding RNA and Cancer Biology Lab, Department of Zoology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India; (K.M.); (S.M.); (A.D.); (A.K.T.); (S.D.); (S.B.); (V.U.)
| |
Collapse
|
293
|
Singh S, Kim GH, Baek KR, Seo SO. Anti-Cancer Strategies Using Anaerobic Spore-Forming Bacteria Clostridium: Advances and Synergistic Approaches. Life (Basel) 2025; 15:465. [PMID: 40141809 PMCID: PMC11943571 DOI: 10.3390/life15030465] [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: 02/25/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Despite ongoing advancements, cancer remains a significant global health concern, with a persistent challenge in identifying a definitive cure. While various cancer therapies have been developed and approved, offering treatments for smaller neoplasms, their efficacy diminishes in solid tumors and hypoxic environments, particularly for chemotherapy and radiation therapy. A novel approach, Clostridium-based therapy, has emerged as a promising candidate for current solid tumor treatments due to its unique affinity for the hypoxic tumor microenvironment. This review examines the potential of Clostridium in cancer treatment, encompassing direct tumor lysis, immune modulation, and synergistic effects with existing cancer therapies. Advancements in synthetic biology have further enhanced its potential through genetic modifications, such as the removal of alpha toxin gene from Clostridium novyi-NT, the implementation of targeted approaches, and reduction in systemic toxicity. Although preclinical and clinical studies have demonstrated that Clostridium-based treatments combined with other therapies hold promise for complete cancer eradication, challenges persist. Through this review, we also propose that the integration of various methods and technologies together with Clostridium-based therapy may lead to the complete eradication of cancer in the future.
Collapse
Affiliation(s)
- Saloni Singh
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
| | - Geun-Hyung Kim
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
| | - Kwang-Rim Baek
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
| | - Seung-Oh Seo
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea (G.-H.K.); (K.-R.B.)
- Research Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
294
|
Ou Z, Zhu L, Chen X, Liu T, Cheng G, Liu R, Zhang S, Tan W, Lin D, Wu C. Hypoxia-Induced Senescent Fibroblasts Secrete IGF1 to Promote Cancer Stemness in Esophageal Squamous Cell Carcinoma. Cancer Res 2025; 85:1064-1081. [PMID: 39661488 DOI: 10.1158/0008-5472.can-24-1185] [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: 04/12/2024] [Revised: 08/12/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Cancer-associated fibroblasts (CAF) contribute to cancer initiation and progression and play a pivotal role in therapeutic response and patient prognosis. CAFs exhibit functional and phenotypic heterogeneity, highlighting the need to clarify the specific subtypes of CAFs to facilitate the development of targeted therapies against protumorigenic CAFs. In this study, using single-cell RNA sequencing on patient samples of esophageal squamous cell carcinoma (ESCC), we identified a CAF subcluster associated with tumor stemness that was enriched in genes associated with hypoxia and senescence. The CAF subpopulation, termed as hypoxia-induced senescent fibroblasts (hsCAF), displayed high secretion of insulin-like growth factor 1 (IGF1). The hsCAFs inhibited AMP-activated protein kinase (AMPK) activity in cancer cells via IGF1 to promote tumor stemness. The formation of hsCAFs was induced by the synergetic effect of hypoxia and cancer cells. Activation of nuclear factor erythroid 2-related factor 2 (NRF2) in cancer cells under hypoxia drove IL1α production to trigger CAF senescence and IGF1 secretion via nuclear factor I A. Knockout of IGF1 in CAFs or nuclear factor erythroid 2-related factor 2 in ESCC cells suppressed the tumor growth and chemotherapy resistance induced by CAFs in vivo. Importantly, patients with high proportions of hsCAFs showed poor survival and a worse response to chemotherapy. In summary, these findings identify a hsCAF subpopulation generated by interplay between cancer cells and CAFs under hypoxic conditions that promotes ESCC stemness and reveal targeting hsCAFs as an effective therapeutic strategy against chemotherapy-resistant ESCC. Significance: A hypoxic microenvironment and cancer cells cooperate to induce a senescent fibroblast subset that supports tumor stemness, suggesting that targeting this cancer-associated fibroblast subpopulation is a potential therapeutic strategy to overcome chemoresistance.
Collapse
Affiliation(s)
- Zhengjie Ou
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Rucheng Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
295
|
Sun YY, Liu NN. Mycobiome: an underexplored kingdom in cancer. Microbiol Mol Biol Rev 2025:e0026124. [PMID: 40084887 DOI: 10.1128/mmbr.00261-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
SUMMARYThe human microbiome, including bacteria, fungi, archaea, and viruses, is intimately linked to both health and disease. The relationship between bacteria and disease has received much attention and intensive investigation, while that of the fungal microbiome, also known as mycobiome, has lagged far behind bacteria. There is growing evidence showing mycobiome dysbiosis in cancer patients, and certain cancer-specific fungi may contribute to cancer progression by interacting with both host and bacteria. It was also demonstrated that the role of fungi-derived products in cancer should also not be underestimated. Therefore, investigating how fungal pathogenesis contributes to the onset and spread of cancer would yield crucial information for cancer diagnosis, prevention, and anti-cancer therapy.
Collapse
Affiliation(s)
- Yan-Yan Sun
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
296
|
Roy D, Bhattacharya B, Chakravarti R, Singh P, Arya M, Kundu A, Patil A, Siva B, Mehta S, Kazi TA, Ghosh D. LncRNAs in oncogenic microenvironment: from threat to therapy. Front Cell Dev Biol 2025; 12:1423279. [PMID: 40176927 PMCID: PMC11962222 DOI: 10.3389/fcell.2024.1423279] [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: 04/25/2024] [Accepted: 12/09/2024] [Indexed: 04/05/2025] Open
Abstract
LncRNAs are RNA molecules of more than 200 nucleotides in length and participate in cellular metabolism and cellular responses through their diverse interactomedespite having no protein-coding capabilities. Such significant interactions also implicate the presence of lncRNAs in complex pathobiological pathways of various diseases, affecting cellular survival by modulating autophagy, inflammation and apoptosis. Proliferating cells harbour a complex microenvironment that mainly stimulate growth-specific activities such as DNA replication, repair, and protein synthesis. They also recognise damages at the macromolecular level, preventing them from reaching the next-generation. LncRNAs have shown significant association with the events occurring towards proliferation, regulating key events in dividing cells, and dysregulation of lncRNA transcriptome affects normal cellular life-cycle, promoting the development of cancer. Furthermore, lncRNAs also demonstrated an association with cancer growth and progression by regulating key pathways governing cell growth, epithelial-mesenchymal transition and metastasis. This makes lncRNAs an attractive target for the treatment of cancer and can also be used as a marker for the diagnosis and prognosis of diseases due to their differential expression in diseased samples. This review delves into the correlation of the lncRNA transcriptome with the fundamental cellular signalling and how this crosstalk shapes the complexity of the oncogenic microhabitat.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Dipanjan Ghosh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata, Kolkata, India
| |
Collapse
|
297
|
Cortellesi E, Savini I, Veneziano M, Gambacurta A, Catani MV, Gasperi V. Decoding the Epigenome of Breast Cancer. Int J Mol Sci 2025; 26:2605. [PMID: 40141248 PMCID: PMC11942310 DOI: 10.3390/ijms26062605] [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: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Breast cancer (BC) is the most prevalent malignancy among women, characterized by extensive heterogeneity stemming from molecular and genetic alterations. This review explores the intricate epigenetic landscape of BC, highlighting the significant role of epigenetic modifications-particularly DNA methylation, histone modifications, and the influence of non-coding RNAs-in the initiation, progression, and prognosis of the disease. Epigenetic alterations drive crucial processes, including gene expression regulation, cell differentiation, and tumor microenvironment interactions, contributing to tumorigenesis and metastatic potential. Notably, aberrations in DNA methylation patterns, including global hypomethylation and hypermethylation of CpG islands, have been associated with distinct BC subtypes, with implications for early detection and risk assessment. Furthermore, histone modifications, such as acetylation and methylation, affect cancer cell plasticity and aggressiveness by profoundly influencing chromatin dynamics and gene transcription. Finally, non-coding RNAs contribute by modulating epigenetic machinery and gene expression. Despite advances in our knowledge, clinical application of epigenetic therapies in BC is still challenging, often yielding limited efficacy when used alone. However, combining epi-drugs with established treatments shows promise for enhancing therapeutic outcomes. This review underscores the importance of integrating epigenetic insights into personalized BC treatment strategies, emphasizing the potential of epigenetic biomarkers for improving diagnosis, prognosis, and therapeutic response in affected patients.
Collapse
Affiliation(s)
- Elisa Cortellesi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Isabella Savini
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Matteo Veneziano
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Alessandra Gambacurta
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
- NAST Centre (Nanoscience & Nanotechnology & Innovative Instrumentation), Tor Vergata University of Rome, 00133 Rome, Italy
| | - Maria Valeria Catani
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Valeria Gasperi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| |
Collapse
|
298
|
Su W, Wang W, Zhang G, Yang L. Epigenetic regulatory protein chromobox family regulates multiple signalling pathways and mechanisms in cancer. Clin Epigenetics 2025; 17:48. [PMID: 40083014 PMCID: PMC11907984 DOI: 10.1186/s13148-025-01852-w] [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: 10/02/2024] [Accepted: 02/20/2025] [Indexed: 03/16/2025] Open
Abstract
Signal transduction plays a pivotal role in modulating a myriad of critical processes, including the tumour microenvironment (TME), cell cycle arrest, proliferation and apoptosis of tumour cells, as well as their migration, invasion, and the epithelial-mesenchymal transition (EMT). Epigenetic mechanisms are instrumental in the genesis and progression of tumours. The Chromobox (CBX) family proteins, which serve as significant epigenetic regulators, exhibit tumour-specific expression patterns and biological functionalities. These proteins are influenced by a multitude of factors and could modulate the activation of diverse signalling pathways within tumour cells through alterations in epigenetic modifications, thereby acting as either oncogenic agents or tumour suppressors. This review aims to succinctly delineate the composition, structure, function, and expression of CBXs within tumour cells, with an emphasis on synthesizing and deliberating the CBXs-mediated activation of intracellular signalling pathways and the intricate mechanisms governing tumourigenesis and progression. Moreover, a plethora of contemporary studies have substantiated that CBXs might represent a promising target for the diagnosis and therapeutic intervention of tumour patients. We have also compiled and scrutinized the current research landscape concerning inhibitors targeting CBXs, aspiring to aid researchers in gaining a deeper comprehension of the biological roles and mechanisms of CBXs in the malignant evolution of tumours, and to furnish novel perspectives for the innovation of targeted tumour therapeutics.
Collapse
Affiliation(s)
- Weiyu Su
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
| | - Weiwen Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
| | - Guanghui Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China.
| | - Lianhe Yang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China.
| |
Collapse
|
299
|
Izzo LT, Reyes T, Meesala S, Ireland AS, Yang S, Sunil HS, Cheng XC, Tserentsoodol N, Hawgood SB, Patz EF, Witt BL, Tyson DR, O’Donnell KA, Oliver TG. KLF4 promotes a KRT13+ hillock-like state in squamous lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.641898. [PMID: 40161723 PMCID: PMC11952405 DOI: 10.1101/2025.03.10.641898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Lung squamous cell carcinoma (LUSC) is basal-like subtype of lung cancer with limited treatment options. While prior studies have identified tumor-propagating cell states in squamous tumors, the broader landscape of intra-tumoral heterogeneity within LUSC remains poorly understood. Here, we employ Sox2-driven mouse models, organoid cultures, and single-cell transcriptomic analyses to uncover previously unrecognized levels of cell fate diversity within LUSC. Specifically, we identify a KRT13+ hillock-like population of slower-dividing tumor cells characterized by immunomodulatory gene expression signatures. The tumor hillock-like state is conserved across multiple animal models and is present in the majority of human LUSCs as well as head and neck and esophageal squamous tumors. Our findings shed light on the cellular origins of lung hillock-like states: normal club cells give rise to tumors with luminal hillock-like populations, while basal-like tumor-propagating cells transition into basal hillock-like states, resembling homeostatic cellular responses to lung injury. Mechanistically, we identify KLF4 as a key transcriptional regulator of the hillock-like state, both necessary and sufficient to induce KRT13 expression. Together, these results provide new molecular insights into cell fate plasticity that underlies intra-tumoral heterogeneity in LUSC, offering potential avenues for new therapeutic strategies.
Collapse
Affiliation(s)
- Luke T. Izzo
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Tony Reyes
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Srijan Meesala
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Abbie S. Ireland
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Steven Yang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Hari Shankar Sunil
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xiao Chun Cheng
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Nomi Tserentsoodol
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Sarah B. Hawgood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Edward F. Patz
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
- Department of Radiology, Duke University, Durham, NC, 27710, USA
| | - Benjamin L. Witt
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Darren R. Tyson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Kathryn A. O’Donnell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Trudy G. Oliver
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Lead contact: Trudy G. Oliver
| |
Collapse
|
300
|
Antonarelli G, Pérez-García JM, Gion M, Rugo H, Schmid P, Bardia A, Hurvitz S, Harbeck N, Tolaney SM, Curigliano G, Llombart-Cussac A, Cortés J. Redefining clinical trial strategic design to support drug approval in medical oncology. Ann Oncol 2025:S0923-7534(25)00111-5. [PMID: 40086733 DOI: 10.1016/j.annonc.2025.03.005] [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: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Randomized clinical trials represent the gold standard for the introduction of innovative therapies in medical oncology, and they provide the highest level of evidence to ascertain the clinical activity of new drugs or novel combinations. However, the current infrastructure of clinical trials supporting innovative drug approvals is challenged by an increased body of knowledge concerning tumor biology and therapy resistance, a fast-growing armamentarium of novel anticancer compounds, an impressively upscaled data analysis capacity, as well as increasing costs related to clinical trials management. In this scenario, modern clinical trial designs need to evolve to expedite new drug approvals by tailoring patients' treatment strategies according to their medical needs. Balanced, patient-oriented clinical trial designs are eagerly warranted to increase their efficiency, to include the fast pace of technological innovations and scientific discoveries, and, ultimately, to face the challenges of the modern medical oncology field.
Collapse
Affiliation(s)
- G Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - J M Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, USA; International Breast Cancer Center (IBCC), Pangaea Oncology, Quirón Group, Barcelona
| | - M Gion
- Hospital Universitario Ramón y Cajal, Madrid, Spain; IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid
| | - H Rugo
- Department of Medicine, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, USA
| | - P Schmid
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - A Bardia
- University of California Los Angeles (UCLA), Los Angeles
| | - S Hurvitz
- Fred Hutchinson Cancer Center, University of Washington School of Medicine, Seattle, USA
| | - N Harbeck
- Breast Center, Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, Germany
| | - S M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - G Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - A Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Arnau de Vilanova Hospital, Universidad Católica de Valencia, Valencia
| | - J Cortés
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, USA; IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain; Oncology Department, Hospital Universitario Torrejón, Ribera Group, Madrid, Spain.
| |
Collapse
|